Next Article in Journal
Study of Seed Ageing in lpa1-1 Maize Mutant and Two Possible Approaches to Restore Seed Germination
Next Article in Special Issue
A Critical Appraisal of the Physicochemical Properties and Biological Effects of Artificial Tear Ingredients and Formulations
Previous Article in Journal
The Association between the Bisphenols Residues in Amniotic Fluid and Fetal Abnormalities in Polish Pregnant Women—Its Potential Clinical Application
Previous Article in Special Issue
Non-Invasive Tear Break-Up Detection with the Kowa DR-1α and Its Relationship to Dry Eye Clinical Severity
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Potential Role of Regulated Cell Death in Dry Eye Diseases and Ocular Surface Dysfunction

Laboratory of Medicinal Chemistry, University of Antwerp, Universiteitsplein 1, B-2160 Antwerp, Belgium
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2023, 24(1), 731; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms24010731
Submission received: 22 November 2022 / Revised: 23 December 2022 / Accepted: 29 December 2022 / Published: 1 January 2023

Abstract

:
The research on new treatments for dry eye diseases (DED) has exponentially grown over the past decades. The increased prevalence of dry eye conditions, particularly in the younger population, has received much attention. Therefore, it is of utmost importance to identify novel therapeutical targets. Regulated cell death (RCD) is an essential process to control the biological homeostasis of tissues and organisms. The identification of different mechanisms of RCD stimulated the research on their involvement in different human pathologies. Whereas apoptosis has been widely studied in DED and included in the DED vicious cycle, the role of RCD still needs to be completely elucidated. In this review, we will explore the potential roles of different types of RCD in DED and ocular surface dysfunction. Starting from the evidence of oxidative stress and inflammation in dry eye pathology, we will analyse the potential therapeutic applications of the following principal RCD mechanisms: ferroptosis, necroptosis, and pyroptosis.

1. Introduction

Considered one of the most prevalent eye syndromes, keratoconjunctivitis sicca, commonly known as dry eye disease (DED), affects millions of people worldwide, with a percentage ranging between 5 to 50% [1]. The Tear Film & Ocular Surface Society (TFOS) Dry Eye Workshop II (TFOS DEWS II) in 2017 defined dry eye as “a multifactorial disease of the ocular surface characterized by a loss of homeostasis of the tear film and accompanied by ocular symptoms, in which tear film instability and hyperosmolarity, ocular surface inflammation and damage, and neurosensory abnormalities play etiological roles” [2]. In the past years, the impact of this disorder has increased worldwide, particularly among the younger population [3]. The major risk factors are frequent use of video screens, environmental causes such as pollution and low humidity, and wearing of contact lenses [4]. The symptoms can be mild or more severe, generally leading to discomfort and visual disturbance [5]. Epidemiological studies underline that age and sex have an impact on the symptoms and signs of DED [6]. Indeed, hormones seem to influence the incidence of eye disorders in the population, particularly DED [7]. The female sex is considered a risk factor with a prevalence between 10–20% of DED, especially in the post-menopause stage [8]. During menopause, reduced estrogen levels promote evaporative DED [9]. However, recent studies hypothesised that menopause evaporative DED was actually due to the reduction of androgens levels rather than estrogens [7,10,11,12]. Therefore, additional studies are needed to further elucidate the role of estrogens in DED [13]. In addition, iatrogenic intervention is one of the most studied causes of DED [14]. In iatrogenic DED, the common risk factors are the use of topical drugs (mainly linked to the presence of preservatives) [15], systemic medications (DED can be a consequence of adverse effects) [16], contact lenses wear, and ophthalmological surgical procedures (related to the procedure itself or as a transitory/permanent side-effect) [14].
Based on an etiological classification, DED can be subdivided into Aqueous Deficient Dry Eye (ADDE) and Evaporative Dry Eye (EDE) [17]. In ADDE, lacrimal tear secretion and volume are reduced, leading to hyperosmolarity and tear film instability [18]. Consequently, the inflammatory mediators are released and detected in the tears of patients suffering from ADDE, particularly in patients affected by Sjögren syndrome dry eye (SSDE) [19]. The Sjögren syndrome is a multifactorial autoimmune endocrinopathy associated with immunologic abnormalities, and is characterised by a severe form of dry eye and/or dry mouth [20]. In EDE, normal lacrimal secretion is followed by excessive evaporation of the tear film, leading to tear hyperosmolarity. The most important aspect of EDE is the meibomian gland dysfunction (MGD), which alters lipid secretion perturbating the corresponding lipid layer of the ocular surface [21]. Consequently, the tear components are modified and the aqueous layers evaporate rapidly [22]. The alteration in the electrolytes equilibrium can cause damage to the ocular surface and tear film, promoting hyperosmolarity and oxidative stress [23]. As a consequence, the concentration of reactive oxygen species (ROS) on the ocular surface increases, propagating dry eyes into a vicious circle [24,25,26]. The tear hyperosmolarity is the main factor responsible for the activation of different pathways leading to the release of inflammatory mediators and proteases [24,27].
Dry eye disease concerns ocular surface composed of cornea, conjunctiva, tear film, lacrimal glands, meibomian glands, eyelids, muscles, and nerves (Figure 1). A healthy human tear film is composed of lipids, water, mucin, proteins, electrolytes, and vitamins [28]. The tear-secreting glands, the lacrimal and meibomian glands, along with the goblet cells in the eye and eyelid, produce the tears and work with the ocular surface to maintain optimum ocular health by lubricating the eye, removing debris, and protecting from infection [29]. The transparent, dome-shaped cornea presents superficial microvilli which aid the tear film anchorage and regulates the secretion of growth factors and cytokines as a shield [30]. The conjunctiva, situated between the corneal rim and the lid margin, is composed of goblet cells responsible for mucin secretion [31]. Externally, the eye is coated with the tear film, formed by three different regions: an external lipid layer, a mild aqueous layer, and an inner mucin layer [32]. The lipid layer is regulated by the meibomian glands lipids and proteins secretion and the main role is to prevent evaporation of the aqueous layer [33]. The lacrimal glands regulate the water, electrolytes, proteins, and mucus content in the aqueous phase, which is extremely important for eye irritation [34]. The electrolytes play a significant role to prevent hyperosmolarity and alter physiological conditions [35]. Additionally, the eyelid contributes to prevent the desiccation of the ocular surface [36]. Internally there is a mucin layer which acts as a surfactant spreading homogeneously through the tear film of the ocular surface [37]. Secreted by the conjunctival goblet cells, the mucus is mainly based on enzymes, mucins, and leukocytes. They also minimise friction and protect the cornea during blinking [36].
The diagnostic methods currently used in DED diagnosis are based on a questionnaire called Dry Eye Questionnaire-5 (DEQ-5) or Ocular Surface Diseases Index (OSDI), collecting all the information to indicate the positivity of DED [38]. Patients normally report burning, photophobia, itching, and foreign body sensation [39,40]. However, the quantification of the symptoms is based on the tear film instability measurement (break up time (BUT)), the tear osmolarity assessment, and the tear volume measurement (Schirmer’s test) [41]. The next step is the discrimination between the two subtypes, ADDE and EDE, followed by medical prescription of the treatment according to the severity of the symptoms [42].
One of the therapeutic strategies is the application of artificial tear substitutes to improve lubrication and decrease evaporation to provide temporary relief. Different formulations, such as eye drops, topical lubricants, gels, and ointments, are available on the market [43]. The first medication used to treat DED is Cyclosporine A ophthalmic emulsion 0.05% (Restasis®, Allergan, Irvine, CA, United States) [44]. To ameliorate the drug delivery into the eye, the novel nanomicellar formulation was used for Cyclosporine A ophthalmic solution 0.09% (Cequa®, Sun Pharmaceutical Industries, Cranbury, NJ, United States) [45]. Cyclosporine acts as an immunomodulator for severe DED, reducing inflammation markers and cell death on the ocular surface [46]. Recently, Lifitegrast ophthalmic solution 5% (Xiidra®, Novartis Pharmaceuticals Corporation, Basel, Switzerland) was the first medication approved in the United States for the treatment of DED signs and symptoms [47]. Lifitegrast A inhibits the release of cytokines, interferon δ, TNF-α, and other interleukins preventing the activation of the ocular inflammatory cycle [48]. An additional corticosteroid drug is Loteprednol etabonate ophthalmic suspension (Eysuvis®, Kala Pharmaceuticals, Wtaertown, MA, United States) approved for the treatment of inflammatory flares [48]. Additionally, in 2021, the Food and Drug Administration (FDA) approved Varenicline (Tyrvaya®, Oyster Point Pharma, Princeton, NJ, United States), a nasal spray which stimulates tears, mucins, and oil production and treat both, signs and symptoms [49]. A novel treatment in phase III of clinical trials, NOV03 is a preservative-free eye drop formulation that alleviates the dryness of the ocular surface [50]. The necessity for novel preservative-free formulations has grown in the last years due to the DED increased risk linked with benzalkonium chloride (BAK), a well-known quaternary ammonium compound commonly used in the formulations of numerous ophthalmic preparations [26,51,52]. However, the current, research is focused on the development of novel devices, processes, and medicines to promote tear secretion rather than substitute them with medications or eye drops [53]. Therefore, the identification of additional biochemical pathways involved in the pathophysiology of DED is fundamental to specifically address the research of novel treatments.

2. Oxidative Stress and Inflammation in DED

Oxidative stress is considered one of the main hallmarks of dry eye, leading to inflammation and general discomfort [26,54,55,56]. An imbalance between ROS production and the antioxidant capacity of the organism results in oxidative stress. Consequently, ROS accumulation can be responsible for damage at different levels and particularly on the membrane and ocular surface [57,58,59]. The reactive hydroxyl radical can initiate the lipid peroxidation of cell membranes and induce the accumulation of lipid hydroperoxides leading to membrane disruption and ultimately cell death [60]. Increased levels of lipid hydroperoxide are detected by two major biomarkers, malondialdehyde (MDA) and 4-hydroxynonenal (4-HNE) [20]. The presence of both MDA and 4-HNE have been detected in the tear film and ocular surface in DED patients [61,62]. The increased concentration of lipid hydroperoxides seems to correlate positively with other parameters detected in DED diagnosis, such as tear film break-up time and Schirmer tear volume [25].
Following ROS -induced lipid peroxidation and in response to cellular stresses, mitogen-activated protein (MAP) kinases and nuclear factor kappa- B (NF-kB) are activated and mediate the release of inflammatory cytokines IL-1α, IL-1β, tumour necrosis factor-alpha (TNFα) and metalloproteinase 9 (MMP-9) [63,64,65]. Moreover, the inflammatory response in cornea and conjunctival cells is enhanced by T-cell upregulation of CD3+, CD4+, and CD8+, and the release of additional pro-inflammatory cytokines such as IL-6 and IL-8 [66,67]. Although the implication of inflammation in DED has been known for more than 40 years, its role as cause and effect of DED was established only recently [68]. Moreover, inflammation is also included in the new concept of the ‘‘vicious cycle of inflammation” (Figure 2) proposed by Baudouin et al., where tear film instability, tear hyperosmolarity, apoptosis of corneal/conjunctival cells, and inflammation in the ocular surface are the key features [24]. Breaking the cycle by targeting one of the main factors is considered a valid therapeutic strategy [24]. Starting with tear film instability and hyperosmolarity, the quality of tears can be affected by a decrease of the water content or an increase of evaporation [69]. As a consequence, inflammatory mediators are released in the tear fluid resulting in the release of inflammatory cytokines, leading to damage to the corneal and conjunctival cells and consequent cell death through apoptosis [70]. All these features aggravate the inflammation and maintain the vicious cycle promoting hyperosmolarity and tear film instability.
Based on the implication of oxidative stress and inflammation in dry eye etiopathology and considering the role played by apoptosis in the dry eye vicious cycle, the possible involvement of different types of regulated cell death (RCD) in DED and other ocular surface diseases was investigated [71,72]. In the first part of this review, we introduce the concept of RCD starting from apoptosis. In the second part, the possible role of the most investigated types of RCD, ferroptosis, necroptosis, and pyroptosis, is examined, as well as their potential as a therapeutic target for the treatment of DED. The specific biochemical mechanisms and the hallmarks for ferroptosis, necroptosis, and pyroptosis are described together with DED ocular surface biomarkers (Table 1).

