Diabetic Keratopathy: History
Please note this is an old version of this entry, which may differ significantly from the current revision.

Diabetes mellitus, the most prevalent endocrine disorder, not only impacts the retina but also significantly involves the ocular surface. Diabetes contributes to the development of dry eye disease and induces morphological and functional corneal alterations, particularly affecting nerves and epithelial cells. These changes manifest as epithelial defects, reduced sensitivity, and delayed wound healing, collectively encapsulated in the context of diabetic keratopathy. In advanced stages of this condition, the progression to corneal ulcers and scarring further unfolds, eventually leading to corneal opacities. This critical complication hampers vision and carries the potential for irreversible visual loss. 

  • diabetic complication
  • cornea
  • keratopathy
  • redox
  • pathways
  • molecular
  • targets

1. Introduction

Corneal alterations in diabetes manifest through increased corneal thickness, epithelial defects, fragility, superficial punctate keratitis, delayed and incomplete wound repair, as well as detectable endothelial changes and neuropathy characterized by decreased corneal sensitivity [1]. In fact, peripheral diabetic neuropathy contributes to diabetic keratopathy by damaging corneal nerve fibers due to hyperglycemia. This results in a significant reduction and alteration in subepithelial nerve fiber morphology, leading to reduced corneal surface sensitivity, a main characteristic of diabetic keratopathy [2][3]. Progressive corneal epithelial cell loss, recurrent erosions, ulcers, and scarring events may culminate in devastating corneal opacities, ultimately causing severe visual impairment or blindness [4][5]. Figure 1 illustrates an example of diabetic keratopathy.
Figure 1. Slit lamp photograph of a patient with diabetic keratopathy, suffering from dry eye symptoms and recurrent corneal erosions. Fluorescein staining of the ocular surface reveals fine punctate keratitis (yellow dots), which is typical of dry eye disease (DED), highlighted by yellow arrowheads. In the inferior third of the cornea, an epithelial ridge (yellow horizontal line) is visible, indicative of a recently closed erosion, highlighted by yellow arrows.

2. Corneal Cell Damage in Diabetic Keratopathy

2.1. Redox Signaling in the Diabetic Cornea

Hyperglycemia, the primary contributor to the pathogenesis of DM and its complications, notably affects the corneal epithelium, a crucial tissue involved in diabetic keratopathy. The corneal epithelium, being a major player in this condition, exhibits insulin-independent glucose uptake through glucose transporter-1 (GLUT1), facilitating a continuous influx of glucose into cells [6][7][8]. In instances of heightened metabolic demand, such as during corneal damage, the expression of GLUT1 transporters increases [9]. Furthermore, the corneal epithelium directly receives glucose through transcorneal transport from the aqueous humor, rather than from tears, thereby maintaining high glycogen levels. During DM, the corneal epithelium becomes exposed to high glucose levels [10]. Notably, hyperglycemia triggers an escalation in the production of advanced glycation end products (AGEs) and significantly elevates the levels of ROS [11][12][13][14][15]. Studies by Kim and colleagues revealed that in diabetic corneal cells, the accumulation of AGEs is linked to oxidative DNA damage, measured through 8′-hydroxy-2′-deoxyguanosine levels. This cascade leads to apoptotic activation via the nuclear factor kappa-light-chain-enhancer of activated B cells (NF-kB), contributing to delayed epithelial wound healing in the diabetic cornea [16]. AGEs, generated under hyperglycemic conditions, promote ROS generation by binding to their receptor RAGE (receptor of AGE). This process stimulates enhanced nicotinamide adenine dinucleotide phosphate (NADPH) oxidase (NOX)-related ROS formation and increased phosphorylation of p47phox. This, in turn, activates the c-Jun N-terminal kinase and p38 mitogen-activated protein kinase pathways, culminating in apoptosis and microvascular complications [17][18][19][20]. Additionally, hyperglycemia increases intracellular concentrations of diacylglycerol (DAG), activating protein kinase C (PKC) and subsequently NOX subtypes (NOX1, NOX2, and NOX4), collectively amplifying ROS formation [19][21][22][23][24]. Furthermore, high glucose levels inhibit the epidermal growth factor receptor (EGFR)–phosphatidylinositol 3-kinase (PI3K)/Ak strain transforming (Akt, also known as protein kinase B) pathway through ROS, attenuating corneal epithelial wound healing [25].
As an additional repercussion of the oxidative stress condition, NF-kB, a crucial transcription factor that regulates immune responses and the expression of adhesion molecules, becomes activated. This activation leads to the abnormal production of proinflammatory cytokines, contributing to chronic inflammation. This establishes a positive loop between oxidative stress, inflammation, and apoptosis in diabetic keratopathy [16]. In addition, ROS and RAGE trigger phospholipase C/PKC/extracellular signal-regulated kinase activator protein 1 signaling, promoting fibrogenesis through transforming growth factor beta (TGF-β) expression [26]. Notably, the TGF-β1 isoform, a major subtype expressed in the cornea, induces delayed re-epithelialization and myofibroblast transformation, suggesting its inhibition as a potential strategy to promote corneal wound healing [27]. Of note, RAGE signaling is activated not only by AGEs but also by other ligands, such as the chemokine high-mobility group box 1 protein (HMGB1), whose expression is reported to be elevated in the corneas of diabetic murine models during wound healing. Its inhibition may be a potential molecular target [28].

2.2. Pyroptosis in the Corneal Epithelium Triggered by Hyperglycemia

ROS abundance not only triggers aberrant inflammation via NF-kB but also induces epithelial cell death via pyroptosis. Oxidative stress can activate the nucleotide-binding domain, leucine-rich-containing family, pyrin domain-containing-3 (NLRP3) inflammasome in corneal epithelial cells [29]. The NLRP3 inflammasome is a cellular multiprotein complex that plays a significant role in the immune response. The protein complex is activated when cells are damaged or infected by certain stress factors such as environmental toxins, bacteria, or viruses [30][31][32]. The NLRP3 inflammasome consists of the NLRP3 protein responsible for detecting harmful signals, the apoptosis-associated speck-like protein containing a caspase activation and recruitment domain (ASC), and the caspase-1 protein responsible for the activation of pro-inflammatory cytokines like interleukin (IL)-1ß and IL-18 [24][33]. When the NLRP3 inflammasome is activated, caspase-1 provides cleavage and activation of IL-1β and IL-18, as well as of gasdermin D, whose fragments generate a membrane pore, allowing proinflammatory cytokine release and membrane disruption, finally leading to pyroptosis [34]. These pathological processes lead to cell death in epithelia and stromal keratocytes.

