Next Article in Journal
Association between the Erythrocyte Membrane Fatty Acid Profile and Cognitive Function in the Overweight and Obese Population Aged from 45 to 75 Years Old
Next Article in Special Issue
The Effect of Diet and Lifestyle on the Course of Diabetic Retinopathy—A Review of the Literature
Previous Article in Journal
Long Term Follow-Up of Sarcopenia and Malnutrition after Hospitalization for COVID-19 in Conventional or Intensive Care Units
Previous Article in Special Issue
Understanding Neurodegeneration from a Clinical and Therapeutic Perspective in Early Diabetic Retinopathy
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Nutritional and Metabolic Imbalance in Keratoconus

Cornea and Ocular Surface Disease Unit, Eye Repair Lab, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
*
Author to whom correspondence should be addressed.
Nutrients 2022, 14(4), 913; https://doi.org/10.3390/nu14040913
Submission received: 31 January 2022 / Accepted: 18 February 2022 / Published: 21 February 2022

Abstract

:
Keratoconus (KC) is a progressive corneal degeneration characterized by structural changes consisting of progressive thinning and steepening of the cornea. These alterations result in biomechanical weakening and, clinically, in vision loss. While the etiology of KC has been the object of study for over a century, no single agent has been found. Recent reviews suggest that KC is a multifactorial disease that is associated with a wide variety of genetic and environmental factors. While KC is typically considered a disease of the cornea, associations with systemic conditions have been well described over the years. In particular, nutritional and metabolic imbalance, such as the redox status, hormones, metabolites, and micronutrients (vitamins and metal ions), can deeply influence KC initiation and progression. In this paper, we comprehensively review the different nutritional (vitamins and minerals) and metabolic (hormones and metabolites) factors that are altered in KC, discussing their possible implication in the pathophysiology of the disease.

Graphical Abstract

1. Introduction

Keratoconus (KC) is the most common ectatic degeneration of the cornea. It is characterized by corneal thinning and steepening, which result in irregular astigmatism and vision loss. Initial presentation typically occurs during infancy/adolescence and usually progresses for 10 to 20 years, after which the disease is generally stable [1].The prevalence varies widely across the globe, from 0.2/100,000 in Russia to 4790/100,000 in Saudi Arabia [2], with an estimated global prevalence of 1.38/1000 [3]. Pediatric/young adult populations usually display a higher prevalence and a more aggressive form of disease, as opposed to later-onset KC [4,5,6]. Moreover, it seems that the genetic background predisposes some ethnicities to KC [7].
The pathogenesis of KC is an object of active debate. While no single etiologic factor has been identified so far, it is generally thought that KC is a multifactorial disease, which requires a genetic predisposition and an environmental factor to develop [8,9]. The role of genetic predisposition is supported by the observation that consanguinity is a risk factor for KC; a positive family history has been reported in 5–20% of KC cases [9,10]. Moreover, the prevalence of KC in first-degree relatives increases to 67% [11]. The exact nature of those genetic and environmental factors remains unknown with limited exceptions [12].
Notably, KC is generally considered an isolated ocular disorder. In fact, there is no clinical evidence of associated systemic diseases. There are, however, some exceptions; atopy [13], sleep apnea [14], mitral valve prolapse [15,16], hypothyroidism [17], and Down’s syndrome [18,19] have been detected in a percentage of KC patients varying from 21% to 70%. It is interesting to note that many of these diseases cause metabolic [20,21,22] and/or nutritional [22,23] imbalance. Therefore, it is reasonable to hypothesize that systemic nutritional and/or metabolic alterations may also play a role in KC pathogenesis. However, this is still a matter of debate, and a solid causal relationship has never been demonstrated.
Lastly, the fact that KC invariably affects both eyes, albeit to a different degree, points to a systemic etiology [24].
In this paper, we comprehensively review the different nutritional (vitamins and minerals) and metabolic (hormones and metabolites) factors that are altered in KC, discussing their possible implication in the pathophysiology of the disease.