3. Regulated Cell Death (RCD): An Emerging Field

Cell death is considered one of the most important mechanisms to maintain cell homeostasis and integrity of multicellular organisms [78]. Primarily observed by Carl Vogt in 1842, it has been studied for more than two centuries [79]. Over the past decades, many experimental evidence unveiled and characterised a detailed set of endogenous genes involved in the cell death machinery [80]. In 1964, Lockshin and Williams introduced the concept of “programmed cell death” (PCD), based on the observed cells capacity to induce a genetically regulated self-destruction [81,82,83]. In 1972, with the definition of the term “apoptosis” by John Kerr and his group, the concept of “regulated cell death” emerged [84]. Traditionally, a distinction was made between regulated (RCD) and accidental cell death (ACD), based on the different morphological features [80]. Specifically, RCD was used as a synonym for apoptosis, while ACD referred to necrosis [85]. While RCD can undergo specific morphological modification that can be potentially targeted, ACD occurs in response to extreme physical stresses with a nonspecific response preventing any molecular intervention [86]. Since 2005, the Nomenclature Committee on Cell Death (NCCD) formulated guidelines for the classification of different cell death types [87]. They proposed unified criteria that moved from the historical oversimplistic classification of RCD and ACD to a more updated classification based on events associated with specific cell death modalities, their biological context, and effectors [80]. With an increased number of research groups working in the field of regulated cell death, the importance of this phenomenon is now widely recognised and many novel types of non-apoptotic RCD have been identified over the last 20 years [85,88].
Among RCD, apoptosis and inflammatory RCD can be separately distinguished [89]. Apoptosis is characterised by cell shrinkage, with consequent reduction of cellular volume, chromatin condensation, membrane blebbing, nuclear fragmentation, and the final separation of the cellular components into apoptotic bodies collected by neighbouring cells with phagocytic activity and degraded by lysosomes [80,84]. These specific biochemical features distinguish it from the other RCD. More importantly, in apoptosis, there is no disruption of the cellular membrane and, therefore, no inflammation process occurs, whereas inflammatory RCD is characterised by an inflammatory response induced by cell membrane lysis and release of cytosolic material [90]. The most widely studied RCDs are: ferroptosis, an iron-dependent form of regulated cell death driven by enhanced lipid peroxidation which reduces the organism antioxidant defence-inducing cell damage [91]; necroptosis, a programmed form of necrosis dependent on RIPK1-RIPK3 activation of MLKL [92]; pyroptosis, an inflammasome gasdermin D dependent form of RCD [93].
The role of apoptosis in DED has been investigated for several years. A correlation between ocular surface inflammation and the expression of proapoptotic markers (Fas, Fas ligand, APO2.7, CD40, and CD40 ligand) has been found [94]. In chronic dry eye in human and dog models, apoptosis has been demonstrated [70]. Recently, the upregulation of apoptosis in human corneal epithelial cells (HCEpiCs) because of hyperosmolarity has been verified [95]. Vitoux M.A. et al. demonstrated that BAK can induce apoptosis in a concentration-dependent manner in HCE [96]. Whereas, a recent study reported BAK not only as oxidative stress and apoptosis inducers, but also as a trigger of emerging cell death mechanisms [97].
More importantly, apoptosis is included in the vicious cycle of DED as reported by Baudouin and co-workers, underlying the relevant role of conjunctival and corneal cell death in the etiopathology of dry eye [98]. Therefore, the possible implication of the inflammatory RCDs ferroptosis, necroptosis, and pyroptosis as a potential therapeutic target for DED is also examined.

4. Targeting RCD as an Innovative Strategy in DED

4.1. Ferroptosis

Ferroptosis is an iron-catalysed non-apoptotic form of cell death initially described in RAS-mutated oncogenic cells and more recently connected with the pathophysiological processes of many diseases in different vital organs (Figure 3) [99,100]. The key features of ferroptosis are the accumulation of lipid peroxides, abnormal iron metabolism, and reduced levels of glutathione (GSH) as well as GPX4, which can lead to a reduced antioxidant defence of the cells, damage on the phospholipids bilayers, and consequently, cell death [101,102]. Specifically, during lipid peroxidation, a chain reaction of bis-allylic hydrogen abstraction and oxygenation of polyunsaturated fatty acids (PUFAs) of phospholipids, catalysed by redox-active iron, results in the accumulation of toxic lipid hydroperoxides [103]. Iron availability is regulated by two main sources: ferrous iron (Fe2+) in the cytosolic labile iron pool (LIP) and in the catalytic centres of non-heme iron proteins, e.g., lipoxygenase (LOXs) [104]. The former leads to non-enzymatic random oxidation based on Fenton reaction, whereas the latter gives enzymatic oxidation of specific substrates [105]. In addition, the enzyme GPX4 can control iron-dependent lipid peroxidation by the reduction of reactive lipid peroxides in their corresponding inactive lipid alcohols [101]. In order to exert its mechanism of action, the selenocysteine GPX4 requires two electrons provided by the intracellular cofactor GSH, synthesised from cysteine [106]. Cysteine availability is regulated by cystine/glutamate antiporter (System Xc-), which transports cystine inside the cytosol in exchange for glutamate in a 1:1 ratio [107]. Activating or blocking the ferroptosis pathway to alleviate the progression of the disease provides a promising therapeutic strategy [100]. Recent studies reported the implication of ferroptosis in the occurrence and progress of many diseases, such as tumours, neurological diseases, acute kidney injury, and ischemia/reperfusion [108,109,110,111,112]. There are different eye disorders where the implication of lipid hydroperoxide and GPX4 have been underlined, without proving the ferroptosis role. Whereas the presence of the main hallmarks has been detected in retinal cells, photoreceptors, cornea, and conjunctival cells, ferroptosis has been confirmed in the ethiopathology of only a few conditions [113]. Therefore, further research should be undertaken to investigate the potential central role of ferroptosis in other eye disorders [114].

Possible Implication of Ferroptosis in Dry Eye and Ocular Surface Dysfunction

ROS accumulation and modification of the lipid layer on the ocular surface are key features in the pathogenesis of DED as described in Section 2. Oxidative stress and inflammation in DED [115]. Different studies underlined how the loss of cell functions is an important contribution to the alteration of the ocular barrier [35,116,117]. We hypothesise, therefore, that ferroptosis can be considered as a key mechanism in the etiopathology of DED.
The cornea is exposed to many different external and environmental factors [118]. Ultraviolet radiation (UV) and global warming are among the causes that can promote oxidative stress and, consequently, cornea cell dysfunction [114,119]. GPX4 is one of the enzymes that maintains redox homeostasis and promotes wound healing [115,120]. As described in Section 4.1 Ferroptosis, reduced availability of GSH or GPX4 promotes lipid peroxidation and decreases regeneration of the corneal epithelium [121]. The addition of α-tocopherol, a lipophilic antioxidant, and known ferroptosis inhibitor, significantly improved the delay in wound healing [120]. This study suggested how ferroptosis inhibition can help to protect the corneal cells. Recently, Katsinas N. et al. demonstrated the antioxidant and anti-inflammatory activity of a phenolic extract from olive pomace, which can control the imbalanced ROS formation [122]. In an additional study reported by Lovatt M. et al., the use of another well-known ferroptosis inhibitor, Ferrostatin-1, in the Fuch’s endothelial corneal dystrophy (FECD) could prevent the accumulation of lipid peroxides [123]. Recently, Yuan J. and co-worker reported a study where excessive ROS production induced ferroptosis in a DED model [124]. They verified the upregulation of the enzyme aldo-keto reductases (AKR1C1), which is responsible for detoxifying 4-HNE in its corresponding non-toxic byproduct. Moreover, they detected a decrease of the common inflammatory biomarkers expression (TNFα, IL-1β) after Ferrostatin 1 treatment in a mouse model. However, partial ferroptosis was detected in HCE cells under hypertonic conditions and AKR1C1 overexpression. Certainly, additional studies are required to fully confirm the potential role of AKR1C1 in ferroptosis and DED.
The tear fluid contains several antioxidants able to protect the ocular surface from different diseases connected with ROS accumulation [125]. In patients suffering from Sjogren’s syndrome, where there is a lack of tear fluid, a high level of late and early-stage lipid peroxidation biomarkers such as 4-HNE and hexanoyl-lysine (HEL) have been detected [20]. ROS overexpression might be induced by exposure to atmospheric oxygen and/or alteration of antioxidant support, contributing to tear film instability and consequent ocular surface damage and inflammation [58]. Among the most common damage caused by ROS, lipid peroxidation of the membrane, oxidative stress of protein, and oxidative damage of DNA have been highlighted [126]. Wakamatsu T.H. et al. detected an increased lipid peroxidation in the tear film and conjunctival cells [59]. Unfortunately, very little is described regarding the role of epithelial lipids in the cell death process and inflammation involved in eye disorders, particularly in DED. In a recent study reported by Magny R et al., lipid markers were analysed as a consequence of BAK and hyperosmolarity inflammation [127]. In patients suffering from dry eye, an alteration of the cornea epithelial cells due to the accumulation of ROS was observed, reducing the differentiation capacity of the cells and hindering the blinking mechanism [128]. Possibly, the use of a radical scavenging agent might be an interesting therapeutic approach, particularly lipid radical trapping antioxidants (RTAs) could tackle the accumulation of lipid peroxides [129].
The exposure of the ocular surface to oxidative stress together with UV light and environmental stress is also responsible for many dysfunctions of the conjunctiva [118,130]. Dry eye, atopic keratoconjunctivitis, and conjunctivochalasis are only a few of the pathologies that can affect the ocular surface, caused by an alteration of the redox balance [31,58,127,131,132]. Together with other antioxidative enzymes such as GPX1, and superoxide dismutase (SOD1 and SOD2), GPX4 controls redox homeostasis [121,133,134]. Specifically, a reduction of its expression could lead to lipid ROS accumulation, promoting cell deaths in conjunctival cells and diseases associated with oxidative stress. Although apoptosis is the main mechanism of cell death associated with GPX4 loss, the recently discovered ferroptosis could also be considered as a possible mechanism involved in the alteration of the conjunctival cell and different eye diseases [135]. Sakai O. et al., verified the fundamental role of GPX4 in human conjunctival epithelial cells to maintain oxidative homeostasis and protect conjunctival cells from cytotoxicity [136]. Although further evidence is needed, all the elements above suggest that ferroptosis and GPX4 could be novel therapeutic targets for dry eye disease and other ocular surface diseases.
The correlation between ferroptosis and other types of eye diseases is beyond the scope of this review. However, it is relevant to mention that previous studies detected the presence of ferroptosis hallmarks in age-related macular degeneration (AMD) [137,138,139,140,141,142], glaucoma [113,143], retinitis pigmentosa [113,144,145], cataract [132,146], Retinal Ischemia-Reperfusion Injury (RIRI) [113,147], and alkali burn [148].

4.2. Necroptosis

Necroptosis is a caspase-independent form of regulated cell death that can be triggered by TNFα, toll-like receptors TLR3-TLR4, interferon receptor 1 (IFNAR1), Z-DNA binding protein 1 (ZBP1), and Fas (Figure 4) [149]. Necroptosis is regulated by the activity of receptor-interacting serine/threonine kinase 1 (RIPK1), a serine/threonine protein kinase which can be activated through the phosphorylation of its kinase domain. Activated RIPK1 then recruits RIPK3 through the interaction with the conservative RIP homotypic interaction motif (RHIM) which forms the so-called Necrosome [150]. Then RIPK3 phosphorylates the Thr357 and Ser358 residues of downstream protein mixed lineage kinase domain-like (MLKL), which oligomerises and translocates to the plasma membrane [151]. Here, pMLKL can execute necroptosis, inducing direct rupture of the plasma membrane, and form a pore-complex to promote leakage of intracellular material, or can dysregulate the Na+ and Ca2+ channels, leading to increased intracellular osmotic pressure and final cell death [152,153]. The membrane permeabilization induces the release of different pro-inflammatory cytokines, promoting inflammation [92]. Moreover, mitochondrial ROS production contributes to necroptosis induction, although the exact mechanism is not completely understood [154]. RIPK1 can be therapeutically targeted in order to inhibit necroptosis and the consequent inflammatory process. In different organs and pathologies, necroptosis plays a fundamental role and, therefore, has been widely investigated since 2005 [155]. Upregulation of RIPK1 was detected in ischaemia-reperfusion injury (IRI), atherosclerosis, acute kidney injury, and neurodegenerative diseases [156,157,158,159,160,161]. Recently, different studies revealed the possible implication of necroptosis in ocular conditions, with particular attention paid to corneal surface dysregulation [144,162,163,164]. Additionally, RIPK1 also regulates cell survival and a form of RIPK1-dependent apoptosis (Figure 4) [165].

Possible Implication in Dry Eye and Ocular Surface Dysfunction

Recently, Shi K. et al. reported the implication of necroptosis in airborne particulate matter (PM) ocular surface injury [71]. Necroptosis can induce cell death in the cornea epithelial cells following PM exposure. Currently, this is the only study that verified necroptosis on the ocular surface, demonstrating a potential link between necroptosis and DED. Indeed, DED is the most frequent clinical condition induced by PM exposure [71]. Treatment with Nec-1 successfully increased cell viability and reduced ROS formation, confirming the potential of using necroptosis inhibitors to prevent cell death. Moreover, considering the etiopathology of DED, it appears that inflammation is one of the main features together with the redox imbalance [63]. Moreover, TNF-α is a well-described mediator of DED pathophysiology and it is also indicated as an initiator of necroptosis. Particularly, a dramatic increase of ROS level is observed when TNF-α interacts with its receptor TNFR1 [166]. Additionally, in a recent study reported by Kessal K. et al., upregulation of RIPK1 was detected in a mouse dry eye model. However, the role of necroptosis in the inflammatory cascade was not confirmed [167]. Whereas the release of pro-inflammatory cytokines such as IL-1α, IL-1β, IL-6, and IL-8 and consequent activation of MAPK kinases is verified, there are no studies available regarding the involvement of RIPK1-RIPK3-MLKL and necroptosis [168].
Necroptosis is also implicated in different eye diseases. Particularly, the use of RIPK1 inhibitor Necrostatin 1 emerged as a promising therapeutic strategy to control the necroptosis-mediated inflammatory process in dry AMD [113,164,169,170], glaucoma [162,163], and retinitis pigmentosa [144,171].