2.3. Involvement of Stroma, Endothelium, and Tight Junctions in Diabetic Damage

Bitirgen et al. demonstrated a reduced basal epithelial cell density, decreased anterior stromal keratocyte counts, and endothelial cell density occurring prior to diabetic retinopathy in individuals with DM type 2 [35]. Zhang and colleagues reported a specific susceptibility of corneal endothelial cells to ultraviolet A-related oxidative damage in a diabetic murine model. This was attributed to an attenuated parkinsonism-associated deglycase 1/nuclear factor erythroid-derived 2-related factor 2 (Nrf2)/nicotinamide adenine dinucleotide (NAD) phosphate (P)H quinone oxidoreductase 1 (NQO1) pathway [36]. Nrf2 activation is closely related to the levels of nuclear deacetylase sirtuin 1 (SIRT1) [37], with an upregulation of SIRT1 associated with increased Nrf2 activation [38]. Both proteins are essential antioxidative regulators, suggesting their significance as molecular targets in antioxidant research [39].
Additionally, hyperglycemia has been reported to impair conjunctival and corneal barrier function, potentially through a loss of laminin-5, a principal component of the corneal basement membrane [40]. Reduced expression of the tight junction component occludin was observed in the human diabetic corneal epithelium by Huang and colleagues [41]. Furthermore, hyperglycemia delays corneal wound healing and reduces cell migration via oxidative stress, through impairing Akt signaling [25][42][43]. More specifically, the phosphatase and tensin homologue protein results upregulated in damaged corneal epithelium, and its inhibition may allow an increase in the PI3K/Akt signaling, accelerating wound healing [44][45].
Figure 2 illustrates the mechanisms leading to a loss of corneal epithelial cells in diabetic keratopathy. The increased loss of corneal epithelial cells can lead to a reduction in the barrier function of the corneal epithelium, thereby favoring superficial punctate keratitis, recurrent erosions, persistent epithelial defects, and infections [46].
Figure 2. Cellular signaling mechanisms that lead to the demise of corneal epithelial cells in the context of diabetic keratopathy. AGEs: advanced glycation end products; Akt: Ak strain transforming (also known as protein kinase B); DAG: diacylglycerol; IL: interleukin; DJ-1: parkinsonism-associated deglycase 1; EGFR: epidermal growth factor receptor; ERK: extracellular signal-regulated kinase; HMGB1: high-mobility group box 1 protein; ICAM-1: intercellular adhesion molecule 1; NF-κB: nuclear factor kappa-light-chain-enhancer of activated B cells; NOX: NADPH oxidase or nicotinamide adenine dinucleotide phosphate oxidase; NQO1: NAD(P)H quinone oxidoreductase 1; Nrf2: nuclear factor erythroid-derived 2-related factor 2; P-p47phox: phosphorylated form of the protein p47phox, part of the NADPH oxidase enzyme system; PKC: protein kinase C; PI3K: phosphatidylinositol 3-kinase; PLC: phospholipase C; PTEN: phosphatase and tensin homologue protein; RAGE: receptor for advanced glycation end products; ROS: reactive oxygen species; TGF-β1: transforming growth factor beta 1; TNF-α: tumor necrosis factor alpha; VCAM-1: vascular cell adhesion molecule 1; VEGF-A: vascular endothelial growth factor A. Upward arrows indicate upregulation or increased activity, downward arrows indicate downregulation or decreased activity.

3. Diabetic Corneal Neuropathy

3.1. Evidence of Corneal Nerve Damage in Diabetes

The cornea represents the most densely innervated structure in the human body, with its nerves contributing to various homeostatic functions such as blinking movements and the release of neuropeptides and growth factors [47][48][49]. The central corneal nerve density is approximately 7000 nociceptors/mm2, making the cornea roughly 300–600 times more sensitive than the skin [50]. Stromal nerve branches are formed by 900–1200 myelinated and unmyelinated axons with a diameter ranging between 0.5 and 5 μm [3]. The peripheral and central nerve endings of the human cornea consist of nociceptive Aδ and C fibers [51]. Stromal nerves are disposed parallelly to the collagen lamellae and subdivide into smaller fascicles as they extend in superficial/anterior direction, creating interconnections within the anterior stromal plexus [49][52]. While some stromal nerves terminate as free endings, a significant number of them directly innervate the keratocytes [52][53], establishing a profound and intricate relationship between corneal epithelial layers and sensory innervation [54], particularly pertinent in cases of DM. The stroma-originated nerves give rise to the sub-basal nerve plexus (SBNP), which innervates the corneal epithelium. SBNP fibers pass through the Bowman’s layer into the epithelium, predominantly in peripheral regions. These fibers form long bundles progressing towards the center, dividing into smaller branches that interconnect, ultimately creating a complex network within the epithelium [48]. Figure 3 illustrates the corneal layers and innervation, emphasizing the distinctive distribution and ramification of stromal nerves throughout the epithelium.
Figure 3. Schematic representation of the corneal layers and innervation. SBNP: sub-basal nerve plexus.
Multiple studies have elucidated the intricate mechanisms of neuroimmune cross-talk within the cornea, primarily mediated through the interplay between neuromodulators expressed by corneal nerves and their receptors in resident corneal leukocytes [55][56]. Corneal sensory innervation releases neurotrophins and neuropeptides, while autonomic innervation secretes catecholamines and acetylcholine [2][50]. Neurotrophic molecules play a crucial role in preserving corneal homeostasis and facilitating corneal wound healing [57]. Notably, neurotrophin-3 (NT-3) and nerve growth factor (NGF) are pivotal neurotrophic agents for ensuring corneal health and repair [58].
The sensory nerves within the cornea produce various neuropeptides responsible for triggering diverse molecular pathways. Among these are substance P, calcitonin gene-related peptide, and α-melanocyte-stimulating hormone, which activate different molecular signaling pathways [59]. Additionally, beyond conventional neurotransmitters, the sympathetic innervation in the cornea also involves serotonin and neuropeptide Y. Meanwhile, as regards the corneal parasympathetic innervation, activity was demonstrated for vasoactive intestinal polypeptide and galanin [59]. Neuropeptides exert their effects by binding to receptors belonging to the superfamily of G protein-coupled receptors, thereby initiating various downstream signaling pathways [59]. Of particular interest in managing diabetic keratopathy might be substances like substance P [60], possibly in combination with insulin-like growth factor 1 [61][62], and vasoactive intestinal polypeptide [63][64]. These substances play roles in promoting wound healing and corneal regeneration. Delayed wound healing, reduced corneal sensitivity, and anomalies in the tear film associated with DM are intricately linked to profound injuries to corneal nerve fibers during the early phases of DM [3][65][66][67]. This correlation underscores an imbalance in the interdependence between corneal epithelium and nerves, leading to damages in both tissues, characteristic of DM-related neurotrophic keratopathy [68][69]. Numerous investigations have detailed critical morphological changes and a pathological decrease in SBNP fibers in DM, even during prediabetes [70][71][72]. Specifically, diabetic patients exhibit reductions in nerve fiber density, branch density, and fiber length [73][74][75]. Structural changes induced by DM extend to the highly innervated stromal cornea, where the accumulation of AGEs contributes to collagen crosslinking and increased corneal thickness [76]. Moreover, abnormal increases in the tortuosity of stromal nerves have been observed in diabetic patients [77]. Overexpression of matrix metalloproteinases (MMP)-3 and -10 in the diabetic stroma indicates significant corneal alterations and remodeling [78]. Rodent diabetic models demonstrated stromal inflammation and edema [79], and in diabetic murine models, dendritic cell infiltration and nerve fiber impairment in the corneal SBNP hint at a neurotrophic function of this cell subpopulation [80]. Moreover, corneal sub-epithelial microneuromas and axonal swelling positively correlate with painful diabetic polyneuropathy, underlining the nuanced relationship between corneal nerve morphology and diabetic complications [81].
In both experimental and clinical studies, the most significant decrease in corneal branch density and nerve fibers occurs in the SBNP, near the epithelium, emphasizing the strong association between corneal neuropathy and diabetic keratopathy [82][83][84][85][86][87]. Reduced corneal keratocytes in diabetic patients have been linked to damage in SBNP fibers [88]. An increase in corneal Langerhans cells is associated with decreased SBNP fiber density [89], while diminished keratocyte density and increased Langerhans cell density have been linked to delayed epithelial wound healing [90]. Furthermore, the variability in serum glucose levels and the duration of DM play crucial roles in the detrimental effects on corneal nerves. For example, individuals with DM under insulin-dependent therapy for five or more years displayed diminished corneal epithelial nerve density and an abundance of nerve loops in the stroma [83]. Longer durations of DM type 2 are positively correlated with the gradual degeneration of SBNP [91]. Moreover, a recent study highlighted morphological changes in corneal stromal nerves in DM, underscoring an association between glucose level variability and the loss of corneal nerve length in the inferior whorl [92].