2. Pathogenesis of Keratoconus

The structural alterations observed in KC corneas affect the epithelium, the epithelial basal membrane, the Bowman layer, and the stroma [25]. Morphological alterations of epithelial basal cells and degradation of extracellular matrix (ECM) occur [26], leading to thinning of the corneal stroma, keratocyte apoptosis, and nerve damage [27,28].
The initiation and progression of KC are deeply influenced by several nutritional and metabolic factors, such as inflammatory mediators, redox status, hormones, metabolites, and micronutrients (vitamins and metal ions) [29]. In fact, ECM remodeling is significantly influenced by these biochemical factors. For instance, metal ions such as copper and iron, and vitamins (e.g., vitamin C) are essential cofactors of enzymes involved in collagen synthesis and crosslinking [30].
In addition, chronic eye rubbing and allergies, well-known risk factors for KC [9], are associated with increased expression of metalloproteinases and proinflammatory mediators, which can favor collagen degradation and KC progression [31].

3. Nutritional and Metabolic Alterations in Keratoconus

An increasing body of evidence supports the role of different metabolic products and micronutrients in many ocular diseases, including KC [29,32,33]. In this section, we discuss the contribution of different nutrients, metabolites, and hormones in the development and progression of KC disease.

3.1. Vitamins

Vitamins are essential nutrients indispensable to many corneal functions. In recent years, the specific role of vitamin D (Vit D) in the maintenance of corneal integrity has been studied in detail [34]. Vit D maintains the corneal epithelial barrier function [35,36] and is needed for survival of endothelial cells [37]. In addition, anti-inflammatory and antimicrobial properties have been described [38]. Lastly, Vit D participates in the redox homeostasis as it has an antioxidant role [39].
Vit D is secreted into the tear fluid and the aqueous humor, from where it reaches the cornea [40,41]. In addition, the cornea possesses the molecular machinery to synthesize and activate native Vit D [36], after UV light activation, similarly to the epidermis.
The involvement of Vit D in KC pathogenesis was suggested several decades ago by Blackberg and Knapp [42]. They discovered that a Vit D-deficient diet was accompanied by ocular changes consistent with KC in preclinical animal models. Some years later, a preliminary study conducted in KC patients suggested that Vit D supplementation improves the disease [43], although interpretation of results is complicated by limited imaging techniques available at the time. The hypothesis that Vit D deficiency is common in KC patients was confirmed years later when it was shown that KC patients displayed lower serum levels of Vit D when compared to age-matched controls [44,45,46]. Interestingly, KC patients exhibit downregulation of proteins involved in Vit D transport (e.g., Vit D-binding protein) and activation [47]. Moreover, an association between Vit D deficiency and KC prevalence was suggested in people living in Middle East regions [46]. Although the serum level of Vit D was not associated with KC severity or progression rate, the probability of developing KC was significantly increased when the levels of Vit D were less than 10 ng/mL [44]. Taken together, these data suggest that Vit D plays a role in the pathogenesis of KC; however, the exact mechanism still needs further elucidation.
Vitamin C (Vit C) is a water-soluble antioxidant that is found at high concentrations in the tear film and the cornea. It is among the most potent antioxidant molecules in the human body and plays a key role in corneal wound healing [48] and ECM assembly (e.g., collagen synthesis) [49,50]. Moreover, Vit C strengthens the cornea by decreasing the distance between collagen fibrils and by enhancing crosslinking in vitro [51]. Interestingly, the levels of Vit C seem to be increased in KC corneas [52,53], suggesting an adaptive response to oxidative stress and impaired collagen synthesis. However, Vit C levels were not altered in the tears [54] or aqueous humor [52] of KC patients when compared to controls. Curiously, Vit C levels increased after corneal crosslinking, which suggests a compensatory mechanism in the wounded cornea [53].
Vitamin A (Vit A) deficiency was also suspected to contribute to KC development [55]. Recently, it has been shown that retinoic acid supplementation, an active metabolite from Vit A, promotes corneal crosslinking by upregulating transglutaminase-2 (TG2) expression in corneal epithelium and keratocyte [56].
Other vitamins, such as B12 and B9, were proposed to be involved in KC pathogenesis, since they can indirectly affect the activity of collagen synthesis enzymes through homocysteine metabolism. However, serum levels of both vitamins are unaltered [45,57].