4.3. Pyroptosis

The term pyroptosis, from the Greek words “pyro” (fire or fever) and “ptosis” (falling), described a novel type of RCD discovered in 2001 by B. Cookson and co-workers (Figure 5) [88,172,173]. The main feature of pyroptosis is the pore formation mediated by gasdermin D (GSDMD) and the activation of the inflammatory response in a caspase 1-dependent (canonical) or independent (non-canonical) way. After stimulation, cells form a cytosolic multiprotein complex, called inflammasome, among which, NOD-like receptor pyrin 3 (NLRP3) inflammasome is the most investigated [174]. NLRP3 is responsible for the release of inflammatory interleukins (IL-1β and IL-18), the formation of an apoptosis-associated speck-like protein (ASC), and the activation of pro-caspase 1. Particularly, caspase 1 (CASP-1) mediates the maturation of pro-IL-1β and pro-IL-18 in their corresponding mature form IL-1β and IL-18, and the cleavage of GSDMD [175]. In the canonical pyroptosis pathway, pathogens can trigger the NLRP3 inflammasome aggregation, lead to CASP-1 activation and GSDMD cleavage [176,177]. The CASP-1-dependent inflammasomes are divided between NLR and non-NLR inflammasomes which can be activated selectively by pathogen-associated molecular patterns (PAMPs) or damage-associated molecular patterns (DAMPs) [178]. Whereas, in the non-canonical pyroptosis pathway, a priming signal represented by microbial lipopolysaccharide (LPS) recognised by TLR4 or endogenous molecules like TNF-α stimulate the oligomerisation of caspase 4 or 5 (in human) or caspase 11 (in mouse) and then cleavage of GSDMD [179]. For both canonical and non-canonical pyroptosis, the final step is the cleavage of GSDMD in the N-terminal fragment (GSDMD-N), which can generate pores on the phospholipids of the plasma membrane and lead to the consequent cell death [180]. In addition, GSDMD cleavage from the non-canonical pathway can also promote an amplification of pyroptosis by stimulating the inflammasome release of CASP-1, the release of IL-1β and IL-18 and activates the inflammatory response [181]. Particularly the latter, can recruit interferon-gamma (IFN-γ) and, therefore, enhance the immune response [182]. In addition, caspase-8 activation by RIPK1 was also recently reported to induce cleavage and activation of the pore-forming molecule GSDMD in macrophages, thereby promoting the induction of a specific form of pyroptosis [183].

Possible Implication in Dry Eye and Ocular Surface Dysfunction

Different cornea disorders can be connected with pyroptosis-pathway activation [162]. In keratitis, which can cause cornea ulcers and consequent blindness, inflammasomes are formed as exogenous infections mediated by DAMPs signalling [184]. Mouse models infected with streptococcus pneumonia and pseudomonas aeruginosa can trigger NLRP3 or NLRC4 inflammatory response [185]. Consequently, caspase 2 can activate the inflammatory mediators leading to the cleavage of GSDMD. Additionally, the non-canonical pyroptosis pathway is involved in the p. aeruginosa keratitis [186]. Targeting caspase 4/5/11 could be a strategy to control the inflammatory response and prevent GSDMD cleavage [187]. In alkali burn, which can affect permanently the cornea transparency, activation of NLPR3 inflammasome and IL-1β were detected and successfully reduced by the use of NLRP3 inhibitors, underlying the role played by pyroptosis in this condition [188].
The elevated level of GSDMD-N detected in dry eye patients supports the hypothesis of pyroptosis implication in DED [189]. Environmental factors, and particularly fine particulate, can induce corneal pyroptosis and, therefore, promote dry eye [190]. A desiccating stress-induced dry eye mouse model with elevated levels of NLRP3 inflammasome, ASC, and CASP1 has been detected together with mature IL-1β and IL-18 [191]. In addition, GSDMD-driven pyroptosis mediated by TLR4 activation has been demonstrated in a mouse model [192]. In vitro, the increased concentration of ROS and hyperosmotic stress promote pyroptosis-mediated inflammation, which was tackled by disulfiram and calcitriol, two pyroptosis inhibitors [193]. The dry eye signs, such as tear loss or inflammatory response, can be aggravated by NLRP12/NLRC4 inflammasome activation mediated GSDMD cleavage together with IL-33 and IL-1β [191]. In dry eye patient tears, the administration of calcitriol effectively alleviates the hyperosmotic stress induced by NLRP3-ASC-CASP1-GSDMD pyroptosis cascade [193]. The administration of dexamethasone attenuated pyroptosis in DED in vitro and in vivo models with decreased expression of inflammatory factors. However, the specific mechanism of action still needs to be completely elucidated [194]. The expression of inflammasome AIM2 and elevated levels of IL-1β was detected in patients suffering from Sjögren’s syndrome [195].
The role of pyroptosis in various eye diseases is also widely studied. The presence of the classical hallmarks are detected not only in vitro, but also in an in vivo rat model, particularly for AMD [196,197,198,199,200]. Additionally, in glaucoma [200,201], cataract [202], and uveitis [203], the implication of pyroptosis was determined. However, different from ferroptosis and necroptosis, the implication of pyroptosis in retinitis pigmentosa is not clarified yet [204].

5. Conclusions

With a higher incidence of dry eye in the worldwide population, the necessity of discovering novel targets and biological pathways involved in the etiopathology of DED has emerged. The vicious cycle is a well-established concept that includes the main features of DED: tear film instability, tear hyperosmolarity, apoptosis, and inflammation. Breaking the vicious cycle is one of the most investigated strategies that can help to alleviate dry eye symptoms and the development of novel therapies. Based on the anatomy of the ocular surface, corneal and conjunctival epithelia are the most exposed to external factors, which can lead to a reduction in the thickness of the external lipid layer and ROS accumulation followed by inflammation and cell death. Among the different cell death mechanisms, only apoptosis has been included in the etiopathology of DED and the vicious cycle [1]. However, considering the complexity of DED, it is reasonable to investigate additional cell death mechanisms. Particularly, the roles of ferroptosis, necroptosis, and pyroptosis are emerging in different ocular diseases and have been verified directly to DED in the case of necroptosis and pyroptosis. The hypothesis of a crosstalk among ferroptosis, necroptosis, and pyroptosis cannot be excluded and left aside, considering the overlay for most of the biochemical pathways involved. Moreover, different studies have underlined the effectiveness of targeting regulated necrosis in various ocular diseases, and more recently, in cornea disorders. Taking all these considerations together and keeping in mind the crosstalk between the redox imbalance and inflammation in DED, as well as the link with the different RCD mechanisms, the research on regulated cell death in DED might be a novel area of study to identify novel therapeutic targets and develop novel therapies for patients.

Author Contributions

Conceptualization, C.S., A.R.L. and K.A.; writing—original draft preparation, C.S.; writing—review and editing, C.S., A.R.L., C.L., G.K. and K.A. All authors have read and agreed to the published version of the manuscript.

Funding

This project has received funding from the European Union’s Horizon 2020 research and innovation programme under the Marie Skłodowska-Curie grant agreements No 765608 (IT-DED3) and No 101065370 (NeuroFerro); from the University of Antwerp, SEP-BOF grant No 44874 and No 44875; from FWO-EOS grant No 30826052 (MODEL-IDI) and grant No 40007512 (CD-INFLADIS); from FWO research project G.0B71.18N and from the FWO-SBO Research project grant No. S001522N (IRONIX).

Data Availability Statement

No new data were created or analyzed in this study. Data sharing is not applicable to this article.

Acknowledgments

Figures created with BioRender.com (accessed on 19 November 2022).

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

4-HNE4-Hydroxynonenal
ACDAccidental Cell Death
ADDEAqueous Deficient Dry Eye
BAKBenzalkonium Chloride
CASP1Caspase 1
DAMPDamage-associated Molecular Patterns
DEDDry Eye Disease
EDEEvaporative dry eye
FDAFood and Drug Administration
GPX4Glutathione Peroxidase 4
GSDMDGasdermin D
GSHGlutathione
HCEHuman corneal epithelial cells
ICAM-1Intracellular adhesion molecule-1
ILInterleukine
IFInterferon
IFNAR1Interferon Receptor 1
JAKJanus Kinases
JNKc-Jun N-Terminal Kinase
LIPLabile Iron Pool
LOXLipoxygenases
MAPKMitogen-Activated Protein Kinase
MDAMalondialdehyde
MGDMeibomian gland dysfunction
MLKLMixed Lineage Kinase Domain Like Pseudokinase
MMPMatric Metalloprotease
NETNeutrophil extracellular trap
NFκBNuclear factor Kappa Beta
NSSDENon-Sjögren Syndrome Dry Eye
OSDIOcular surface disease index
PAMPPathogen-associated Molecular Patterns
PCDProgrammed cell death
PMParticulate Matter
PUFAPolyunsaturated Fatty Acids
RASRat Sarcoma virus protein
RCDRegulated Cell Death
RIPK1Receptor-Interacting serine/threonine-Protein Kinase 1
RIPK3Receptor-Interacting serine/threonine-Protein Kinase 3
ROSRadical Oxygen Species
SODSuperoxide Dismutase
SSDESjögren Syndrome Dry Eye
TLR4Toll-like Receptor 4
TNFTumour Necrosis Factor
ZBP1Z-DNA binding protein 1