3.2. Molecular Pathways Leading to Corneal Nerve Damage

From a molecular standpoint, hyperglycemia triggers the activation of several pathways involving mitochondria, AGEs, the polyol pathway, and the PKC cascade. These pathways collectively contribute to an excess of ROS, ultimately leading to cell death and corneal nerve injuries [2][72].

Hyperglycemia-Related Mitochondrial Dysfunction

In the context of DM, the influx of glucose into the mitochondria accelerates the electron transport chain, indirectly promoting the increased formation of superoxide (O2●−), resulting in oxidative stress [11]. Hyperglycemia induces alterations in mitochondrial dynamics, with short-term exposure leading to fission, caspase-3 activation, and oxidative stress, while prolonged exposure may trigger mitochondrial biogenesis [93]. Chronic hyperglycemia contributes to impaired mitochondrial axonal transport due to the accumulation of larger, swollen mitochondria, resulting in decreased nerve conduction velocity, axonal demyelination, and neuron apoptosis [94][95][96]. Importantly, these mitochondrial anomalies can disrupt the corneal neurotrophic interaction by decreasing the levels of NT-3 and NGF [58][97].

AGEs and Axonal Degeneration

During chronic hyperglycemia, AGEs not only overproduce ROS but also form cross-links with structural proteins in peripheral nerves. This process induces the formation of glycated myelin proteins, dysfunctional entities that promote demyelination, impaired regenerative activity, and axonal degeneration [98][99][100]. AGEs also cross-link with collagen molecules in the corneal stroma [76], leading to the glycation of laminin and fibronectin in Descemet’s membrane. This glycation decreases the adhesion of corneal endothelial cells [101], potentially causing endothelial dysfunction and microangiopathy [2].

Effects of the Polyol Pathway and PKC Cascade

The polyol pathway converts glucose into sorbitol and then into fructose, and its activation is a result of hyperglycemia-induced overload in the glycolytic pathway. This alternative metabolic pathway necessitates NADPH and NAD+, leading to a decrease in glutathione (GSH) synthesis and subsequently hydrogen peroxide (H2O2) generation. As NADPH is vital for the regeneration of the antioxidant GSH, its reduced availability results in a diminished antioxidant capacity [102]. Furthermore, an abundance of sorbitol and fructose is negatively correlated with free nerve myoinositol, a molecule crucial for normal nerve conduction velocity [103]. The reduced bioavailability of myoinositol leads to decreased functionality of the membrane sodium–potassium adenosine triphosphatase (Na+/K+ ATPase), compromising nerve conduction velocity [10].
As described above, the PKC pathway is activated in response to high glucose-induced intracellular DAG, causing enhanced ROS generation through NOX activation in corneal epithelial cells [19][21][22][23][24]. Additionally, PKC activation can impair Na+/K+ ATPase function, resulting in reduced nerve conduction and regeneration [104][105].
Figure 4 summarizes the main molecular pathways associated with hyperglycemia that contribute to corneal nerve damage, along with the principal changes in the corneal stroma and epithelium, focusing particularly on the SBNP during diabetic keratopathy.
Figure 4. Hyperglycemia-related molecular pathways initiating corneal nerve damage and changes in the corneal stroma and SBNP during diabetic keratopathy. Hyperglycemia causes mitochondrial dysfunction, altered mitochondrial dynamics, and neurotrophic deficits, which trigger ROS overproduction. In addition, during diabetes, the accumulation of AGEs promotes the glycation of myelin, favoring axonal degeneration, and collagen, inducing endothelial dysfunction. Furthermore, activation of the polyol and PKC signaling pathways trigger, via ROS overproduction, corneal nerve damage, impairing nerve conduction. AGEs: advanced glycated end products; ETC: electron transport chain; Na+/K+ ATPase: sodium–potassium adenosine triphosphatase; NGF: nerve growth factor; NOX: nicotinamide adenine dinucleotide phosphate oxidase; NT-3: neurotrophin-3; PKC: protein kinase C; SBNP: sub-basal nerve plexus. Upward arrows indicate upregulation or increased activity, downward arrows indicate downregulation or decreased activity.