3.2. Minerals

Nutritional deficiency can also affect the concentration of metal ions, such as copper (Cu), iron (Fe), selenium (Se), and zinc (Zn). These are all cofactors of enzymes involved in collagen synthesis, crosslinking, or antioxidant activity. Therefore, an imbalance in these minerals can be an independent risk factor for KC development [58].
In line with this hypothesis, KC patients display lower serum levels of Cu, Se, and Zn [46,59,60,61]. Moreover, Cu deficiency can be exacerbated in KC by increased tear alkalinity. This would inhibit transfer of Cu ions to the center of the cornea [62], thus generating a localized deficit of Cu. In this vein, peripheral Cu deposition is a common clinical feature in KC patients (i.e., Fleischer ring) [62,63,64]. The mechanism(s) through which deficiency of metal ions may promote KC formation are manifold. Some relevant examples are the Cu-dependent lysyl oxidase (LOX), which promotes collagen crosslinking [65], antioxidant enzymes such as Cu,Zn-Superoxide dismutase (SOD1 and SOD3) [60,61,66,67] and Se-dependent glutathione peroxidase (GPx) [66], and Zn-dependent matrix metalloproteinases (MMPs), which regulate collagen degradation [60].
Dysregulated iron metabolism has also been hypothesized in KC corneas. In fact, Fe has been detected, together with Cu, in the corneal Fleischer ring [62,63]. Due to Fe peripheral deposition, it was hypothesized that central unavailability of Fe contributes to KC development, because it works as a cofactor of lysyl hydroxylase (LH), which is required for the collagen crosslinking process. While no significant differences were found in the serum levels of Fe [59], KC patients show lower levels of iron-binding proteins serotransferrin and lactoferrin in tears [47,68] and the corneal epithelium [69], suggesting that iron homeostasis is locally altered in KC cornea. Interestingly, certain polymorphisms of the transferrin gene have been identified as risk factors for KC [70]. Deranged iron metabolism could contribute to KC development not only by reducing the function of crosslinking enzymes, but also by producing reactive oxygen species (ROS) through the Fenton reaction [71]. This may result in oxidative stress, a well-established cause of KC development.

3.3. Hormones

3.3.1. Thyroid Hormones

The contribution of thyroid hormones is still under scrutiny. Some studies have reported a higher thyroid gland dysfunction prevalence among KC patients [72,73], but others failed to prove an association between them [74,75,76]. Nevertheless, increased levels of thyroxine hormone (T4) were found in tears [72,77] and aqueous humor [78]. Moreover, the expression of thyroxine receptor (T4R) was elevated in keratocytes of KC patients compared to controls [72], suggesting an active role of T4 in the pathophysiology of KC.

3.3.2. Sex Hormones

The imbalance of sexual hormone levels has been studied in KC patients. However, data are heterogeneous, and it is still unclear when (age and disease stage) those hormones impact on KC corneas or which molecular pathways are involved [79]. A longitudinal evaluation of middle-aged KC patients failed to show any difference in progression between males and females, or in females with or without hormone treatment [80]. Interestingly, KC typically develops in puberty with stabilization after 40 years of age [9,81]. This is consistent with the profile of significant hormonal changes, supporting a potential role in KC pathophysiology. Lastly, there are reports of KC development and progression during or immediately after pregnancy [82,83].
Some studies have reported an increased expression of androgen [84,85] and estrogen [85] receptor in corneal epithelium retrieved from KC patients, while progesterone receptor showed significantly lower expression [84]. Interestingly, the levels of salivary and plasma estriol and estrone were downregulated in KC patients when compared to controls, while dehydroepiandrosterone sulfate levels were upregulated [53,86,87]. In addition, an alteration of the luteinizing hormone/follicle-stimulating hormone ratio was observed in KC patients, suggesting a role for gonadotropins in KC pathogenesis [88]. Lastly, prolactin-induced protein (PIP), secreted by a number of normal apocrine cells, has been proposed as a marker for KC progression [78,86], as it is altered in several body fluids (e.g., aqueous humor, tear fluid, plasma, saliva) of KC patients.