References

  1. Stapleton, F.; Alves, M.; Bunya, V.Y.; Jalbert, I.; Lekhanont, K.; Malet, F.; Na, K.S.; Schaumberg, D.; Uchino, M.; Vehof, J.; et al. TFOS DEWS II Epidemiology Report. Ocul. Surf. 2017, 15, 334–365. [Google Scholar] [CrossRef] [PubMed]
  2. Craig, J.P.; Nelson, J.D.; Azar, D.T.; Belmonte, C.; Bron, A.J.; Chauhan, S.K.; de Paiva, C.S.; Gomes, J.A.P.; Hammitt, K.M.; Jones, L.; et al. TFOS DEWS II Report Executive Summary. Ocul. Surf. 2017, 15, 802–812. [Google Scholar] [CrossRef] [PubMed]
  3. Hu, J.W.; Zhu, X.P.; Pan, S.Y.; Yang, H.; Xiao, X.H. Prevalence and Risk Factors of Dry Eye Disease in Young and Middle-Aged Office Employee: A Xi’an Study. Int. J. Ophthalmol. 2021, 14, 567–573. [Google Scholar] [CrossRef] [PubMed]
  4. Rao, S.K.; Mohan, R.; Gokhale, N.; Matalia, H.; Mehta, P. Inflammation and Dry Eye Disease-Where Are We? Int. J. Ophthalmol. 2022, 15, 820–827. [Google Scholar] [CrossRef] [PubMed]
  5. Perrigan, D.; Morgan, A.; Quintero, S.; Perrigin, J.; Brown, S.; Bergmanson, J. Comparison of Osmolarity Values of Selected Ocular Lubricants. Investig. Ophthalmol. Vis. Sci. 2004, 45, 3901. [Google Scholar]
  6. Sullivan, D.A.; Rocha, E.M.; Aragona, P.; Clayton, J.A.; Ding, J.; Golebiowski, B.; Hampel, U.; McDermott, A.M.; Schaumberg, D.A.; Srinivasan, S.; et al. TFOS DEWS II Sex, Gender, and Hormones Report. Ocul. Surf. 2017, 15, 284–333. [Google Scholar] [CrossRef]
  7. Truong, S.; Cole, N.; Stapleton, F.; Golebiowski, B. Sex Hormones and the Dry Eye. Clin. Exp. Optom. 2014, 97, 324–336. [Google Scholar] [CrossRef]
  8. Matossian, C.; McDonald, M.; Donaldson, K.E.; Nichols, K.K.; Maciver, S.; Gupta, P.K. Dry Eye Disease: Consideration for Women’s Health. J. Women’s Health 2019, 28, 502–514. [Google Scholar] [CrossRef] [Green Version]
  9. Gagliano, C.; Caruso, S.; Napolitano, G.; Malaguarnera, G.; Cicinelli, M.V.; Amato, R.; Reibaldi, M.; Incarbone, G.; Bucolo, C.; Drago, F.; et al. Low Levels of 17-β-Oestradiol, Oestrone and Testosterone Correlate with Severe Evaporative Dysfunctional Tear Syndrome in Postmenopausal Women: A Case-Control Study. Br. J. Ophthalmol. 2014, 98, 371–376. [Google Scholar] [CrossRef] [Green Version]
  10. Ablamowicz, A.F.; Nichols, J.J.; Nichols, K.K. Association between Serum Levels of Testosterone and Estradiol with Meibomian Gland Assessments in Postmenopausal Women. Investig. Ophthalmol. Vis. Sci. 2016, 57, 295–300. [Google Scholar] [CrossRef]
  11. Sullivan, D.A.; Sullivan, B.D.; Evans, J.E.; Schirra, F.; Yamagami, H.; Liu, M.; Richards, S.M.; Suzuki, T.; Schaumberg, D.A.; Sullivan, R.M.; et al. Androgen Deficiency, Meibomian Gland Dysfunction, and Evaporative Dry Eye. Ann. N. Y. Acad. Sci. 2002, 966, 211–222. [Google Scholar] [CrossRef]
  12. Sriprasert, I.; Warren, D.W.; Mircheff, A.K.; Stanczyk, F.Z. Dry Eye in Postmenopausal Women: A Hormonal Disorder. Menopause 2016, 23, 343–351. [Google Scholar] [CrossRef]
  13. Nuzzi, R.; Caselgrandi, P. Sex Hormones and Their Effects on Ocular Disorders and Pathophysiology: Current Aspects and Our Experience. Int. J. Mol. Sci. 2022, 23, 3269. [Google Scholar] [CrossRef] [PubMed]
  14. Gomes, J.A.P.; Azar, D.T.; Baudouin, C.; Efron, N.; Hirayama, M.; Horwath-Winter, J.; Kim, T.; Mehta, J.S.; Messmer, E.M.; Pepose, J.S.; et al. TFOS DEWS II Iatrogenic Report. Ocul. Surf. 2017, 15, 511–538. [Google Scholar] [CrossRef] [PubMed]
  15. Baudouin, C.; Labbé, A.; Liang, H.; Pauly, A. Progress in Retinal and Eye Research Preservatives in Eyedrops: The Good, the Bad and the Ugly Q. Prog. Retin. Eye Res. 2010, 29, 312–334. [Google Scholar] [CrossRef] [PubMed]
  16. Fraunfelder, F.T.; Sciubba, J.J.; Mathers, W.D. The Role of Medications in Causing Dry Eye. J. Ophthalmol. 2012, 2012, 285851. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  17. Craig, J.P.; Nichols, K.K.; Akpek, E.K.; Caffery, B.; Dua, H.S.; Joo, C.K.; Liu, Z.; Nelson, J.D.; Nichols, J.J.; Tsubota, K.; et al. TFOS DEWS II Definition and Classification Report. Ocul. Surf. 2017, 15, 276–283. [Google Scholar] [CrossRef] [PubMed]
  18. Jarad, N. Treatment of Dry Eye Disease. Table of Artificial Tears Solutions. Chron. Respir. Dis. 2016, 13, 173–188. [Google Scholar] [CrossRef] [Green Version]
  19. Akpek, E.K.; Bunya, V.Y.; Saldanha, I.J. Sjögren’s Syndrome: More Than Just Dry Eye. Cornea 2019, 38, 658–661. [Google Scholar] [CrossRef] [PubMed]
  20. Wakamatsu, T.H.; Dogru, M.; Matsumoto, Y.; Kojima, T.; Kaido, M.; Ibrahim, O.M.A.; Sato, E.A.; Igarashi, A.; Ichihashi, Y.; Satake, Y.; et al. Evaluation of Lipid Oxidative Stress Status in Sjögren Syndrome Patients. Investig. Ophthalmol. Vis. Sci. 2013, 54, 201–210. [Google Scholar] [CrossRef] [Green Version]
  21. Chhadva, P.; Raquel, G.; Galor, A. Meibomian Gland Disease: The Role of Gland Dysfunction in Dry Eye Disease. Physiol. Behav. 2016, 176, 139–148. [Google Scholar] [CrossRef]
  22. Foulks, G.N. Introduction to the Report of the International Dry Eye WorkShop; Ethis Communications: New York, NY, USA, 2007; ISBN 9786468600. [Google Scholar]
  23. Woodward, A.M.; Senchyna, M.; Argüeso, P. Differential Contribution of Hypertonic Electrolytes to Corneal Epithelial Dysfunction. Exp. Eye Res. 2012, 100, 98–100. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Baudouin, C.; Aragona, P.; Messmer, E.M.; Tomlinson, A.; Calonge, M.; Boboridis, K.G.; Akova, Y.A.; Geerling, G.; Labetoulle, M.; Rolando, M. Role of Hyperosmolarity in the Pathogenesis and Management of Dry Eye Disease: Proceedings of the Ocean Group Meeting. Ocul. Surf. 2013, 11, 246–258. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Nakamura, S.; Shibuya, M.; Nakashima, H.; Hisamura, R.; Masuda, N.; Imagawa, T.; Uehara, M.; Tsubota, K. Involvement of Oxidative Stress on Corneal Epithelial Alterations in a Blink-Suppressed Dry Eye. Investig. Ophthalmol. Vis. Sci. 2007, 48, 1552–1558. [Google Scholar] [CrossRef]
  26. Seen, S.; Tong, L. Dry Eye Disease and Oxidative Stress. Acta Ophthalmol. 2018, 96, e412–e420. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Joossen, C.; Baan, A.; Moreno-Cinos, C.; Joossens, J.; Cools, N.; Moons, L.; Lemmens, K.; Lambeir, A.; Fransen, E.; Delputte, P.; et al. A Novel Serine Protease Inhibitor as Potential Treatment for Dry Eye Syndrome and Ocular Inflammation. Sci. Rep. 2020, 10, 17268. [Google Scholar] [CrossRef] [PubMed]
  28. Clayton, J.A. Dry Eye. N. Engl. J. Med. 2018, 378, 2212–2223. [Google Scholar] [CrossRef]
  29. Hampel, U.; Garreis, F. The Human Meibomian Gland Epithelial Cell Line as a Model to Study Meibomian Gland Dysfunction. Exp. Eye Res. 2017, 163, 46–52. [Google Scholar] [CrossRef]
  30. Balasopoulou, A.; Κokkinos, P.; Pagoulatos, D.; Plotas, P.; Makri, O.E.; Georgakopoulos, C.D.; Vantarakis, A.; Li, Y.; Liu, J.J.; Qi, P.; et al. Anatomy of Cornea and Ocular Surface. BMC Ophthalmol. 2017, 17, 1. [Google Scholar] [CrossRef]
  31. Kumagai, N.; Fukuda, K.; Fujitsu, Y.; Yamamoto, K.; Nishida, T. Role of Structural Cells of the Cornea and Conjunctiva in the Pathogenesis of Vernal Keratoconjunctivitis. Prog. Retin. Eye Res. 2006, 25, 165–187. [Google Scholar] [CrossRef]
  32. Buckner, C.A.; Lafrenie, R.M.; Dénommée, J.A.; Caswell, J.M.; Want, D.A.; Gan, G.G.; Leong, Y.C.; Bee, P.C.; Chin, E.; Teh, A.K.H.; et al. The Physiology of Tear Film. Intech 2021, 11, 13. [Google Scholar]
  33. Cwiklik, L. Tear Film Lipid Layer: A Molecular Level View. Biochim. Biophys. Acta - Biomembr. 2016, 1858, 2421–2430. [Google Scholar] [CrossRef] [PubMed]
  34. Conrady, C.D.; Joos, Z.P.; Patel, B.C.K. Review: The Lacrimal Gland and Its Role in Dry Eye. J. Ophthalmol. 2016, 2016. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Bron, A.J.; Tiffany, J.M.; Gouveia, S.M.; Yokoi, N.; Voon, L.W. Functional Aspects of the Tear Film Lipid Layer. Exp. Eye Res. 2004, 78, 347–360. [Google Scholar] [CrossRef]
  36. Zhang, X.; Vimalin Jeyalatha, M.; Qu, Y.; He, X.; Ou, S.; Bu, J.; Jia, C.; Wang, J.; Wu, H.; Liu, Z.; et al. Dry Eye Management: Targeting the Ocular Surface Microenvironment. Int. J. Mol. Sci. 2017, 18, 1398. [Google Scholar] [CrossRef] [Green Version]
  37. Hori, Y. Secreted Mucins on the Ocular Surface. Investig. Ophthalmol. Vis. Sci. 2018, 59, DES151–DES156. [Google Scholar] [CrossRef] [Green Version]
  38. Shimazaki, J. Definition and Diagnostic Criteria of Dry Eye Disease: Historical Overview and Future Directions. Investig. Ophthalmol. Vis. Sci. 2018, 59, DES7–DES12. [Google Scholar] [CrossRef] [Green Version]
  39. Arif, S.A.; Khan, M.I.; Abid, M.S.; Babar, A.; Arif, M.A.; Jahanzaib, H.M.; Khan, I. Frequency and Impact of Individual Symptoms on Quality of Life in Dry Eye Disease in Patients Presenting to a Tertiary Care Hospital. J. Pak. Med. Assoc. 2021, 71, 1063–1068. [Google Scholar] [CrossRef]
  40. Nichols, K.K. Patient-Reported Symptoms in Dry Dye Disease. Ocul. Surf. 2006, 4, 137–145. [Google Scholar] [CrossRef]
  41. Khanal, S.; Tomlinson, A.; McFadyen, A.; Diaper, C.; Ramaesh, K. Dry Eye Diagnosis. Investig. Ophthalmol. Vis. Sci. 2008, 49, 1407–1414. [Google Scholar] [CrossRef]
  42. Wolffsohn, J.S.; Arita, R.; Chalmers, R.; Djalilian, A.; Dogru, M.; Dumbleton, K.; Gupta, P.K.; Karpecki, P.; Lazreg, S.; Pult, H.; et al. TFOS DEWS II Diagnostic Methodology Report. Ocul. Surf. 2017, 15, 539–574. [Google Scholar] [CrossRef] [PubMed]
  43. Ling, J.; Chan, B.C.L.; Tsang, M.S.M.; Gao, X.; Leung, P.C.; Lam, C.W.K.; Hu, J.M.; Wong, C.K. Current Advances in Mechanisms and Treatment of Dry Eye Disease: Toward Anti-Inflammatory and Immunomodulatory Therapy and Traditional Chinese Medicine. Front. Med. 2022, 8, 815075. [Google Scholar] [CrossRef] [PubMed]
  44. Agarwal, P.; Rupenthal, I.D. Modern Approaches to the Ocular Delivery of Cyclosporine A. Drug Discov. Today 2016, 21, 977–988. [Google Scholar] [CrossRef] [PubMed]
  45. Mandal, A.; Gote, V.; Pal, D.; Ogundele, A.; Mitra, A.K. Ocular Pharmacokinetics of a Topical Ophthalmic Nanomicellar Solution of Cyclosporine (Cequa®) for Dry Eye Disease. Pharm. Res. 2019, 36. [Google Scholar] [CrossRef] [PubMed]
  46. Othman, T.M.; Mousa, A.; Gikandi, P.W.; AbdelMabod, M.; Abdelrahman, A.M. Efficacy and Safety of Using Topical Cyclosporine A for Treatment of Moderate to Severe Dry Eye Disease. Saudi J. Ophthalmol. 2018, 32, 217–221. [Google Scholar] [CrossRef]
  47. Soiza, R.L.; Donaldson, A.I.C.; Myint, P.K. Vaccine against Arteriosclerosis: An Update. Ther. Adv. Vaccines 2018, 9, 259–261. [Google Scholar] [CrossRef] [Green Version]
  48. Beckman, K.; Katz, J.; Majmudar, P.; Rostov, A. Loteprednol Etabonate for the Treatment of Dry Eye Disease. J. Ocul. Pharmacol. Ther. 2020, 36, 497–511. [Google Scholar] [CrossRef]
  49. Katroscik, J.T. FDA Approves New Prescription Medication for Dry Eye Disease. Pharm. Today 2022, 28, 18. [Google Scholar] [CrossRef]
  50. Tauber, J.; Wirta, D.L.; Sall, K.; Majmudar, P.A.; Willen, D.; Krösser, S. A Randomized Clinical Study (SEECASE) to Assess Efficacy, Safety, and Tolerability of NOV03 for Treatment of Dry Eye Disease. Cornea 2021, 40, 1132–1140. [Google Scholar] [CrossRef]