4. Diabetes-Associated Dry Eye Disease Promotes Oxidatives Stress and Inflammation

The tear function unit, encompassing cornea, conjunctiva, lacrimal gland, Meibomian glands, eyelids, and sensory/motor nerve fibers, intricately regulates tear fluid secretion and tear film formation, which are crucial for maintaining and safeguarding the tear film [106]. Disruptions in any component of this unit can result in DED. This condition is not only linked to a deficiency in tear production, but also associated with a status of chronic inflammation and oxidative stress. In this regard, a recent innovative study by Yang and colleagues demonstrated the effectiveness of a gabapentin/nanoceria nanoformulation as a carrier with anti-inflammatory, antiangiogenic, antiapoptotic, and neuroprotective properties in a rabbit model of DED [107].
Regarding DM, chronic hyperglycemia can potentially harm nerve fibers that supply the lacrimal gland, resulting in histological alterations within the lacrimal gland itself [108]. DM-induced corneal nerve fiber loss diminishes the corneal sensitivity, subsequently impairing tear secretion [47]. Moreover, patients with DM often exhibit a reduced blink frequency, a common factor in evaporative DED [109][110]. Conjunctival squamous metaplasia and a decline in conjunctival goblet cells contribute to decreased mucin secretion, diminishing tear film adhesion to the ocular surface and causing tear film instability [111]. Elevated levels of insulin-like growth factor-binding protein 3 (IGFBP3) in diabetic tears act as a negative modulator of IGF-1 signaling in the corneal epithelium, exacerbating corneal impairment and DED in diabetic corneas [112]. Furthermore, increased concentrations of AGEs in the tear fluid initiate inflammatory reactions and oxidative stress on the ocular surface, further impairing corneal epithelial cell function [113][114]. Notably, HMGB1, an alternative ligand of the receptor for RAGE, and dual oxidase 2 (DUOX2), a subtype of NOX, play crucial roles in DED. Hyperosmolarity-induced DUOX2 activation, mediated by toll-like receptor 4 (TLR4)-dependent signaling and exacerbated by ROS excess, leads to HMGB1 release, cell death, and inflammation in a positive loop in human corneal epithelial cells exposed to DED conditions [115]. In a diabetic murine model, hyperglycemia induces a more severe mitochondrial bioenergetic deficiency in lacrimal gland cells compared to corneal cells. This deficiency results in elevated mtDNA damage, reduced mtDNA copies, decreased levels of GSH, contributing to the early onset of DM-induced DED [116].
Collectively, diabetic alterations affecting tear film composition, secretion, and stability manifest as reduced tear volume and decreased break-up time and conjunctival goblet cell density. There are enhanced tear film osmolarity, squamous conjunctival metaplasia, disturbances in the ocular surface microbiota, heightening the risk of bacterial conjunctivitis, and impairment of Meibomian glands [117].
Figure 5 illustrates the tear film sublayers and the detrimental impact of hyperglycemia and ROS on tear production and stability, leading to DED.
Figure 5. Normal and diabetic tear film in comparison, with particular focus on the ROS-related changes in tear film production, composition and stability. AGEs: advanced glycated end products; DED: dry eye disease; DUOX2: dual oxidase 2; GSH: glutathione; HMGB1: high-mobility group box 1 protein; mtDNA: mitochondrial DNA; RAGE: receptor of advanced glycated end products; ROS: reactive oxygen species; TLR4: toll-like receptor 4. Upward arrows indicate upregulation or increased activity, downward arrows indicate downregulation or decreased activity.