3.4. Metabolites

3.4.1. Redox Metabolism

The redox metabolism involves the broad range of biochemical reactions that maintain the balance between oxidant and antioxidant compounds [89]. When this balance shifts toward the pro-oxidant species, oxidative stress takes place, which may result in massive cell damage [90,91]. The eyes, particularly the cornea, are constantly exposed to environmental stressors, such as UV radiation, pollutants, and injuries. For this reason, enzymatic and nonenzymatic antioxidants are highly expressed in the human cornea [92]. In fact, alterations of the redox balance are involved in several corneal diseases, including KC [93]. The role of redox metabolism alterations in the pathogenesis of KC has been extensively studied [94]. It has been shown that KC corneas display increased production of reactive oxygen and nitrogen species (ROS and RNS, respectively) [95,96]. As a consequence, large amounts of cytotoxic byproducts from both lipid peroxidation (e.g., malondialdehyde) and nitric oxide pathways (e.g., nitrotyrosine, peroxynitrites) were observed [95,97]. In addition, mitochondrial DNA damage has been reported [98].
A possible explanation for the increased oxidative stress is a reduction in the total antioxidant capacity of the KC cornea. In line with this hypothesis, levels of non-enzymatic antioxidants (e.g., glutathione) [54,97,99] and antioxidant enzymes, including SOD and aldehyde dehydrogenase (ALDH) [94,98,100,101], are reduced in KC corneas. Another indicator of oxidative stress is the increase in lactate/pyruvate (L/P) ratio, which was found in both in vitro [102] and in vivo [99] studies.
Interestingly, redox imbalance was not restricted to the ocular surface. In fact, systemic alterations of redox homeostasis were demonstrated in KC patients. Specifically, serum ROS levels were increased, while the activity of antioxidant enzymes (SOD, GPx, catalase) and glutathione levels were reduced in KC patients [66,67,103]. These findings support the hypothesis of systemic metabolic changes that may contribute to KC initiation and progression.
In conclusion, the increased levels of ROS and RNS and the concomitant decrease of the antioxidant defenses leave the cornea more susceptible to oxidative damage and to the degradation of the extracellular matrix, which could result, clinically, in KC corneal thinning [104].

3.4.2. Arachidonic Acid Pathway

Arachidonic acid (AA) is a polyunsaturated fatty acid that is implicated in several biological functions. Due to its four cis double bonds, AA helps in maintaining cell membrane fluidity and ability to interact with proteins [105]. Moreover, AA can be metabolized by phospholipase A2s (PLA2s), cyclooxygenases (COXs), and lipoxygenases into prostaglandins and leukotrienes, important proinflammatory mediators [106]. The AA pathway is involved in different pathophysiological processes in the eye [107]. In particular, an abnormal activity of AA-related metabolic enzymes (e.g., COXs) and of its derived products has been reported in in vitro models of KC [108,109,110]. These findings were confirmed by further studies on KC patients, in which high serum levels of prostaglandin F2α, A2, and E2, as well as 5-hydroxyeicosatetraenoic acid, were reported [111]. These observations are recently leading many authors to consider KC as a systemic inflammatory disease [112].

3.4.3. Tricarboxylic Acid Cycle

The tricarboxylic acid cycle, also known as the citric acid cycle or Krebs cycle, is an amphibolic series of chemical reactions occurring in respiring organisms that eventually lead to the production of energy [113]. Defects in this cycle are associated with pathological conditions in different body sites, including the eye [114,115]. Metabolic imbalances involving the citric acid cycle have been reported in KC patients and are confirmed by detection of increased lactate production [53,87,102]. This has deep implications for the cell fate, since abundant lactate synthesis (anaerobic respiration) results in lower extracellular pH, increased oxidative stress, and apoptosis [116]. For these reasons, the lactate/malate ratio has been proposed as an indicator of oxidative stress, which may play a role in KC progression [96,102]. Interestingly, it has been shown that in vitro models of KC receiving collagen crosslinking respond with a shift toward aerobic respiration, with marked increase in ATP synthesis [53].
On the contrary, metabolomics analysis of KC patients’ tears revealed a strong increase of ATP, malate, and malonyl-CoA compared to controls [99]. This difference could be due to the contribution of other ocular surface structures (e.g., conjunctiva, lacrimal glands) to the tear composition.

3.4.4. Glycolysis and Gluconeogenesis

Glycolysis and gluconeogenesis are two metabolic pathways involved in glucose catabolism and anabolism, respectively, which play a key role in cell survival [117,118]. Studies performed on in vitro models of KC demonstrated that glucose metabolism is upregulated [119,120,121]. In particular, anaerobic glycolysis appears to be the most affected, since concentrations of lactate are higher than pyruvate [102]. Interestingly, it was shown that sex hormones are able to alter glucose metabolism in human keratoconus cells (HKCs), a finding that is corroborated by the observation that KC typically develops in puberal age [122,123]. Metabolomic analysis of tears from KC patients showed a significant increase in metabolic intermediates involved in anaerobic glycolysis, confirming the in vitro results [99].
Findings on glucose metabolism alterations led to the investigation of a possible role of diabetes mellitus (DM) in KC pathogenesis. However, results obtained from different studies were controversial, and clinical data suggest that DM is a protective rather than a risk factor for KC onset and progression [121,124].