  51. Walsh, K.; Jones, L. The Use of Preservatives in Dry Eye Drops. Clin. Ophthalmol. 2019, 13, 1409–1425. [Google Scholar] [CrossRef] [Green Version]
  52. Tong, L.; Lan, W.; Lim, R.R.; Chaurasia, S.S. S100A Proteins as Molecular Targets in the Ocular Surface Inflammatory Diseases. Ocul. Surf. 2014, 12, 23–31. [Google Scholar] [CrossRef]
  53. Mohamed, H.B.; Abd El-Hamid, B.N.; Fathalla, D.; Fouad, E.A. Current Trends in Pharmaceutical Treatment of Dry Eye Disease: A Review. Eur. J. Pharm. Sci. 2022, 175, 106206. [Google Scholar] [CrossRef]
  54. Talens-Estarelles, C.; García-Marqués, J.V.; Cerviño, A.; García-Lázaro, S. Digital Display Use and Contact Lens Wear: Effects on Dry Eye Signs and Symptoms. Ophthalmic Physiol. Opt. 2022, 797–806. [Google Scholar] [CrossRef]
  55. Al-Mohtaseb, Z.; Schachter, S.; Lee, B.S.; Garlich, J.; Trattler, W. The Relationship between Dry Eye Disease and Digital Screen Use. Clin. Ophthalmol. 2021, 15, 3811–3820. [Google Scholar] [CrossRef]
  56. Behar-Cohen, F.; Baillet, G.; de Ayguavives, T.; Garcia, P.O.; Krutmann, J.; Peña-García, P.; Reme, C.; Wolffsohn, J.S. Ultraviolet Damage to the Eye Revisited: Eye-Sun Protection Factor (E-SPF®), a New Ultraviolet Protection Label for Eyewear. Clin. Ophthalmol. 2014, 8, 87–104. [Google Scholar] [CrossRef] [Green Version]
  57. Deng, R.; Hua, X.; Li, J.; Chi, W.; Zhang, Z.; Lu, F.; Zhang, L.; Pflugfelder, S.C.; Li, D.Q. Oxidative Stress Markers Induced by Hyperosmolarity in Primary Human Corneal Epithelial Cells. PLoS ONE 2015, 10, e0126561. [Google Scholar] [CrossRef] [Green Version]
  58. Macri, A.; Scanarotti, C.; Bassi, A.M.; Giuffrida, S.; Sangalli, G.; Traverso, C.E.; Iester, M. Evaluation of Oxidative Stress Levels in the Conjunctival Epithelium of Patients with or without Dry Eye, and Dry Eye Patients Treated with Preservative-Free Hyaluronic Acid 0.15 % and Vitamin B12 Eye Drops. Graefe’s Arch. Clin. Exp. Ophthalmol. 2015, 253, 425–430. [Google Scholar] [CrossRef]
  59. Wakamatsu, T.H.; Dogru, M.; Ayako, I.; Takano, Y.; Matsumoto, Y.; Ibrahim, O.M.A.; Okada, N.; Satake, Y.; Fukagawa, K.; Shimazaki, J.; et al. Evaluation of Lipid Oxidative Stress Status and Inflammation in Atopic Ocular Surface Disease. Mol. Vis. 2010, 16, 2465–2475. [Google Scholar]
  60. Gaschier, M.M.; Stockwell, B.R. Lipid Peroxidation in Cell Death. Physiol. Behav. 2018, 176, 139–148. [Google Scholar] [CrossRef]
  61. Choi, W.; Lian, C.; Ying, L.; Kim, G.E.; You, I.C.; Park, S.H.; Yoon, K.C. Expression of Lipid Peroxidation Markers in the Tear Film and Ocular Surface of Patients with Non-Sjogren Syndrome: Potential Biomarkers for Dry Eye Disease. Curr. Eye Res. 2016, 41, 1143–1149. [Google Scholar] [CrossRef]
  62. Li, Y.; Zhao, T.; Li, J.; Xia, M.; Li, Y.; Wang, X.; Liu, C.; Zheng, T.; Chen, R.; Kan, D.; et al. Oxidative Stress and 4-Hydroxy-2-Nonenal (4-HNE): Implications in the Pathogenesis and Treatment of Aging-Related Diseases. J. Immunol. Res. 2022, 2022. [Google Scholar] [CrossRef]
  63. Hessen, M.; Akpek, E.K. Dry Eye: An Inflammatory Ocular Disease. J. Ophthalmic Vis. Res. 2014, 9, 240–250. [Google Scholar]
  64. Yamaguchi, T. Inflammatory Response in Dry Eye. Investig. Ophthalmol. Vis. Sci. 2018, 59, DES192–DES199. [Google Scholar] [CrossRef] [Green Version]
  65. Ganesalingam, K.; Ismail, S.; Sherwin, T.; Craig, J.P. Molecular Evidence for the Role of Inflammation in Dry Eye Disease. Clin. Exp. Optom. 2019, 102, 446–454 . [Google Scholar] [CrossRef]
  66. Liu, H.; Gambino, F.; Algenio, C.S.; Wu, C.; Gao, Y.; Bouchard, C.S.; Qiao, L.; Bu, P.; Zhao, S. Inflammation and Oxidative Stress Induced by Lipid Peroxidation Metabolite 4-Hydroxynonenal in Human Corneal Epithelial Cells. Graefe’s Arch. Clin. Exp. Ophthalmol. 2020, 258, 1717–1725. [Google Scholar] [CrossRef]
  67. Lan, W.; Petznick, A.; Heryati, S.; Rifada, M.; Tong, L. Nuclear Factor-ΚB: Central Regulator in Ocular Surface Inflammation and Diseases. Ocul. Surf. 2012, 10, 137–148. [Google Scholar] [CrossRef]
  68. Stern, M.E.; Pflugfelder, S.C. Inflammation in Dry Eye. Ocul. Surf. 2004, 2, 124–130. [Google Scholar] [CrossRef]
  69. Rolando, M.; Zierhut, M. The Ocular Surface and Tear Film and Their Dysfunction in Dry Eye Disease. Surv. Ophthalmol. 2001, 45, 203–210. [Google Scholar] [CrossRef]
  70. Yeh, S.; Song, X.J.; Farley, W.; Li, D.Q.; Stern, M.E.; Pflugfelder, S.C. Apoptosis of Ocular Surface Cells in Experimentally Induced Dry Eye. Investig. Ophthalmol. Vis. Sci. 2003, 44, 124–129. [Google Scholar] [CrossRef] [Green Version]
  71. Shi, K.; Yin, Q.; Tang, X.; Yu, X.; Zheng, S.; Shentu, X. Necroptosis Contributes to Airborne Particulate Matter-Induced Ocular Surface Injury. Toxicology 2022, 470, 153140. [Google Scholar] [CrossRef]
  72. Chen, M.; Rong, R.; Xia, X. Spotlight on Pyroptosis: Role in Pathogenesis and Therapeutic Potential of Ocular Diseases. J. Neuroinflamm. 2022, 19, 183. [Google Scholar] [CrossRef] [PubMed]
  73. Hantera, M. Tear Film Biomarkers in Dry Eye Disease. US Ophthalmic Rev. 2020, 13, 68. [Google Scholar] [CrossRef]
  74. Ahmad, S.S. Update on the Role of Impression Cytology in Ocular Surface Disease. Taiwan J. Ophthalmol. 2017, 8, 53–55. [Google Scholar] [CrossRef]
  75. Chiva, A. Tear Biomarkers in Dry Eye Disease. Eur. Ophthalmic Rev. 2019, 13, 21. [Google Scholar] [CrossRef]
  76. Navel, V.; Sapin, V.; Henrioux, F.; Blanchon, L.; Labbé, A.; Chiambaretta, F.; Baudouin, C.; Dutheil, F. Oxidative and Antioxidative Stress Markers in Dry Eye Disease: A Systematic Review and Meta-Analysis. Acta Ophthalmol. 2022, 100, 45–57. [Google Scholar] [CrossRef]
  77. Akkurt Arslan, M.; Kolman, I.; Pionneau, C.; Chardonnet, S.; Magny, R.; Baudouin, C.; Brignole-Baudouin, F.; Kessal, K. Proteomic Analysis of Tears and Conjunctival Cells Collected with Schirmer Strips Using Timstof pro: Preanalytical Considerations. Metabolites 2022, 12, 2. [Google Scholar] [CrossRef]
  78. Long, J.S.; Ryan, K.M. New Frontiers in Promoting Tumour Cell Death: Targeting Apoptosis, Necroptosis and Autophagy. Oncogene 2012, 31, 5045–5060. [Google Scholar] [CrossRef] [Green Version]
  79. Vogt, C. ALYTES OBSTETRICANS. Biol. Cent. 1842, 2, 1–114. [Google Scholar]
  80. Galluzzi, L.; Vitale, I.; Aaronson, S.A.; Abrams, J.M.; Adam, D.; Agostinis, P.; Alnemri, E.S.; Altucci, L.; Amelio, I.; Andrews, D.W.; et al. Molecular Mechanisms of Cell Death: Recommendations of the Nomenclature Committee on Cell Death 2018. Cell Death Differ. 2018, 25, 486–541. [Google Scholar] [CrossRef]
  81. Lockshin, A.; Williams, M. Programmed Cell Death. Endocrine Potentiation of the Breakdown of the Intersegmental Muscles of Silkmoths. J. Insect Physiol. 1964, 10, 643–649. [Google Scholar] [CrossRef]
  82. Lockshin, R.A.; Williams, C.M. Programmed Cell Death-I. Cytology of Degeneration in the Intersegmental Muscles of the Pernyi Silkmoth. J. Insect Physiol. 1965, 11, 123–133. [Google Scholar] [CrossRef] [PubMed]
  83. Ameisen, J.C. On the Origin, Evolution, and Nature of Programmed Cell Death: A Timeline of Four Billion Years. Cell Death Differ. 2002, 9, 367–393. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Fadeel, B.; Orrenius, S. Apoptosis: A Basic Biological Phenomenon with Wide-Ranging Implications in Human Disease. J. Intern. Med. 2005, 258, 479–517. [Google Scholar] [CrossRef] [PubMed]
  85. Armenta, D.A.; Dixon, S.J. Investigating Nonapoptotic Cell Death Using Chemical Biology Approaches. Cell Chem. Biol. 2020, 27, 376–386. [Google Scholar] [CrossRef] [PubMed]
  86. Galluzzi, L.; Bravo-San Pedro, J.M.; Vitale, I.; Aaronson, S.A.; Abrams, J.M.; Adam, D.; Alnemri, E.S.; Altucci, L.; Andrews, D.; Annicchiarico-Petruzzelli, M.; et al. Essential versus Accessory Aspects of Cell Death: Recommendations of the NCCD 2015. Cell Death Differ. 2015, 22, 58–73. [Google Scholar] [CrossRef] [PubMed]
  87. Kroemer, G.; El-Deiry, W.S.; Golstein, P.; Peter, M.E.; Vaux, D.; Vandenabeele, P.; Zhivotovsky, B.; Blagosklonny, M.V.; Malorni, W.; Knight, R.A.; et al. Classification of Cell Death: Recommendations of the Nomenclature Committee on Cell Death. Cell Death Differ. 2005, 12, 1463–1467. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  88. Tang, D.; Kang, R.; Berghe, T.V.; Vandenabeele, P.; Kroemer, G. The Molecular Machinery of Regulated Cell Death. Cell Res. 2019, 29, 347–364. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  89. Berghe, T.V.; Linkermann, A.; Jouan-Lanhouet, S.; Walczak, H.; Vandenabeele, P. Regulated Necrosis: The Expanding Network of Non-Apoptotic Cell Death Pathways. Nat. Rev. Mol. Cell Biol. 2014, 15, 135–147. [Google Scholar] [CrossRef]
  90. Linkermann, A.; Stockwell, B.R.; Krautwald, S.; Anders, H.J. Regulated Cell Death and Inflammation: An Auto-Amplification Loop Causes Organ Failure. Nat. Rev. Immunol. 2014, 14, 759–767. [Google Scholar] [CrossRef]
  91. Dixon, S.J.; Lemberg, K.M.; Lamprecht, M.R.; Skouta, R.; Zaitsev, E.M.; Gleason, C.E.; Patel, D.N.; Bauer, A.J.; Cantley, A.M.; Yang, W.S.; et al. Ferroptosis: An Iron-Dependent Form of Nonapoptotic Cell Death. Cell 2012, 149, 1060–1072. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  92. Dhuriya, Y.K.; Sharma, D. Necroptosis: A Regulated Inflammatory Mode of Cell Death. J. Neuroinflamm. 2018, 15, 199. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Pasparakis, M.; Vandenabeele, P. Necroptosis and Its Role in Inflammation. Nature 2015, 517, 311–320. [Google Scholar] [CrossRef] [PubMed]
  94. Brignole, F.; De Saint-Jean, M.; Goldschild, M.; Becquet, F.; Goguel, A.; Baudouin, C. Expression of Fas-Fas Ligand Antigens and Apoptotic Marker APO2.7 by the Human Conjunctival Epithelium. Positive Correlation with Class II HLA DR Expression in Inflammatory Ocular Surface Disorders. Exp. Eye Res. 1998, 67, 687–697. [Google Scholar] [CrossRef] [PubMed]
  95. Li, Y.; Liu, H.; Zeng, W.; Wei, J. Edaravone Protects against Hyperosmolarity-Induced Oxidative Stress and Apoptosis in Primary Human Corneal Epithelial Cells. PLoS ONE 2017, 12, e0174437. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. Vitoux, M.A.; Kessal, K.; Melik Parsadaniantz, S.; Claret, M.; Guerin, C.; Baudouin, C.; Brignole-Baudouin, F.; Réaux-Le Goazigo, A. Benzalkonium Chloride-Induced Direct and Indirect Toxicity on Corneal Epithelial and Trigeminal Neuronal Cells: Proinflammatory and Apoptotic Responses in Vitro. Toxicol. Lett. 2020, 319, 74–84. [Google Scholar] [CrossRef]
  97. Bonneau, N.; Brignole-Baudouin, F.; Guerin, C.; Melik-Parsadaniantz, S.; Baudouin, C.; Réaux-Le Goazigo, A. Effects of CX3CR1 deficiency on trigeminal cell death in an in vitro Toxicity-Induced Dry Eye model. Investig. Ophthalmol. Vis. Sci. 2022, 63, 684-F0138. [Google Scholar]
  98. Baudouin, C.; Messmer, E.M.; Aragona, P.; Geerling, G.; Akova, Y.A.; Benítez-Del-Castillo, J.; Boboridis, K.G.; Merayo-Lloves, J.; Rolando, M.; Labetoulle, M. Revisiting the Vicious Circle of Dry Eye Disease: A Focus on the Pathophysiology of Meibomian Gland Dysfunction. Br. J. Ophthalmol. 2016, 100, 300–306. [Google Scholar] [CrossRef]
  99. Hassannia, B.; Vandenabeele, P.; Vanden Berghe, T. Targeting Ferroptosis to Iron Out Cancer. Cancer Cell 2019, 35, 830–849. [Google Scholar] [CrossRef]