This entry is adapted from the peer-reviewed paper 10.3390/antiox13010120

References

  1. Ljubimov, A.V. Diabetic complications in the cornea. Vis. Res. 2017, 139, 138–152.
  2. Markoulli, M.; Flanagan, J.; Tummanapalli, S.S.; Wu, J.; Willcox, M. The impact of diabetes on corneal nerve morphology and ocular surface integrity. Ocul. Surf. 2018, 16, 45–57.
  3. Murphy, P.J.; Patel, S.; Kong, N.; Ryder, R.E.; Marshall, J. Noninvasive assessment of corneal sensitivity in young and elderly diabetic and nondiabetic subjects. Investig. Ophthalmol. Vis. Sci. 2004, 45, 1737–1742.
  4. Mukhija, R.; Gupta, N.; Vashist, P.; Tandon, R.; Gupta, S.K. Population-based assessment of visual impairment and pattern of corneal disease: Results from the CORE (Corneal Opacity Rural Epidemiological) study. Br. J. Ophthalmol. 2020, 104, 994–998.
  5. Chang, Y.-S.; Tai, M.-C.; Ho, C.-H.; Chu, C.-C.; Wang, J.-J.; Tseng, S.-H.; Jan, R.-L. Risk of Corneal Ulcer in Patients with Diabetes Mellitus: A Retrospective Large-Scale Cohort Study. Sci. Rep. 2020, 10, 7388.
  6. Friend, J.; Snip, R.C.; Kiorpes, T.C.; Thoft, R.A. Insulin sensitivity and sorbitol production of the normal rabbit corneal epithelium in vitro. Investig. Ophthalmol. Vis. Sci. 1980, 19, 913–919.
  7. Cunha, D.A.; Carneiro, E.M.; Alves, M.d.C.; Jorge, A.G.; Sousa, S.M.d.; Boschero, A.C.; Saad, M.J.A.; Velloso, L.A.; Rocha, E.M. Insulin secretion by rat lachrymal glands: Effects of systemic and local variables. Am. J. Physiol.-Endocrinol. Metab. 2005, 289, E768–E775.
  8. Mueckler, M. Family of glucose-transporter genes. Implications for glucose homeostasis and diabetes. Diabetes 1990, 39, 6–11.
  9. Stuard, W.L.; Titone, R.; Robertson, D.M. The IGF/Insulin-IGFBP Axis in Corneal Development, Wound Healing, and Disease. Front. Endocrinol. 2020, 11, 24.
  10. Zhu, L.; Titone, R.; Robertson, D.M. The impact of hyperglycemia on the corneal epithelium: Molecular mechanisms and insight. Ocul. Surf. 2019, 17, 644–654.
  11. Babizhayev, M.A.; Strokov, I.A.; Nosikov, V.V.; Savel’yeva, E.L.; Sitnikov, V.F.; Yegorov, Y.E.; Lankin, V.Z. The Role of Oxidative Stress in Diabetic Neuropathy: Generation of Free Radical Species in the Glycation Reaction and Gene Polymorphisms Encoding Antioxidant Enzymes to Genetic Susceptibility to Diabetic Neuropathy in Population of Type I Diabetic Patients. Cell Biochem. Biophys. 2015, 71, 1425–1443.
  12. González, P.; Lozano, P.; Ros, G.; Solano, F. Hyperglycemia and Oxidative Stress: An Integral, Updated and Critical Overview of Their Metabolic Interconnections. Int. J. Mol. Sci. 2023, 24, 9352.
  13. Ramya, R.; Coral, K.; Bharathidevi, S.R. RAGE silencing deters CML-AGE induced inflammation and TLR4 expression in endothelial cells. Exp. Eye Res. 2021, 206, 108519.
  14. Buonfiglio, F.; Böhm, E.W.; Pfeiffer, N.; Gericke, A. Oxidative Stress: A Suitable Therapeutic Target for Optic Nerve Diseases? Antioxidants 2023, 12, 1465.
  15. Böhm, E.W.; Buonfiglio, F.; Voigt, A.M.; Bachmann, P.; Safi, T.; Pfeiffer, N.; Gericke, A. Oxidative stress in the eye and its role in the pathophysiology of ocular diseases. Redox Biol. 2023, 68, 102967.
  16. Kim, J.; Kim, C.-S.; Sohn, E.; Jeong, I.-H.; Kim, H.; Kim, J.S. Involvement of advanced glycation end products, oxidative stress and nuclear factor-kappaB in the development of diabetic keratopathy. Graefe’s Arch. Clin. Exp. Ophthalmol. 2011, 249, 529–536.
  17. Shi, L.; Yu, X.; Yang, H.; Wu, X. Advanced Glycation End Products Induce Human Corneal Epithelial Cells Apoptosis through Generation of Reactive Oxygen Species and Activation of JNK and p38 MAPK Pathways. PLoS ONE 2013, 8, e66781.
  18. Cepas, V.; Collino, M.; Mayo, J.C.; Sainz, R.M. Redox Signaling and Advanced Glycation Endproducts (AGEs) in Diet-Related Diseases. Antioxidants 2020, 9, 142.
  19. Chen, Y.-H.; Chen, Z.-W.; Li, H.-M.; Yan, X.-F.; Feng, B. AGE/RAGE-induced EMP release via the NOX-derived ROS pathway. J. Diabetes Res. 2018, 2018, 6823058.
  20. Dobi, A.; Bravo, S.B.; Veeren, B.; Paradela-Dobarro, B.; Álvarez, E.; Meilhac, O.; Viranaicken, W.; Baret, P.; Devin, A.; Rondeau, P. Advanced glycation end-products disrupt human endothelial cells redox homeostasis: New insights into reactive oxygen species production. Free Radic. Res. 2019, 53, 150–169.
  21. Hink, U.; Li, H.; Mollnau, H.; Oelze, M.; Matheis, E.; Hartmann, M.; Skatchkov, M.; Thaiss, F.; Stahl, R.A.K.; Warnholtz, A.; et al. Mechanisms Underlying Endothelial Dysfunction in Diabetes Mellitus. Circ. Res. 2001, 88, e14–e22.
  22. Giordo, R.; Nasrallah, G.K.; Posadino, A.M.; Galimi, F.; Capobianco, G.; Eid, A.H.; Pintus, G. Resveratrol-Elicited PKC Inhibition Counteracts NOX-Mediated Endothelial to Mesenchymal Transition in Human Retinal Endothelial Cells Exposed to High Glucose. Antioxidants 2021, 10, 224.
  23. Kolczynska, K.; Loza-Valdes, A.; Hawro, I.; Sumara, G. Diacylglycerol-evoked activation of PKC and PKD isoforms in regulation of glucose and lipid metabolism: A review. Lipids Health Dis. 2020, 19, 113.
  24. Yu, W.; Tao, M.; Zhao, Y.; Hu, X.; Wang, M. 4′-Methoxyresveratrol Alleviated AGE-Induced Inflammation via RAGE-Mediated NF-κB and NLRP3 Inflammasome Pathway. Molecules 2018, 23, 1447.
  25. Xu, K.P.; Li, Y.; Ljubimov, A.V.; Yu, F.S.X. High glucose suppresses epidermal growth factor receptor/phosphatidylinositol 3-kinase/Akt signaling pathway and attenuates corneal epithelial wound healing. Diabetes 2009, 58, 1077–1085.
  26. Taguchi, K.; Fukami, K. RAGE signaling regulates the progression of diabetic complications. Front. Pharmacol. 2023, 14, 1128872.
  27. Carrington, L.M.; Albon, J.; Anderson, I.; Kamma, C.; Boulton, M. Differential Regulation of Key Stages in Early Corneal Wound Healing by TGF-β Isoforms and Their Inhibitors. Investig. Ophthalmol. Vis. Sci. 2006, 47, 1886–1894.
  28. Hou, Y.; Lan, J.; Zhang, F.; Wu, X. Expression profiles and potential corneal epithelial wound healing regulation targets of high-mobility group box 1 in diabetic mice. Exp. Eye Res. 2021, 202, 108364.
  29. Wan, L.; Bai, X.; Zhou, Q.; Chen, C.; Wang, H.; Liu, T.; Xue, J.; Wei, C.; Xie, L. The advanced glycation end-products (AGEs)/ROS/NLRP3 inflammasome axis contributes to delayed diabetic corneal wound healing and nerve regeneration. Int. J. Biol. Sci. 2022, 18, 809–825.