3.4.5. Urea Cycle Metabolism

The urea cycle, also known as the ornithine cycle, is a human metabolic pathway, which serves for ammonia detoxification [125]. Stimulation of HKCs with dehydroepiandrosterone (DHEA), a sex hormone upregulated in the serum of KC patients, directly upregulated urea cycling [111]. This stimulation led to an altered availability of precursors necessary for the biosynthesis of proline and hydroxyproline, two major components of collagen [123], thereby altering collagen metabolism. Similarly, the metabolomic signature from tears of KC patients showed low levels of ornithine and an upregulation of aspartate, supporting an imbalance in the urea cycle [99].

3.4.6. Fatty Acid Metabolism

Lipids and fatty acids are two of the major components of the human cornea. They are involved in different cellular pathways, modulating pro- and anti-inflammatory reactions, promoting corneal tissue proliferation and repair, and contributing to neovascularization [126]. The most representative fatty acids in the human cornea are oleic, stearic, and palmitic acids [127]. In a recent study, gas chromatography and mass spectrometry were used to define a metabolomic signature to discriminate between healthy and KC corneas [128]. A significantly reduced amount of both saturated fatty acids, such as stearic, palmitic, myristic, and pentadecanoic acid, and unsaturated fatty acids (e.g., linoleic and trans-13-octadecenoic acid) was reported. Lower levels of fatty acids in KC patients may be explained by the reduced amount of malonyl CoA observed in previous studies [99,129]. In fact, malonyl CoA is an essential precursor for fatty acid biosynthesis [130].

4. Conclusions

The existing literature supports a role for many nutrients, hormones, and metabolites in KC pathogenesis. The proposed mechanisms of action are manifold, but generally converge toward the promotion of collagen degradation and inhibition of its synthesis/crosslinking. It remains unclear, however, if these alterations are the primary cause of KC or, rather, if they are consequent to the deeply altered KC metabolome.
It should be noted, however, that existing data support the existence of systemic, rather than eye-isolated nutritional and/or metabolic imbalance. Interestingly, some of these alterations have been associated with higher prevalence or worse prognosis. In this context, it is tempting to speculate that these alterations may translate into predictive/monitoring biochemical markers measured in the tear fluid/peripheral blood. Perhaps more importantly, the time has come to consider KC as a systemic disease, where nutritional/metabolic imbalances may play a role.