  100. Li, J.; Cao, F.; Yin, H.L.; Huang, Z.J.; Lin, Z.T.; Mao, N.; Sun, B.; Wang, G. Ferroptosis: Past, Present and Future. Cell Death Dis. 2020, 11. [Google Scholar] [CrossRef] [Green Version]
  101. Ursini, F.; Maiorino, M. Lipid Peroxidation and Ferroptosis: The Role of GSH and GPx4. Free Radic. Biol. Med. 2020, 152, 175–185. [Google Scholar] [CrossRef]
  102. Dixon, S.J.; Stockwell, B.R. The Hallmarks of Ferroptosis. Annu. Rev. Cancer Biol. 2019, 3, 35–54. [Google Scholar] [CrossRef]
  103. Bayır, H.; Anthonymuthu, T.S.; Tyurina, Y.Y.; Patel, S.J.; Amoscato, A.A.; Lamade, A.M.; Yang, Q.; Vladimirov, G.K.; Philpott, C.C.; Kagan, V.E. Achieving Life through Death: Redox Biology of Lipid Peroxidation in Ferroptosis. Cell Chem. Biol. 2020, 27, 387–408. [Google Scholar] [CrossRef] [PubMed]
  104. Jiang, X.; Stockwell, B.R.; Conrad, M. Ferroptosis: Mechanisms, Biology and Role in Disease. Nat. Rev. Mol. Cell Biol. 2021, 22, 266–282. [Google Scholar] [CrossRef] [PubMed]
  105. Conrad, M.; Pratt, D.A. The Chemical Basis of Ferroptosis. Nat. Chem. Biol. 2019, 15, 1137–1147. [Google Scholar] [CrossRef] [PubMed]
  106. Conrad, M.; Proneth, B. Selenium: Tracing Another Essential Element of Ferroptotic Cell Death. Cell Chem. Biol. 2020, 27, 409–419. [Google Scholar] [CrossRef] [PubMed]
  107. Yang, W.S.; Stockwell, B.R. Ferroptosis: Death by Lipid Peroxidation. Trends Cell Biol. 2016, 26, 165–176. [Google Scholar] [CrossRef]
  108. Zhang, C.; Liu, X.; Jin, S.; Chen, Y.; Guo, R. Ferroptosis in Cancer Therapy: A Novel Approach to Reversing Drug Resistance. Mol. Cancer 2022, 21, 47. [Google Scholar] [CrossRef]
  109. Tan, Q.; Fang, Y.; Gu, Q. Mechanisms of Modulation of Ferroptosis and Its Role in Central Nervous System Diseases. Front. Pharmacol. 2021, 12, 657033. [Google Scholar] [CrossRef]
  110. Ni, L.; Yuan, C.; Wu, X. Targeting Ferroptosis in Acute Kidney Injury. Cell Death Dis. 2022, 13, 182. [Google Scholar] [CrossRef]
  111. Van Coillie, S.; Van San, E.; Goetschalckx, I.; Wiernicki, B.; Mukhopadhyay, B.; Tonnus, W.; Choi, S.M.; Roelandt, R.; Dumitrascu, C.; Lamberts, L.; et al. Targeting Ferroptosis Protects against Experimental (Multi)Organ Dysfunction and Death. Nat. Commun. 2022, 13, 657033. [Google Scholar] [CrossRef]
  112. Luo, L.; Mo, G.; Huang, D. Ferroptosis in Hepatic Ischemia-Reperfusion Injury: Regulatory Mechanisms and New Methods for Therapy (Review). Mol. Med. Rep. 2021, 23, 1. [Google Scholar] [CrossRef] [PubMed]
  113. Peng, J.J.; Song, W.T.; Yao, F.; Zhang, X.; Peng, J.; Luo, X.J.; Xia, X.B. Involvement of Regulated Necrosis in Blinding Diseases: Focus on Necroptosis and Ferroptosis. Exp. Eye Res. 2020, 191, 107922. [Google Scholar] [CrossRef] [PubMed]
  114. Zhang, J.; Sheng, S.; Wang, W.; Dai, J.; Zhong, Y. Molecular Mechanisms of Iron Mediated Programmed Cell Death and Its Roles in Eye Diseases. Front. Nutr. 2022, 9, 844757. [Google Scholar] [CrossRef] [PubMed]
  115. Rowsey, T.G.; Karamichos, D. The Role of Lipids in Corneal Diseases and Dystrophies: A Systematic Review. Clin. Transl. Med. 2017, 6, 30. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  116. Leong, Y.Y.; Tong, L. Barrier Function in the Ocular Surface: From Conventional Paradigms to New Opportunities. Ocul. Surf. 2015, 13, 103–109. [Google Scholar] [CrossRef] [PubMed]
  117. de Paiva, C.S.; St. Leger, A.J.; Caspi, R.R. Mucosal Immunology of the Ocular Surface. Mucosal Immunol. 2022, 15, 1143–1157. [Google Scholar] [CrossRef] [PubMed]
  118. Johnson, G.J. The Environment and the Eye. Eye 2004, 18, 1235–1250. [Google Scholar] [CrossRef] [Green Version]
  119. Cejková, J.; Cejka, C. The Role of Oxidative Stress in Corneal Diseases and Injuries. Histol. Histopathol. 2015, 30, 893–900. [Google Scholar] [CrossRef]
  120. Sakai, O.; Uchida, T.; Imai, H.; Ueta, T. Glutathione Peroxidase 4 Plays an Important Role in Oxidative Homeostasis and Wound Repair in Corneal Epithelial Cells. FEBS Open Bio 2016, 6, 1238–1247. [Google Scholar] [CrossRef]
  121. Ogawa, Y. Glutathione Peroxidase 4, a Unique Antioxidant Enzyme, Plays a Role in Protecting Ocular Surface Mucosal Epithelia. Investig. Ophthalmol. Vis. Sci. 2015, 56, 1657. [Google Scholar] [CrossRef] [Green Version]
  122. Katsinas, N.; Rodríguez-Rojo, S.; Enríquez-De-salamanca, A. Olive Pomace Phenolic Compounds and Extracts Can Inhibit Inflammatory-and Oxidative-Related Diseases of Human Ocular Surface Epithelium. Antioxidants 2021, 10, 1150. [Google Scholar] [CrossRef] [PubMed]
  123. Lovatt, M.; Adnan, K.; Kocaba, V.; Dirisamer, M.; Peh, G.S.L.; Mehta, J.S. Peroxiredoxin-1 Regulates Lipid Peroxidation in Corneal Endothelial Cells. Redox Biol. 2020, 30, 101417. [Google Scholar] [CrossRef] [PubMed]
  124. Zuo, X.; Zeng, H.; Wang, B.; Yang, X.; He, D.; Wang, L.; Ouyang, H.; Yuan, J. AKR1C1 Protects Corneal Epithelial Cells Against Oxidative Stress-Mediated Ferroptosis in Dry Eye. Invest. Ophthalmol. Vis. Sci. 2022, 63, 3. [Google Scholar] [CrossRef] [PubMed]
  125. Bucolo, C.; Fidilio, A.; Platania, C.B.M.; Geraci, F.; Lazzara, F.; Drago, F. Antioxidant and Osmoprotecting Activity of Taurine in Dry Eye Models. J. Ocul. Pharmacol. Ther. 2018, 34, 188–194. [Google Scholar] [CrossRef] [PubMed]
  126. Čejková, J.; Ardan, T.; Šimonová, Z.; Čejka, Č.; Malec, J.; Dotřelova, D.; Brůnová, B. Decreased Expression of Antioxidant Enzymes in the Conjunctival Epithelium of Dry Eye (Sjögren’s Syndrome) and Its Possible Contribution to the Development of Ocular Surface Oxidative Injuries. Histol. Histopathol. 2008, 23, 1477–1483. [Google Scholar] [CrossRef]
  127. Magny, R.; Kessal, K.; Regazzetti, A.; Ben Yedder, A.; Baudouin, C.; Mélik Parsadaniantz, S.; Brignole-Baudouin, F.; Laprévote, O.; Auzeil, N. Lipidomic Analysis of Epithelial Corneal Cells Following Hyperosmolarity and Benzalkonium Chloride Exposure: New Insights in Dry Eye Disease. Biochim. Biophys. Acta - Mol. Cell Biol. Lipids 2020, 1865, 158728. [Google Scholar] [CrossRef] [PubMed]
  128. Augustin, A.J.; Spitznas, M.; Kaviani, N.; Meller, D.; Koch, F.H.J.; Grus, F.; Göbbels, M.J. Oxidative Reactions in the Tear Fluid of Patients Suffering from Dry Eyes. Graefe’s Arch. Clin. Exp. Ophthalmol. 1995, 233, 694–698. [Google Scholar] [CrossRef] [PubMed]
  129. Zilka, O.; Shah, R.; Li, B.; Friedmann Angeli, J.P.; Griesser, M.; Conrad, M.; Pratt, D.A. On the Mechanism of Cytoprotection by Ferrostatin-1 and Liproxstatin-1 and the Role of Lipid Peroxidation in Ferroptotic Cell Death. ACS Cent. Sci. 2017, 3, 232–243. [Google Scholar] [CrossRef]
  130. Dogru, M.; Kojima, T.; Simsek, C.; Tsubotav, K. Potential Role of Oxidative Stress in Ocular Surface Inflammation and Dry Eye Disease. Investig. Ophthalmol. Vis. Sci. 2018, 59, DES163–DES168. [Google Scholar] [CrossRef] [Green Version]
  131. Chhadva, P.; Alexander, A.; McClellan, A.L.; McManus, K.T.; Seiden, B.; Galor, A. The Impact of Conjunctivochalasis on Dry Eye Symptoms and Signs. Investig. Ophthalmol. Vis. Sci. 2015, 56, 2867–2871. [Google Scholar] [CrossRef] [Green Version]
  132. Ward, S.K.; Wakamatsu, T.H.; Dogru, M.; Ibrahim, O.M.A.; Kaido, M.; Ogawa, Y.; Matsumoto, Y.; Igarashi, A.; Ishida, R.; Shimazaki, J.; et al. The Role of Oxidative Stress and Inflammation in Conjunctivochalasis. Investig. Ophthalmol. Vis. Sci. 2010, 51, 1994–2002. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  133. Maiorino, M.; Conrad, M.; Ursini, F. GPx4, Lipid Peroxidation, and Cell Death: Discoveries, Rediscoveries, and Open Issues. Antioxidants Redox Signal. 2018, 29, 61–74. [Google Scholar] [CrossRef] [PubMed]
  134. Nezzar, H.; Mbekeani, J.N.; Noblanc, A.; Chiambaretta, F.; Drevet, J.R.; Kocer, A. Investigation of Antioxidant Systems in Human Meibomian Gland and Conjunctival Tissues. Exp. Eye Res. 2017, 165, 99–104. [Google Scholar] [CrossRef] [PubMed]
  135. Ran, Q.; Liang, H.; Ikeno, Y.; Qi, W.; Prolla, T.A.; Ii, L.J.R.; Wolf, N.; Vanremmen, H.; Richardson, A. Reduction in Glutathione Peroxidase 4 Increases Life Span Through Increased Sensitivity to Apoptosis. J. Gerontol. Ser. A Biol. Sci. Med. Sci. 2007, 62, 932–942. [Google Scholar] [CrossRef] [Green Version]
  136. Sakai, O.; Uchida, T.; Imai, H.; Ueta, T.; Amano, S. Role of Glutathione Peroxidase 4 in Conjunctival Epithelial Cells. Investig. Ophthalmol. Vis. Sci. 2015, 56, 538–543. [Google Scholar] [CrossRef] [Green Version]
  137. Telegina, D.V.; Kozhevnikova, O.S.; Kolosova, N.G. Molecular Mechanisms of Cell Death in Retina during Development of Age-Related Macular Degeneration. Adv. Gerontol. 2017, 7, 17–24. [Google Scholar] [CrossRef]
  138. Totsuka, K.; Ueta, T.; Uchida, T.; Roggia, M.F.; Nakagawa, S.; Vavvas, D.G.; Honjo, M.; Aihara, M. Oxidative Stress Induces Ferroptotic Cell Death in Retinal Pigment Epithelial Cells. Exp. Eye Res. 2019, 181, 316–324. [Google Scholar] [CrossRef]
  139. Henning, Y.; Blind, U.S.; Larafa, S.; Matschke, J.; Fandrey, J. Hypoxia Aggravates Ferroptosis in RPE Cells by Promoting the Fenton Reaction. Cell Death Dis. 2022, 13, 662. [Google Scholar] [CrossRef]
  140. Shu, W.; Dunaief, J.L. Potential Treatment of Retinal Diseases with Iron Chelators. Pharmaceuticals 2018, 11, 112. [Google Scholar] [CrossRef] [Green Version]
  141. Sun, Y.; Zheng, Y.; Wang, C.; Liu, Y. Glutathione Depletion Induces Ferroptosis, Autophagy, and Premature Cell Senescence in Retinal Pigment Epithelial Cells Article. Cell Death Dis. 2018, 9, 753. [Google Scholar] [CrossRef] [Green Version]
  142. Zhao, X.; Gao, M.; Liang, J.; Chen, Y.; Wang, Y. SLC7A11 Reduces Laser-Induced Choroidal Neovascularization by Inhibiting RPE Ferroptosis and VEGF Production. Front. Cell Dev. Biol. 2021, 9, 639851. [Google Scholar] [CrossRef]
  143. Sakamoto, K.; Suzuki, T.; Takahashi, K.; Koguchi, T.; Hirayama, T.; Mori, A.; Nakahara, T.; Nagasawa, H.; Ishii, K. Iron-Chelating Agents Attenuate NMDA-Induced Neuronal Injury via Reduction of Oxidative Stress in the Rat Retina. Exp. Eye Res. 2018, 171, 30–36. [Google Scholar] [CrossRef] [PubMed]
  144. Newton, F.; Megaw, R. Mechanisms of Photoreceptor Death in Retinitis Pigmentosa. Genes (Basel) 2020, 11, 1120. [Google Scholar] [CrossRef] [PubMed]
  145. Lu, L.; Oveson, B.C.; Jo, Y.J.; Lauer, T.W.; Usui, S.; Komeima, K.; Xie, B.; Campochiaro, P.A. Increased Expression of Glutathione Peroxidase 4 Strongly Protects Retina from Oxidative Damage. Antioxid. Redox Signal. 2009, 11, 715–724. [Google Scholar] [CrossRef]
  146. Raman, T.; Ramar, M.; Arumugam, M.; Nabavi, S.M.; Varsha, M.K.N.S. Cytoprotective Mechanism of Action of Curcumin against Cataract. Pharmacol. Reports 2016, 68, 561–569. [Google Scholar] [CrossRef]
  147. Yan, H.F.; Tuo, Q.Z.; Yin, Q.Z.; Lei, P. The Pathological Role of Ferroptosis in Ischemia/Reperfusion-Related Injury. Zool. Res. 2020, 41, 220–230. [Google Scholar] [CrossRef] [PubMed]
  148. Wang, K.; Jiang, L.; Zhong, Y.; Zhang, Y.; Yin, Q.; Li, S.; Zhang, X.; Han, H.; Yao, K. Ferrostatin-1-Loaded Liposome for Treatment of Corneal Alkali Burn via Targeting Ferroptosis. Bioeng. Transl. Med. 2022, 7, e10276. [Google Scholar] [CrossRef] [PubMed]