  30. Yu, P.; Zhang, X.; Liu, N.; Tang, L.; Peng, C.; Chen, X. Pyroptosis: Mechanisms and diseases. Signal Transduct. Target. Ther. 2021, 6, 128.
  31. Wooff, Y.; Fernando, N.; Wong, J.H.C.; Dietrich, C.; Aggio-Bruce, R.; Chu-Tan, J.A.; Robertson, A.A.B.; Doyle, S.L.; Man, S.M.; Natoli, R. Caspase-1-dependent inflammasomes mediate photoreceptor cell death in photo-oxidative damage-induced retinal degeneration. Sci. Rep. 2020, 10, 2263.
  32. Evavold, C.L.; Hafner-Bratkovič, I.; Devant, P.; D’Andrea, J.M.; Ngwa, E.M.; Boršić, E.; Doench, J.G.; LaFleur, M.W.; Sharpe, A.H.; Thiagarajah, J.R. Control of gasdermin D oligomerization and pyroptosis by the Ragulator-Rag-mTORC1 pathway. Cell 2021, 184, 4495–4511.e4419.
  33. Abderrazak, A.; Syrovets, T.; Couchie, D.; El Hadri, K.; Friguet, B.; Simmet, T.; Rouis, M. NLRP3 inflammasome: From a danger signal sensor to a regulatory node of oxidative stress and inflammatory diseases. Redox Biol. 2015, 4, 296–307.
  34. Mauro, A.G.; Hunter, K.; Salloum, F.N. Chapter Five—Cardiac complications of cancer therapies. In Advances in Cancer Research; Gewirtz, D.A., Fisher, P.B., Eds.; Academic Press: Cambridge, MA, USA, 2022; Volume 155, pp. 167–214.
  35. Bitirgen, G.; Ozkagnici, A.; Malik, R.; Kerimoglu, H. Corneal nerve fibre damage precedes diabetic retinopathy in patients with type 2 diabetes mellitus. Diabet. Med. 2014, 31, 431–438.
  36. Zhang, X.; Qiu, J.; Huang, F.; Shan, K.; Zhang, C. Type 2 Diabetes Mellitus Makes Corneal Endothelial Cells Vulnerable to Ultraviolet A-Induced Oxidative Damage Via Decreased DJ-1/Nrf2/NQO1 Pathway. Invest. Ophthalmol. Vis. Sci. 2022, 63, 25.
  37. Xu, J.J.; Cui, J.; Lin, Q.; Chen, X.Y.; Zhang, J.; Gao, E.H.; Wei, B.; Zhao, W. Protection of the enhanced Nrf2 deacetylation and its downstream transcriptional activity by SIRT1 in myocardial ischemia/reperfusion injury. Int. J. Cardiol. 2021, 342, 82–93.
  38. Singh, C.K.; Chhabra, G.; Ndiaye, M.A.; Garcia-Peterson, L.M.; Mack, N.J.; Ahmad, N. The Role of Sirtuins in Antioxidant and Redox Signaling. Antioxid. Redox Signal 2018, 28, 643–661.
  39. Wang, M.X.; Zhao, J.; Zhang, H.; Li, K.; Niu, L.Z.; Wang, Y.P.; Zheng, Y.J. Potential Protective and Therapeutic Roles of the Nrf2 Pathway in Ocular Diseases: An Update. Oxid. Med. Cell Longev. 2020, 2020, 9410952.
  40. Lu, W.; Ebihara, N.; Miyazaki, K.; Murakami, A. Reduced Expression of Laminin-5 in Corneal Epithelial Cells Under High Glucose Condition. Cornea 2006, 25, 61–67.
  41. Huang, C.; Liao, R.; Wang, F.; Tang, S. Characteristics of Reconstituted Tight Junctions After Corneal Epithelial Wounds and Ultrastructure Alterations of Corneas in Type 2 Diabetic Rats. Curr. Eye Res. 2016, 41, 783–790.
  42. Wang, Y.; Zhao, X.; Shi, D.; Chen, P.; Yu, Y.; Yang, L.; Xie, L. Overexpression of SIRT1 Promotes High Glucose–Attenuated Corneal Epithelial Wound Healing via p53 Regulation of the IGFBP3/IGF-1R/AKT Pathway. Investig. Ophthalmol. Vis. Sci. 2013, 54, 3806–3814.
  43. Jiang, Q.-W.; Kaili, D.; Freeman, J.; Lei, C.-Y.; Geng, B.-C.; Tan, T.; He, J.-F.; Shi, Z.; Ma, J.-J.; Luo, Y.-H. Diabetes inhibits corneal epithelial cell migration and tight junction formation in mice and human via increasing ROS and impairing Akt signaling. Acta Pharmacol. Sin. 2019, 40, 1205–1211.
  44. Cao, L.; Graue-Hernandez, E.O.; Tran, V.; Reid, B.; Pu, J.; Mannis, M.J.; Zhao, M. Downregulation of PTEN at Corneal Wound Sites Accelerates Wound Healing through Increased Cell Migration. Investig. Ophthalmol. Vis. Sci. 2011, 52, 2272–2278.
  45. Li, J.; Qi, X.; Wang, X.; Li, W.; Li, Y.; Zhou, Q. PTEN Inhibition Facilitates Diabetic Corneal Epithelial Regeneration by Reactivating Akt Signaling Pathway. Transl. Vis. Sci. Technol. 2020, 9, 5.
  46. Gekka, M.; Miyata, K.; Nagai, Y.; Nemoto, S.; Sameshima, T.; Tanabe, T.; Maruoka, S.; Nakahara, M.; Kato, S.; Amano, S. Corneal epithelial barrier function in diabetic patients. Cornea 2004, 23, 35–37.
  47. Mansoor, H.; Tan, H.C.; Lin, M.T.Y.; Mehta, J.S.; Liu, Y.C. Diabetic Corneal Neuropathy. J. Clin. Med. 2020, 9, 3956.
  48. Shaheen, B.S.; Bakir, M.; Jain, S. Corneal nerves in health and disease. Surv. Ophthalmol. 2014, 59, 263–285.
  49. He, J.; Bazan, N.G.; Bazan, H.E.P. Mapping the entire human corneal nerve architecture. Exp. Eye Res. 2010, 91, 513–523.
  50. Müller, L.J.; Marfurt, C.F.; Kruse, F.; Tervo, T.M. Corneal nerves: Structure, contents and function. Exp. Eye Res. 2003, 76, 521–542.
  51. Müller, L.J.; Vrensen, G.F.; Pels, L.; Cardozo, B.N.; Willekens, B. Architecture of human corneal nerves. Invest. Ophthalmol. Vis. Sci. 1997, 38, 985–994.
  52. Müller, L.J.; Pels, L.; Vrensen, G. Ultrastructural organization of human corneal nerves. Investig. Ophthalmol. Vis. Sci. 1996, 37, 476–488.
  53. Dartt, D.A.; Bex, P.; D’Amore, P.; Dana, R.; Mcloon, L.; Niederkorn, J. Ocular Periphery and Disorders; Academic Press: Cambridge, MA, USA, 2011.
  54. Belmonte, C.; Garcia-Hirschfeld, J.; Gallar, J. Neurobiology of ocular pain. Prog. Retin. Eye Res. 1997, 16, 117–156.
  55. Maugeri, G.; D’Amico, A.G.; Amenta, A.; Saccone, S.; Federico, C.; Reibaldi, M.; Russo, A.; Bonfiglio, V.; Avitabile, T.; Longo, A.; et al. Protective effect of PACAP against ultraviolet B radiation-induced human corneal endothelial cell injury. Neuropeptides 2020, 79, 101978.
  56. Wu, M.; Hill, L.J.; Downie, L.E.; Chinnery, H.R. Neuroimmune crosstalk in the cornea: The role of immune cells in corneal nerve maintenance during homeostasis and inflammation. Prog. Retin. Eye Res. 2022, 91, 101105.
  57. Bikbova, G.; Oshitari, T.; Baba, T.; Yamamoto, S. Neuronal changes in the diabetic cornea: Perspectives for neuroprotection. BioMed Res. Int. 2016, 2016, 5140823.
  58. Tomlinson, D.; Fernyhough, P.; Diemel, L. Role of neurotrophins in diabetic neuropathy and treatment with nerve growth factors. Diabetes 1997, 46, S43–S49.
  59. Puri, S.; Kenyon, B.M.; Hamrah, P. Immunomodulatory Role of Neuropeptides in the Cornea. Biomedicines 2022, 10, 1985.
  60. Yang, L.; Di, G.; Qi, X.; Qu, M.; Wang, Y.; Duan, H.; Danielson, P.; Xie, L.; Zhou, Q. Substance P promotes diabetic corneal epithelial wound healing through molecular mechanisms mediated via the neurokinin-1 receptor. Diabetes 2014, 63, 4262–4274.