Author Contributions

R.M.L.V., F.B. and G.F. wrote the manuscript. R.M.L.V. designed, drew, and edited the figures. All authors revised the manuscript for important intellectual content. G.F. and P.R. provided the financial support. G.F. gave final approval of the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by Ospedale San Raffaele (OSR) internal funding given to G.F.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Tuft, S.J.; Moodaley, L.C.; Gregory, W.M.; Davison, C.R.; Buckley, R.J. Prognostic Factors for the Progression of Keratoconus. Ophthalmology 1994, 101, 439–447. [Google Scholar] [CrossRef]
  2. Ferrari, G.; Rama, P. The keratoconus enigma: A review with emphasis on pathogenesis. Ocul. Surf. 2020, 18, 363–373. [Google Scholar] [CrossRef]
  3. Hashemi, H.; Heydarian, S.; Hooshmand, E.; Saatchi, M.; Yekta, A.; Aghamirsalim, M.; Valadkhan, M.; Mortazavi, M.; Hashemi, A.; Khabazkhoob, M. The Prevalence and Risk Factors for Keratoconus: A Systematic Review and Meta-Analysis. Cornea 2020, 39, 263–270. [Google Scholar] [CrossRef]
  4. Anitha, V.; Vanathi, M.; Raghavan, A.; Rajaraman, R.; Ravindran, M.; Tandon, R. Pediatric keratoconus—Current perspectives and clinical challenges. Indian J. Ophthalmol. 2021, 69, 214–225. [Google Scholar] [CrossRef]
  5. Mukhtar, S.; Ambati, B.K. Pediatric keratoconus: A review of the literature. Int. Ophthalmol. 2018, 38, 2257. [Google Scholar] [CrossRef]
  6. Chan, E.; Chong, E.W.; Lingham, G.; Stevenson, L.J.; Sanfilippo, P.G.; Hewitt, A.W.; Mackey, D.A.; Yazar, S. Prevalence of Keratoconus Based on Scheimpflug Imaging: The Raine Study. Ophthalmology 2021, 128, 515–521. [Google Scholar] [CrossRef]
  7. Woodward, M.A.; Blachley, T.S.; Stein, J.D. The association between sociodemographic factors, common systemic diseases, and keratoconus an analysis of a nationwide heath care claims database. Ophthalmology 2016, 123, 457–465.e2. [Google Scholar] [CrossRef] [Green Version]
  8. Gordon-Shaag, A.; Millodot, M.; Shneor, E.; Liu, Y. The genetic and environmental factors for keratoconus. BioMed Res. Int. 2015, 2015, 795738. [Google Scholar] [CrossRef] [Green Version]
  9. Rabinowitz, Y.S. Keratoconus. Surv. Ophthalmol. 1998, 42, 297–319. [Google Scholar] [CrossRef]
  10. Lass, J.H.; Lembach, R.G.; Park, S.B.; Hom, D.L.; Fritz, M.E.; Svilar, G.M.; Nuamah, I.F.; Reinhart, W.J.; Stocker, E.G.; Keates, R.H.; et al. Clinical Management of Keratoconus: A Multicenter Analysis. Ophthalmology 1990, 97, 433–445. [Google Scholar] [CrossRef]
  11. Wang, Y.; Rabinowitz, Y.S.; Rotter, J.I.; Yang, H. Genetic epidemiological study of keratoconus: Evidence for major gene determination. Am. J. Med. Genet. 2000, 93, 403–409. [Google Scholar] [CrossRef]
  12. Hawkes, E.; Nanavaty, M.A. Eye Rubbing and Keratoconus: A Literature Review. Int. J. Keratoconus Ectatic Corneal Dis. 2014, 3, 118–121. [Google Scholar] [CrossRef]
  13. Bawazeer, A.M.; Hodge, W.G.; Lorimer, B. Atopy and keratoconus: A multivariate analysis. Br. J. Ophthalmol. 2000, 84, 834–836. [Google Scholar] [CrossRef] [Green Version]
  14. Pedrotti, E.; Demasi, C.L.; Fasolo, A.; Bonacci, E.; Brighenti, T.; Gennaro, N.; Ferrari, M.; Marchini, G. Obstructive Sleep Apnea Assessed by Overnight Polysomnography in Patients With Keratoconus. Cornea 2018, 37, 470–473. [Google Scholar] [CrossRef]
  15. Sharif, K.W.; Casey, T.A.; Coltart, J. Prevalence of mitral valve prolapse in keratoconus patients. J. R. Soc. Med. 1992, 85, 446–448. [Google Scholar] [CrossRef]
  16. Beardsley, T.L.; Foulks, G.N. An Association of Keratoconus and Mitral Valve Prolapse. Ophthalmology 1982, 89, 35–37. [Google Scholar] [CrossRef]
  17. Lee, R.; Hafezi, F.; Bradley Randleman, J. Bilateral keratoconus induced by secondary hypothyroidism after radioactive iodine therapy. J. Refract. Surg. 2018, 34, 351–353. [Google Scholar] [CrossRef]
  18. Marsack, J.D.; Benoit, J.S.; Kollbaum, P.S.; Anderson, H.A. Application of Topographical Keratoconus Detection Metrics to Eyes of Individuals with Down Syndrome. Optom. Vis. Sci. 2019, 96, 664–669. [Google Scholar] [CrossRef]