  149. Grootjans, S.; Berghe, T.V.; Vandenabeele, P. Initiation and Execution Mechanisms of Necroptosis: An Overview. Cell Death Differ. 2017, 24, 1184–1195. [Google Scholar] [CrossRef] [Green Version]
  150. Xie, T.; Peng, W.; Liu, Y.; Yan, C.; Maki, J.; Degterev, A.; Yuan, J.; Shi, Y. Structural Basis of RIP1 Inhibition by Necrostatins. Structure 2013, 21, 493–499. [Google Scholar] [CrossRef] [Green Version]
  151. Cai, Z.; Jitkaew, S.; Zhao, J.; Chiang, H.C.; Choksi, S.; Liu, J.; Ward, Y.; Wu, L.G.; Liu, Z.G. Plasma Membrane Translocation of Trimerized MLKL Protein Is Required for TNF-Induced Necroptosis. Nat. Cell Biol. 2014, 16, 55–65. [Google Scholar] [CrossRef]
  152. Wang, H.; Sun, L.; Su, L.; Rizo, J.; Liu, L.; Wang, L.F.; Wang, F.S.; Wang, X. Mixed Lineage Kinase Domain-like Protein MLKL Causes Necrotic Membrane Disruption upon Phosphorylation by RIP3. Mol. Cell 2014, 54, 133–146. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  153. Gutierrez, K.D.; Davis, M.A.; Daniels, B.P.; Olsen, T.M.; Ralli- Jain, P.; Gale, M., Jr.; Oberst, A. MLKL Activation Triggers NLRP3-Mediated Processing and Release of IL-1β Independent of Gasdermin-D. Physiol. Behav. 2017, 176, 139–148. [Google Scholar] [CrossRef]
  154. Zhang, Y.; Su, S.S.; Zhao, S.; Yang, Z.; Zhong, C.Q.; Chen, X.; Cai, Q.; Yang, Z.H.; Huang, D.; Wu, R.; et al. RIP1 Autophosphorylation Is Promoted by Mitochondrial ROS and Is Essential for RIP3 Recruitment into Necrosome. Nat. Commun. 2017, 8, 14329. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  155. Kearney, C.J.; Martin, S.J. An Inflammatory Perspective on Necroptosis. Mol. Cell 2017, 65, 965–973. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  156. Liu, X.; Xie, X.; Ren, Y.; Shao, Z.; Zhang, N.; Li, L.; Ding, X.; Zhang, L. The Role of Necroptosis in Disease and Treatment. MedComm 2021, 2, 730–755. [Google Scholar] [CrossRef] [PubMed]
  157. Baidya, R.; Crawford, D.H.G.; Gautheron, J.; Wang, H.; Bridle, K.R. Necroptosis in Hepatosteatotic Ischaemia-Reperfusion Injury. Int. J. Mol. Sci. 2020, 21, 5931. [Google Scholar] [CrossRef] [PubMed]
  158. Anders, H.J. Necroptosis in Acute Kidney Injury. Nephron 2018, 139, 342–348. [Google Scholar] [CrossRef]
  159. Coornaert, I.; Hofmans, S.; Devisscher, L.; Augustyns, K.; Van Der Veken, P.; De Meyer, G.R.Y.; Martinet, W. Novel Drug Discovery Strategies for Atherosclerosis That Target Necrosis and Necroptosis. Expert Opin. Drug Discov. 2018, 13, 477–488. [Google Scholar] [CrossRef]
  160. Dionísio, P.A.; Amaral, J.D.; Rodrigues, C.M.P. Molecular Mechanisms of Necroptosis and Relevance for Neurodegenerative Diseases, 1st ed.; Elsevier Inc.: Amsterdam, The Netherlands, 2020; Volume 353. [Google Scholar]
  161. Chalkias, A.; Xanthos, T. Acute Kidney Injury. Lancet 2012, 380, 1904. [Google Scholar] [CrossRef]
  162. Lin, W.; Chen, M.; Cissé, Y.; Chen, X.; Bai, L. Roles and Mechanisms of Regulated Necrosis in Corneal Diseases: Progress and Perspectives. J. Ophthalmol. 2022, 2022, 2695212. [Google Scholar] [CrossRef]
  163. Do, Y.J.; Sul, J.W.; Jang, K.H.; Kang, N.S.; Kim, Y.H.; Kim, Y.G.; Kim, E. A Novel RIPK1 Inhibitor That Prevents Retinal Degeneration in a Rat Glaucoma Model. Exp. Cell Res. 2017, 359, 30–38. [Google Scholar] [CrossRef] [PubMed]
  164. Hanus, J.; Anderson, C.; Wang, S. RPE Necroptosis in Response to Oxidative Stress and in AMD. Physiol. Behav. 2017, 176, 139–148. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  165. Li, X.; Zhang, M.; Huang, X.; Liang, W.; Li, G.; Lu, X.; Li, Y.; Pan, H.; Shi, L.; Zhu, H.; et al. Ubiquitination of RIPK1 Regulates Its Activation Mediated by TNFR1 and TLRs Signaling in Distinct Manners. Nat. Commun. 2020, 11, 6364. [Google Scholar] [CrossRef] [PubMed]
  166. Liang, W.; Chen, M.; Zheng, D.; He, J.; Song, M.; Mo, L.; Feng, J.; Lan, J. A Novel Damage Mechanism: Contribution of the Interaction between Necroptosis and ROS to High Glucose-Induced Injury and Inflammation in H9c2 Cardiac Cells. Int. J. Mol. Med. 2017, 40, 201–208. [Google Scholar] [CrossRef] [Green Version]
  167. Kessal, K.; Daull, P.; Cimbolini, N.; Feraille, L.; Grillo, S.; Docquier, M.; Baudouin, C.; Brignole-Baudouin, F.; Garrigue, J.S. Comparison of Two Experimental Mouse Dry Eye Models through Inflammatory Gene Set Enrichment Analysis Based on a Multiplexed Transcriptomic Approach. Int. J. Mol. Sci. 2021, 22, 10770. [Google Scholar] [CrossRef]
  168. Pflugfelder, S.C.; De Paiva, C.S.; Tong, L.; Luo, L.; Stern, M.E.; Li, D.Q. Stress-Activated Protein Kinase Signaling Pathways in Dry Eye and Ocular Surface Disease. Ocul. Surf. 2005, 3, S154–S157. [Google Scholar] [CrossRef]
  169. Datta, S.; Cano, M.; Ebrahiki, K.; Wang, L.; Handa, J.T. The Impact of Oxidative Stress and Inflammation on RPE Degeneration in Non-Neovascular AMD. Rev. del Col. Am. Cardiol. 2018, 72, 2964–2979. [Google Scholar] [CrossRef]
  170. Jang, K.H.; Do, Y.J.; Koo, T.S.; Choi, J.S.; Song, E.J.; Hwang, Y.; Bae, H.J.; Lee, J.H.; Kim, E. Protective Effect of RIPK1-Inhibitory Compound in in Vivo Models for Retinal Degenerative Disease. Exp. Eye Res. 2019, 180, 8–17. [Google Scholar] [CrossRef]
  171. Murakami, Y.; Matsumoto, H.; Roh, M.; Suzuki, J.; Hisatomi, T.; Ikeda, Y.; Miller, J.W. Receptor Interacting Protein Kinase Mediates Necrotic Cone but Not Rod Cell Death in a Mouse Model of Inherited Degeneration. Proc. Natl. Acad. Sci. USA 2012, 109, 14598–14603. [Google Scholar] [CrossRef] [Green Version]
  172. Fink, S.L.; Cookson, B.T. Apoptosis, Pyroptosis, and Necrosis: Mechanistic Description of Dead and Dying Eukaryotic Cells. Infect. Immun. 2005, 73, 1907–1916. [Google Scholar] [CrossRef] [Green Version]
  173. Cookson, B.T.; Brennan, M.A. Pro-Inflammatory Programmed Cell Death. Trends Microbiol. 2001, 9, 113–114. [Google Scholar] [CrossRef] [PubMed]
  174. Kelley, N.; Jeltema, D.; Duan, Y.; He, Y. The NLRP3 Inflammasome: An Overview of Mechanisms of Activation and Regulation. Int. J. Mol. Sci. 2019, 20, 3328. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  175. Man, S.M.; Karki, R.; Kanneganti, T.-D. Molecular Mechanisms and Functions of Atg11 in Selective Autophagy. Immunol Rev. 2018, 277, 61–75. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  176. Lei, Q.; Yi, T.; Chen, C. NF-ΚB-Gasdermin D (GSDMD) Axis Couples Oxidative Stress and NACHT, LRR and PYD Domains-Containing Protein 3 (NLRP3) Inflammasome-Mediated Cardiomyocyte Pyroptosis Following Myocardial Infarction. Med. Sci. Monit. 2018, 24, 6044–6052. [Google Scholar] [CrossRef] [PubMed]
  177. Wang, Y.; Zhu, X.; Yuan, S.; Wen, S.; Liu, X.; Wang, C.; Qu, Z.; Li, J.; Liu, H.; Sun, L.; et al. TLR4/NF-ΚB Signaling Induces GSDMD-Related Pyroptosis in Tubular Cells in Diabetic Kidney Disease. Front. Endocrinol. (Lausanne) 2019, 10, 603. [Google Scholar] [CrossRef] [Green Version]
  178. Sangiuliano, B.; Pérez, N.M.; Moreira, D.F.; Belizário, J.E. Cell Death-Associated Molecular-Pattern Molecules: Inflammatory Signaling and Control. Mediators Inflamm. 2014, 2014, 821043. [Google Scholar] [CrossRef]
  179. Gritsenko, A.; Yu, S.; Martin-Sanchez, F.; Diaz-del-Olmo, I.; Nichols, E.M.; Davis, D.M.; Brough, D.; Lopez-Castejon, G. Priming Is Dispensable for NLRP3 Inflammasome Activation in Human Monocytes In Vitro. Front. Immunol. 2020, 11, 2318. [Google Scholar] [CrossRef]
  180. Wang, C.; Ruan, J. Mechanistic Insights into Gasdermin Pore Formation and Regulation in Pyroptosis. J. Mol. Biol. 2022, 434, 167297. [Google Scholar] [CrossRef]
  181. Bertheloot, D.; Latz, E.; Franklin, B.S. Necroptosis, Pyroptosis and Apoptosis: An Intricate Game of Cell Death. Cell. Mol. Immunol. 2021, 18, 1106–1121. [Google Scholar] [CrossRef]
  182. Kopitar-Jerala, N. The Role of Interferons in Inflammation and Inflammasome Activation. Front. Immunol. 2017, 8, 1331. [Google Scholar] [CrossRef] [Green Version]
  183. Fritsch, M.; Günther, S.D.; Schwarzer, R.; Albert, M.C.; Schorn, F.; Werthenbach, J.P.; Schiffmann, L.M.; Stair, N.; Stocks, H.; Seeger, J.M.; et al. Caspase-8 Is the Molecular Switch for Apoptosis, Necroptosis and Pyroptosis. Nature 2019, 575, 683–687. [Google Scholar] [CrossRef] [PubMed]
  184. Ting, D.S.J.; Ho, C.S.; Deshmukh, R.; Said, D.G.; Dua, H.S. Infectious Keratitis: An Update on Epidemiology, Causative Microorganisms, Risk Factors, and Antimicrobial Resistance. Eye 2021, 35, 1084–1101. [Google Scholar] [CrossRef]
  185. Sharma, P.; Sharma, N.; Mishra, P.; Joseph, J.; Mishra, D.K.; Garg, P.; Roy, S. Differential Expression of Antimicrobial Peptides in Streptococcus Pneumoniae Keratitis and STAT3-Dependent Expression of LL-37 by Streptococcus Pneumoniae in Human Corneal Epithelial Cells. Pathogens 2019, 8, 31. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  186. Xu, S.; Liu, X.; Liu, X.; Shi, Y.; Jin, X.; Zhang, N.; Li, X. Wedelolactone Ameliorates Pseudomonas aeruginosa -Induced Inflammation and Corneal Injury by Suppressing Caspase-4 / 5 / 11 / GSDMD-Mediated Non-Canonical Pyroptosis. Exp. Eye Res. 2021, 211, 108750. [Google Scholar] [CrossRef]
  187. Qu, W.; Wang, Y.; Wu, Y.; Liu, Y.; Chen, K.; Liu, X.; Zou, Z.; Huang, X.; Wu, M. Triggering Receptors Expressed on Myeloid Cells 2 Promotes Corneal Resistance against Pseudomonas aeruginosa by Inhibiting Caspase-1-Dependent Pyroptosis. Front. Immunol. 2018, 9, 1121. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  188. Bian, F.; Xiao, Y.; Zaheer, M.; Volpe, E.A.; Pflugfelder, S.C.; Li, D.; Paiva, C.S. De Inhibition of NLRP3 Inflammasome Pathway by Butyrate Improves Corneal Wound Healing in Corneal Alkali Burn. Int. J. Mol. Sci. 2017, 18, 562. [Google Scholar] [CrossRef]
  189. Zheng, Q.; Ren, Y.; Reinach, P.S.; She, Y.; Xiao, B.; Hua, S.; Qu, J.; Chen, W. Reactive Oxygen Species Activated NLRP3 Inflammasomes Prime Environment-Induced Murine Dry Eye. Exp. Eye Res. 2014, 125, 1–8. [Google Scholar] [CrossRef]
  190. Niu, L.; Li, L.; Xing, C.; Luo, B.; Hu, C.; Song, M.; Niu, J.; Ruan, Y.; Sun, X.; Lei, Y. Airborne Particulate Matter (PM2.5) Triggers Cornea Inflammation and Pyroptosis via NLRP3 Activation. Ecotoxicol. Environ. Saf. 2021, 207, 111306. [Google Scholar] [CrossRef]
  191. Chen, H.; Gan, X.; Li, Y.; Gu, J.; Liu, Y.; Deng, Y.; Wang, X.; Hong, Y.; Hu, Y.; Su, L.; et al. NLRP12- and NLRC4-Mediated Corneal Epithelial Pyroptosis Is Driven by GSDMD Cleavage Accompanied by IL-33 Processing in Dry Eye. Ocul. Surf. 2020, 18, 783–794. [Google Scholar] [CrossRef]
  192. Wang, B.; Zeng, H.; Zuo, X.; Yang, X.; Wang, X.; He, D.; Yuan, J. TLR4-Dependent DUOX2 Activation Triggered Oxidative Stress and Promoted HMGB1 Release in Dry Eye. Front. Med. 2022, 8, 781616. [Google Scholar] [CrossRef]
  193. Zhang, J.; Dai, Y.; Yang, Y.; Xu, J. Calcitriol Alleviates Hyperosmotic Stress-Induced Corneal Epithelial Cell Damage via Inhibiting the NLRP3-ASC-Caspase-1-GSDMD Pyroptosis Pathway in Dry Eye Disease. J. Inflamm. Res. 2021, 14, 2955. [Google Scholar] [CrossRef] [PubMed]
  194. Li, X.; Jin, X.; Wang, J.; Li, X.; Zhang, H. Dexamethasone Attenuates Dry Eye-Induced Pyroptosis by Regulating the Kcnq1ot1/Mir-214 Cascade. SSRN Electron. J. 2022, 186, 109073. [Google Scholar] [CrossRef]