  61. Chikamoto, N.; Chikama, T.-i.; Yamada, N.; Nishida, T.; Ishimitsu, T.; Kamiya, A. Efficacy of substance P and insulin-like growth factor-1 peptides for preventing postsurgical superficial punctate keratopathy in diabetic patients. Jpn. J. Ophthalmol. 2009, 53, 464–469.
  62. Nagano, T.; Nakamura, M.; Nakata, K.; Yamaguchi, T.; Takase, K.; Okahara, A.; Ikuse, T.; Nishida, T. Effects of Substance P and IGF-1 in Corneal Epithelial Barrier Function and Wound Healing in a Rat Model of Neurotrophic Keratopathy. Investig. Ophthalmol. Vis. Sci. 2003, 44, 3810–3815.
  63. Yang, L.W.Y.; Mehta, J.S.; Liu, Y.-C. Corneal neuromediator profiles following laser refractive surgery. Neural Regen. Res. 2021, 16, 2177.
  64. Zhang, Y.; Gao, N.; Wu, L.; Lee, P.S.; Me, R.; Dai, C.; Xie, L.; Yu, F.-S.X. Role of VIP and sonic hedgehog signaling pathways in mediating epithelial wound healing, sensory nerve regeneration, and their defects in diabetic corneas. Diabetes 2020, 69, 1549–1561.
  65. Dogru, M.; Katakami, C.; Inoue, M. Tear function and ocular surface changes in noninsulin-dependent diabetes mellitus. Ophthalmology 2001, 108, 586–592.
  66. Cousen, P.; Cackett, P.; Bennett, H.; Swa, K.; Dhillon, B. Tear production and corneal sensitivity in diabetes. J. Diabetes Its Complicat. 2007, 21, 371–373.
  67. Kabosova, A.; Kramerov, A.A.; Aoki, A.M.; Murphy, G.; Zieske, J.D.; Ljubimov, A.V. Human diabetic corneas preserve wound healing, basement membrane, integrin and MMP-10 differences from normal corneas in organ culture. Exp. Eye Res. 2003, 77, 211–217.
  68. Al-Aqaba, M.A.; Dhillon, V.K.; Mohammed, I.; Said, D.G.; Dua, H.S. Corneal nerves in health and disease. Prog. Retin. Eye Res. 2019, 73, 100762.
  69. Dua, H.S.; Said, D.G.; Messmer, E.M.; Rolando, M.; Benitez-del-Castillo, J.M.; Hossain, P.N.; Shortt, A.J.; Geerling, G.; Nubile, M.; Figueiredo, F.C. Neurotrophic keratopathy. Prog. Retin. Eye Res. 2018, 66, 107–131.
  70. Barsegian, A.; Lee, J.; Salifu, M.O.; McFarlane, S.I. Corneal neuropathy: An underrated manifestation of diabetes mellitus. J. Clin. Endocrinol. Diabetes 2018, 2.
  71. De Clerck, E.E.B.; Schouten, J.; Berendschot, T.; Koolschijn, R.S.; Nuijts, R.; Schram, M.T.; Schaper, N.C.; Henry, R.M.A.; Dagnelie, P.C.; Ruggeri, A.; et al. Reduced corneal nerve fibre length in prediabetes and type 2 diabetes: The Maastricht Study. Acta Ophthalmol. 2020, 98, 485–491.
  72. Zhou, T.; Lee, A.; Lo, A.C.Y.; Kwok, J. Diabetic Corneal Neuropathy: Pathogenic Mechanisms and Therapeutic Strategies. Front. Pharmacol. 2022, 13, 816062.
  73. Chen, X.; Graham, J.; Petropoulos, I.N.; Ponirakis, G.; Asghar, O.; Alam, U.; Marshall, A.; Ferdousi, M.; Azmi, S.; Efron, N. Corneal nerve fractal dimension: A novel corneal nerve metric for the diagnosis of diabetic sensorimotor polyneuropathy. Investig. Ophthalmol. Vis. Sci. 2018, 59, 1113–1118.
  74. Li, Q.; Zhong, Y.; Zhang, T.; Zhang, R.; Zhang, Q.; Zheng, H.; Ji, L.; Sun, W.; Zhu, X.; Zhang, S. Quantitative analysis of corneal nerve fibers in type 2 diabetics with and without diabetic peripheral neuropathy: Comparison of manual and automated assessments. Diabetes Res. Clin. Pract. 2019, 151, 33–38.
  75. Lewis, E.J.; Lovblom, L.E.; Ferdousi, M.; Halpern, E.M.; Jeziorska, M.; Pacaud, D.; Pritchard, N.; Dehghani, C.; Edwards, K.; Srinivasan, S. Rapid corneal nerve fiber loss: A marker of diabetic neuropathy onset and progression. Diabetes Care 2020, 43, 1829–1835.
  76. Sady, C.; Khosrof, S.; Nagaraj, R. Advanced Maillard reaction and crosslinking of corneal collagen in diabetes. Biochem. Biophys. Res. Commun. 1995, 214, 793–797.
  77. Mocan, M.C.; Durukan, I.; Irkec, M.; Orhan, M. Morphologic alterations of both the stromal and subbasal nerves in the corneas of patients with diabetes. Cornea 2006, 25, 769–773.
  78. Saghizadeh, M.; Brown, D.J.; Castellon, R.; Chwa, M.; Huang, G.H.; Ljubimova, J.Y.; Rosenberg, S.; Spirin, K.S.; Stolitenko, R.B.; Adachi, W. Overexpression of matrix metalloproteinase-10 and matrix metalloproteinase-3 in human diabetic corneas: A possible mechanism of basement membrane and integrin alterations. Am. J. Pathol. 2001, 158, 723–734.
  79. Gül, M.; Emre, S.; Esrefoglu, M.; Vard, N. Protective effects of melatonin and aminoguanidine on the cornea in streptozotocin-induced diabetic rats. Cornea 2008, 27, 795–801.
  80. Leppin, K.; Behrendt, A.K.; Reichard, M.; Stachs, O.; Guthoff, R.F.; Baltrusch, S.; Eule, J.C.; Vollmar, B. Diabetes mellitus leads to accumulation of dendritic cells and nerve fiber damage of the subbasal nerve plexus in the cornea. Invest. Ophthalmol. Vis. Sci. 2014, 55, 3603–3615.
  81. Sierra-Silvestre, E.; Andrade, R.J.; Holguín-Colorado, L.; Edwards, K.; Coppieters, M.W. Occurrence of corneal sub-epithelial microneuromas and axonal swelling in people with diabetes with and without (painful) diabetic neuropathy. Diabetologia 2023, 66, 1719–1734.
  82. De Cilla, S.; Ranno, S.; Carini, E.; Fogagnolo, P.; Ceresara, G.; Orzalesi, N.; Rossetti, L.M. Corneal subbasal nerves changes in patients with diabetic retinopathy: An in vivo confocal study. Investig. Ophthalmol. Vis. Sci. 2009, 50, 5155–5158.
  83. He, J.; Bazan, H.E. Mapping the nerve architecture of diabetic human corneas. Ophthalmology 2012, 119, 956–964.
  84. Davidson, E.P.; Coppey, L.J.; Kardon, R.H.; Yorek, M.A. Differences and similarities in development of corneal nerve damage and peripheral neuropathy and in diet-induced obesity and type 2 diabetic rats. Investig. Ophthalmol. Vis. Sci. 2014, 55, 1222–1230.
  85. Wang, F.; Gao, N.; Yin, J.; Fu-Shin, X.Y. Reduced innervation and delayed re-innervation after epithelial wounding in type 2 diabetic Goto-Kakizaki rats. Am. J. Pathol. 2012, 181, 2058–2066.
  86. Stem, M.S.; Hussain, M.; Lentz, S.I.; Raval, N.; Gardner, T.W.; Pop-Busui, R.; Shtein, R.M. Differential reduction in corneal nerve fiber length in patients with type 1 or type 2 diabetes mellitus. J. Diabetes Its Complicat. 2014, 28, 658–661.
  87. Zhivov, A.; Winter, K.; Hovakimyan, M.; Peschel, S.; Harder, V.; Schober, H.-C.; Kundt, G.; Baltrusch, S.; Guthoff, R.F.; Stachs, O. Imaging and quantification of subbasal nerve plexus in healthy volunteers and diabetic patients with or without retinopathy. PLoS ONE 2013, 8, e52157.
  88. Kalteniece, A.; Ferdousi, M.; Azmi, S.; Marshall, A.; Soran, H.; Malik, R.A. Keratocyte density is reduced and related to corneal nerve damage in diabetic neuropathy. Investig. Ophthalmol. Vis. Sci. 2018, 59, 3584–3590.
  89. Ferdousi, M.; Romanchuk, K.; Mah, J.K.; Virtanen, H.; Millar, C.; Malik, R.A.; Pacaud, D. Early corneal nerve fibre damage and increased Langerhans cell density in children with type 1 diabetes mellitus. Sci. Rep. 2019, 9, 8758.
  90. Qu, J.-h.; Li, L.; Tian, L.; Zhang, X.-y.; Thomas, R.; Sun, X.-G. Epithelial changes with corneal punctate epitheliopathy in type 2 diabetes mellitus and their correlation with time to healing. BMC Ophthalmol. 2018, 18, 1.
  91. Lagali, N.S.; Allgeier, S.; Guimaraes, P.; Badian, R.A.; Ruggeri, A.; Köhler, B.; Utheim, T.P.; Peebo, B.; Peterson, M.; Dahlin, L.B. Reduced corneal nerve fiber density in type 2 diabetes by wide-area mosaic analysis. Investig. Ophthalmol. Vis. Sci. 2017, 58, 6318–6327.
  92. Issar, T.; Tummanapalli, S.S.; Kwai, N.C.G.; Chiang, J.C.B.; Arnold, R.; Poynten, A.M.; Markoulli, M.; Krishnan, A.V. Associations between acute glucose control and peripheral nerve structure and function in type 1 diabetes. Diabet. Med. 2020, 37, 1553–1560.
  93. Vincent, A.M.; Edwards, J.L.; McLean, L.L.; Hong, Y.; Cerri, F.; Lopez, I.; Quattrini, A.; Feldman, E.L. Mitochondrial biogenesis and fission in axons in cell culture and animal models of diabetic neuropathy. Acta Neuropathol. 2010, 120, 477–489.
  94. Hamid, H.S.; Mervak, C.M.; Münch, A.E.; Robell, N.J.; Hayes, J.M.; Porzio, M.T.; Singleton, J.R.; Smith, A.G.; Feldman, E.L.; Lentz, S.I. Hyperglycemia-and neuropathy-induced changes in mitochondria within sensory nerves. Ann. Clin. Transl. Neurol. 2014, 1, 799–812.
  95. Vincent, A.M.; Mclean, L.L.; Backus, C.; Feldman, E.L. Short-term hyperglycemia produces oxidative damage and apoptosis in neurons. FASEB J. 2005, 19, 1–24.
  96. Ishibashi, F.; Kojima, R.; Taniguchi, M.; Kosaka, A.; Uetake, H.; Tavakoli, M. The expanded bead size of corneal C-nerve fibers visualized by corneal confocal microscopy is associated with slow conduction velocity of the peripheral nerves in patients with type 2 diabetes mellitus. J. Diabetes Res. 2016, 2016, 3653459.
  97. Zherebitskaya, E.; Akude, E.; Smith, D.R.; Fernyhough, P. Development of selective axonopathy in adult sensory neurons isolated from diabetic rats: Role of glucose-induced oxidative stress. Diabetes 2009, 58, 1356–1364.
  98. Ryle, C.; Donaghy, M. Non-enzymatic glycation of peripheral nerve proteins in human diabetics. J. Neurol. Sci. 1995, 129, 62–68.
  99. Sugimoto, K.; Yasujima, M.; Yagihashi, S. Role of advanced glycation end products in diabetic neuropathy. Curr. Pharm. Des. 2008, 14, 953–961.
  100. Sugimoto, K.; Nishizawa, Y.; Horiuchi, S.; Yagihashi, S. Localization in human diabetic peripheral nerve of Nɛ-carboxymethyllysine-protein adducts, an advanced glycation endproduct. Diabetologia 1997, 40, 1380–1387.
  101. Kaji, Y.; Amano, S.; Usui, T.; Suzuki, K.; Tanaka, S.; Oshika, T.; Nagai, R.; Horiuchi, S. Advanced glycation end products in Descemet’s membrane and their effect on corneal endothelial cell. Curr. Eye Res. 2001, 23, 469–477.
  102. Lyu, Y.; Zeng, X.; Li, F.; Zhao, S. The effect of the duration of diabetes on dry eye and corneal nerves. Contact Lens Anterior Eye 2019, 42, 380–385.
  103. Greene, D.A.; Sima, A.A.; Stevens, M.J.; Feldman, E.L.; Lattimer, S.A. Complications: Neuropathy, pathogenetic considerations. Diabetes Care 1992, 15, 1902–1925.
  104. Lehning, E.J.; LoPachin, R.M.; Mathew, J.; Eichberg, J. Changes in Na-K ATPase and protein kinase C activities in peripheral nerve of acrylamide-treated rats. J. Toxicol. Environ. Health Part. A Curr. Issues 1994, 42, 331–342.
  105. Geraldes, P.; King, G.L. Activation of protein kinase C isoforms and its impact on diabetic complications. Circ. Res. 2010, 106, 1319–1331.
  106. Rieger, G. The importance of the precorneal tear film for the quality of optical imaging. Br. J. Ophthalmol. 1992, 76, 157–158.
  107. Yang, C.-J.; Anand, A.; Huang, C.-C.; Lai, J.-Y. Unveiling the Power of Gabapentin-Loaded Nanoceria with Multiple Therapeutic Capabilities for the Treatment of Dry Eye Disease. ACS Nano 2023, 17, 25118–25135.
  108. Imam, S.; Elagin, R.B.; Jaume, J.C. Diabetes-associated dry eye syndrome in a new humanized transgenic model of type 1 diabetes. Mol. Vis. 2013, 19, 1259–1267.
  109. Zhang, X.; Zhao, L.; Deng, S.; Sun, X.; Wang, N. Dry Eye Syndrome in Patients with Diabetes Mellitus: Prevalence, Etiology, and Clinical Characteristics. J. Ophthalmol. 2016, 2016, 8201053.
  110. Research in Dry Eye: Report of the Research Subcommittee of the International Dry Eye WorkShop (2007). Ocul. Surf. 2007, 5, 179–193.
  111. Kesarwani, D.; Rizvi, S.W.A.; Khan, A.A.; Amitava, A.K.; Vasenwala, S.M.; Siddiqui, Z. Tear film and ocular surface dysfunction in diabetes mellitus in an Indian population. Indian. J. Ophthalmol. 2017, 65, 301–304.
  112. Wu, Y.-C.; Buckner, B.R.; Zhu, M.; Cavanagh, H.D.; Robertson, D.M. Elevated IGFBP3 Levels in Diabetic Tears: A Negative Regulator of IGF-1 Signaling in the Corneal Epithelium. Ocul. Surf. 2012, 10, 100–107.
  113. Zhao, Z.; Liu, J.; Shi, B.; He, S.; Yao, X.; Willcox, M.D. Advanced glycation end product (AGE) modified proteins in tears of diabetic patients. Mol. Vis. 2010, 16, 1576–1584.
  114. Liu, H.; Sheng, M.; Liu, Y.; Wang, P.; Chen, Y.; Chen, L.; Wang, W.; Li, B. Expression of SIRT1 and oxidative stress in diabetic dry eye. Int. J. Clin. Exp. Pathol. 2015, 8, 7644–7653.
  115. 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. 2021, 8, 781616.
  116. Qu, M.; Wan, L.; Dong, M.; Wang, Y.; Xie, L.; Zhou, Q. Hyperglycemia-induced severe mitochondrial bioenergetic deficit of lacrimal gland contributes to the early onset of dry eye in diabetic mice. Free Radic. Biol. Med. 2021, 166, 313–323.
  117. Richdale, K.; Chao, C.; Hamilton, M. Eye care providers’ emerging roles in early detection of diabetes and management of diabetic changes to the ocular surface: A review. BMJ Open Diabetes Res. Care 2020, 8, e001094.
More
This entry is offline, you can click here to edit this entry!
Video Production Service