  195. Vakrakou, A.G.; Svolaki, I.P.; Evangelou, K.; Gorgoulis, V.G.; Manoussakis, M.N. Cell-Autonomous Epithelial Activation of AIM2 (Absent in Melanoma-2) in Fl Ammasome by Cytoplasmic DNA Accumulations in Primary Sjögren’s Syndrome. J. Autoimmun. 2019, 2, 102381. [Google Scholar] [CrossRef]
  196. Ambati, J.; Atkinson, J.P.; Gelfand, B.D. Immunology of Age-Related Macular Degeneration. Nat. Rev. Immunol. 2013, 13, 438–451. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  197. Doyle, S.L.; Campbell, M.; Ozaki, E.; Salomon, R.G.; Mori, A.; Kenna, P.F.; Farrar, G.J.; Kiang, A.S.; Humphries, M.M.; Lavelle, E.C.; et al. NLRP3 Has a Protective Role in Age-Related Macular Degeneration through the Induction of IL-18 by Drusen Components. Nat. Med. 2012, 18, 791–798. [Google Scholar] [CrossRef] [PubMed]
  198. Wang, M.; Su, S.; Jiang, S.; Sun, X.; Wang, J. Role of Amyloid β-Peptide in the Pathogenesis of Age-Related Macular Degeneration. BMJ Open Ophthalmol. 2021, 6, 1–6. [Google Scholar] [CrossRef]
  199. Liu, R.T.; Gao, J.; Cao, S.; Sandhu, N.; Cui, J.Z.; Chou, C.L.; Fang, E.; Matsubara, J.A. Inflammatory Mediators Induced by Amyloid-Beta in the Retina and RPE In Vivo: Implications for Inflammasome Activation in Age-Related Macular Degeneration. Investig. Ophthalmol. Vis. Sci. 2013, 54, 2225–2237. [Google Scholar] [CrossRef] [Green Version]
  200. Kauppinen, A.; Niskanen, H.; Suuronen, T.; Kinnunen, K.; Salminen, A.; Kaarniranta, K. Oxidative Stress Activates NLRP3 Inflammasomes in ARPE-19 Cells-Implications for Age-Related Macular Degeneration (AMD). Immunol. Lett. 2012, 147, 29–33. [Google Scholar] [CrossRef]
  201. Baudouin, C.; Kolko, M.; Melik-Parsadaniantz, S.; Messmer, E.M. Inflammation in Glaucoma: From the Back to the Front of the Eye, and Beyond. Prog. Retin. Eye Res. 2021, 83, 100916. [Google Scholar] [CrossRef]
  202. Rossi, T.; Romano, M.R.; Iannetta, D.; Romano, V.; Gualdi, L.; D’Agostino, I.; Ripandelli, G. Cataract Surgery Practice Patterns Worldwide: A Survey. BMJ Open Ophthalmol. 2021, 6, e000464. [Google Scholar] [CrossRef]
  203. Rosenzweig, H.L.; Woods, A.; Clowers, J.S.; Planck, S.R.; Rosenbaum, J.T. The NLRP3 Inflammasome Is Active but Not Essential in Endotoxin-Induced Uveitis. Inflamm. Res. 2012, 61, 225–231. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  204. Liu, W.; Liu, S.; Li, P.; Yao, K. Retinitis Pigmentosa: Progress in Molecular Pathology and Biotherapeutical Strategies. Int. J. Mol. Sci. 2022, 23, 4883. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Representation of the ocular surface and tear film composition. The tear film is constituted by the external lipid layer which prevents evaporation in physiological conditions, the aqueous layer, responsible for lubrication, and the nutrients content to maintain osmolarity, and the mucin layer which protects the internal corneal epithelium.
Figure 1. Representation of the ocular surface and tear film composition. The tear film is constituted by the external lipid layer which prevents evaporation in physiological conditions, the aqueous layer, responsible for lubrication, and the nutrients content to maintain osmolarity, and the mucin layer which protects the internal corneal epithelium.
Ijms 24 00731 g001
Figure 2. Representation of the vicious cycle of inflammation. Figure adapted from Baudouin C. et al. [24].
Figure 2. Representation of the vicious cycle of inflammation. Figure adapted from Baudouin C. et al. [24].
Ijms 24 00731 g002
Figure 3. Overview of ferroptosis pathway. The system Xc- exchanges glutamate and cystine in a 1:1 ratio. From cystine, GSH is synthesised and available as a substrate for GPX4, responsible for controlling the lipid peroxidation process. Transferrin is responsible for the intake of Fe3+, which is transformed into its reduced form ferrous iron (Fe2+) by the metalloreductase STEAP3. Fe2+ constitutes the labile iron pool and mediates lipid peroxidation via the Fenton reaction. The lipid peroxides chain process is depicted in the central area of the figures. PL = phospholipids; PL• = phospholipid radicals; PLOOH = phospholipid hydroperoxides; PLO• = alkoxyl phospholipid radicals; PLOO• = phospholipid peroxyl radicals; PLOH = phospholipids alcohols.
Figure 3. Overview of ferroptosis pathway. The system Xc- exchanges glutamate and cystine in a 1:1 ratio. From cystine, GSH is synthesised and available as a substrate for GPX4, responsible for controlling the lipid peroxidation process. Transferrin is responsible for the intake of Fe3+, which is transformed into its reduced form ferrous iron (Fe2+) by the metalloreductase STEAP3. Fe2+ constitutes the labile iron pool and mediates lipid peroxidation via the Fenton reaction. The lipid peroxides chain process is depicted in the central area of the figures. PL = phospholipids; PL• = phospholipid radicals; PLOOH = phospholipid hydroperoxides; PLO• = alkoxyl phospholipid radicals; PLOO• = phospholipid peroxyl radicals; PLOH = phospholipids alcohols.
Ijms 24 00731 g003
Figure 4. Overview of the TNF-mediated RIPK1 activation pathway. (1) RIPK1 can mediate cell survival when cIAP1/2 and LUBAC polyubiquitinate different components of TNFR1-complex I, resulting in the downstream activation of TAB2/TAB3/TAK1 and NEMO/IKKα/IKKβ complex. The activation of IKKα/β can promote cell survival by NF-kB-dependent upregulation of pro-survival genes. (2) When complex IIa is formed, it can mediate apoptosis. CYLD deubiquitinates RIPK1 which subsequently is released from complex I. The TRADD-RIPK1 complex recruits FADD, activates caspase 8, and leads to cell death by apoptosis. (3) Complex IIb-mediated apoptosis which is dependent on RIPK1 activity. When no RIPK1 ubiquitination can occur, complex IIb or the riptosome is formed. This results in the induction of apoptotic cell death through a pathway similar to that of complex IIa. (4) Complex IIc (Necrosome)-mediate necroptosis. When deubiquitinated RIPK1 is present and caspases are inactivated, necroptosis will occur as a rescue mechanism, since caspase 8-dependent apoptosis can not happen. The execution of necroptosis is dependent on the kinase activity of both RIPK1 and RIPK3, which activate downstream protein MLKL.
Figure 4. Overview of the TNF-mediated RIPK1 activation pathway. (1) RIPK1 can mediate cell survival when cIAP1/2 and LUBAC polyubiquitinate different components of TNFR1-complex I, resulting in the downstream activation of TAB2/TAB3/TAK1 and NEMO/IKKα/IKKβ complex. The activation of IKKα/β can promote cell survival by NF-kB-dependent upregulation of pro-survival genes. (2) When complex IIa is formed, it can mediate apoptosis. CYLD deubiquitinates RIPK1 which subsequently is released from complex I. The TRADD-RIPK1 complex recruits FADD, activates caspase 8, and leads to cell death by apoptosis. (3) Complex IIb-mediated apoptosis which is dependent on RIPK1 activity. When no RIPK1 ubiquitination can occur, complex IIb or the riptosome is formed. This results in the induction of apoptotic cell death through a pathway similar to that of complex IIa. (4) Complex IIc (Necrosome)-mediate necroptosis. When deubiquitinated RIPK1 is present and caspases are inactivated, necroptosis will occur as a rescue mechanism, since caspase 8-dependent apoptosis can not happen. The execution of necroptosis is dependent on the kinase activity of both RIPK1 and RIPK3, which activate downstream protein MLKL.
Ijms 24 00731 g004
Figure 5. Overview of the pyroptosis pathway. After external stimulation mediated by pathogens, pyroptosis can occur via a canonical or a non-canonical pathway. In the canonical pyroptosis, following the external stimuli, the release of DAMPs or PAMPs can induce the formation of a multiprotein complex, inflammasome. The most studied is NLRP3. Inflammasome mediates the activation of CASP-1, which induces the maturation of IL-1β and IL-18 and the cleavage of GSDMD in the N-terminal fragment GSDMD-N responsible for the pore formation in the phospholipids bilayer. In the non-canonical pyroptosis, microbial LPS induces the oligomerisation of caspase 4/5 (human) or 11 (mouse), which mediates GSDMD cleavage in GSDMD-N and pore formation without enabling maturation of interleukins. At the same time, GSDMD activation via the non-canonical pathway can also promote the amplification of pyroptosis, stimulating the canonical pathway.
Figure 5. Overview of the pyroptosis pathway. After external stimulation mediated by pathogens, pyroptosis can occur via a canonical or a non-canonical pathway. In the canonical pyroptosis, following the external stimuli, the release of DAMPs or PAMPs can induce the formation of a multiprotein complex, inflammasome. The most studied is NLRP3. Inflammasome mediates the activation of CASP-1, which induces the maturation of IL-1β and IL-18 and the cleavage of GSDMD in the N-terminal fragment GSDMD-N responsible for the pore formation in the phospholipids bilayer. In the non-canonical pyroptosis, microbial LPS induces the oligomerisation of caspase 4/5 (human) or 11 (mouse), which mediates GSDMD cleavage in GSDMD-N and pore formation without enabling maturation of interleukins. At the same time, GSDMD activation via the non-canonical pathway can also promote the amplification of pyroptosis, stimulating the canonical pathway.
Ijms 24 00731 g005
Table 1. Overview of the different DED biomarkers of the ocular surface and their possible connection with the different RCD.
Table 1. Overview of the different DED biomarkers of the ocular surface and their possible connection with the different RCD.
DED Biomarkers on the Ocular SurfaceBiomarkers [73]Possible Correlation with RCD
Inflammatory biomarkers [74] TNF-α✓ Necroptosis
MMP-9✕ (Apoptosis)
IL-1β✓ Pyroptosis
IL-6✓ Necroptosis
IL-17A✕ (Apoptosis)
IL-18✓ Pyroptosis
Tear film biomarkers [75,76,77]ROS✓ Ferroptosis, necroptosis
Hyperosmolarity✓ Ferroptosis
Lipid peroxidation✓ Ferroptosis
↓Lactoferrin ✕ n.d.*
↓Lysozyme✕ n.d.*
Chemokyne/cytokines ✓ Necroptosis, pyroptosis
* n.d. = not defined. It is not associated with a specific cell death mechanism.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Scarpellini, C.; Ramos Llorca, A.; Lanthier, C.; Klejborowska, G.; Augustyns, K. The Potential Role of Regulated Cell Death in Dry Eye Diseases and Ocular Surface Dysfunction. Int. J. Mol. Sci. 2023, 24, 731. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms24010731

AMA Style

Scarpellini C, Ramos Llorca A, Lanthier C, Klejborowska G, Augustyns K. The Potential Role of Regulated Cell Death in Dry Eye Diseases and Ocular Surface Dysfunction. International Journal of Molecular Sciences. 2023; 24(1):731. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms24010731

Chicago/Turabian Style

Scarpellini, Camilla, Alba Ramos Llorca, Caroline Lanthier, Greta Klejborowska, and Koen Augustyns. 2023. "The Potential Role of Regulated Cell Death in Dry Eye Diseases and Ocular Surface Dysfunction" International Journal of Molecular Sciences 24, no. 1: 731. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms24010731

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

Article Metrics

Back to TopTop