  19. Alio, J.L.; Vega-Estrada, A.; Sanz, P.; Osman, A.A.; Kamal, A.M.; Mamoon, A.; Soliman, H. Corneal morphologic characteristics in patients with down syndrome. JAMA Ophthalmol. 2018, 136, 971–978. [Google Scholar] [CrossRef] [Green Version]
  20. Ottas, A.; Fishman, D.; Okas, T.L.; Püssa, T.; Toomik, P.; Märtson, A.; Kingo, K.; Soomets, U. Blood serum metabolome of atopic dermatitis: Altered energy cycle and the markers of systemic inflammation. PLoS ONE 2017, 12, e0188580. [Google Scholar] [CrossRef] [Green Version]
  21. Shechter, A. Obstructive sleep apnea and energy balance regulation: A systematic review. Sleep Med. Rev. 2017, 34, 59–69. [Google Scholar] [CrossRef] [Green Version]
  22. Mazurek, D.; Wyka, J. Down syndrome—Genetic and nutritional aspects of accompanying disorders. Rocz. Państwowego Zakładu Hig. 2015, 66, 189–194. [Google Scholar]
  23. Penders, J.; Thijs, C.; Van Den Brandt, P.A.; Kummeling, I.; Snijders, B.; Stelma, F.; Adams, H.; Van Ree, R.; Stobberingh, E.E. Gut microbiota composition and development of atopic manifestations in infancy: The KOALA birth cohort study. Gut 2007, 56, 661–667. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Burns, D.M.; Johnston, F.M.; Frazer, D.G.; Patterson, C.; Jackson, A.J. Keratoconus: An analysis of corneal asymmetry. Br. J. Ophthalmol. 2004, 88, 1252–1255. [Google Scholar] [CrossRef]
  25. Ambekar, R.; Toussaint, K.C.; Wagoner Johnson, A. The effect of keratoconus on the structural, mechanical, and optical properties of the cornea. J. Mech. Behav. Biomed. Mater. 2011, 4, 223–236. [Google Scholar] [CrossRef]
  26. Kenney, M.C.; Nesburn, A.B.; Burgeson, R.E.; Butkowski, R.J.; Ljubimov, A. V Abnormalities of the extracellular matrix in keratoconus corneas. Cornea 1997, 16, 345–351. [Google Scholar] [CrossRef]
  27. Kaldawy, R.M.; Wagner, J.; Ching, S.; Seigel, G.M. Evidence of apoptotic cell death in keratoconus. Cornea 2002, 21, 206–209. [Google Scholar] [CrossRef]
  28. Mannion, L.S.; Tromans, C.; O’Donnell, C. An evaluation of corneal nerve morphology and function in moderate keratoconus. Contact Lens Anterior Eye 2005, 28, 185–192. [Google Scholar] [CrossRef]
  29. Shetty, R.; D’Souza, S.; Khamar, P.; Ghosh, A.; Nuijts, R.M.M.A.; Sethu, S. Biochemical markers and alterations in keratoconus. Asia-Pac. J. Ophthalmol. 2020, 9, 533–540. [Google Scholar] [CrossRef]
  30. Pinnell, S.R. Regulation of collagen biosynthesis by ascorbic acid: A review. Yale J. Biol. Med. 1985, 58, 553–559. [Google Scholar]
  31. Balasubramanian, S.A.; Pye, D.C.; Willcox, M.D.P. Effects of eye rubbing on the levels of protease, protease activity and cytokines in tears: Relevance in keratoconus. Clin. Exp. Optom. 2013, 96, 214–218. [Google Scholar] [CrossRef] [PubMed]
  32. Pellegrini, M.; Senni, C.; Bernabei, F.; Cicero, A.F.G.; Vagge, A.; Maestri, A.; Scorcia, V.; Giannaccare, G. The role of nutrition and nutritional supplements in ocular surface diseases. Nutrients 2020, 12, 952. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. 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]
  34. McMillan, J. Spectrum of Darkness, Agent of Light: Myopia, Keratoconus, Ocular Surface Disease, and Evidence for a Profoundly Vitamin D-dependent Eye. Cureus 2018, 10, e2744. [Google Scholar] [CrossRef] [Green Version]
  35. Lu, X.; Watsky, M.A. Influence of Vitamin D on corneal epithelial cell desmosomes and hemidesmosomes. Investig. Ophthalmol. Vis. Sci. 2019, 60, 4074–4083. [Google Scholar] [CrossRef] [Green Version]
  36. Yin, Z.; Pintea, V.; Lin, Y.; Hammock, B.D.; Watsky, M.A. Vitamin D enhances corneal epithelial barrier function. Investig. Ophthalmol. Vis. Sci. 2011, 52, 7359–7364. [Google Scholar] [CrossRef] [Green Version]
  37. Cankaya, C.; Cumurcu, T.; Gunduz, A. Corneal endothelial changes in patients with Vitamin D deficiency. Indian J. Ophthalmol. 2018, 66, 1256–1261. [Google Scholar] [CrossRef]
  38. Reins, R.Y.; Baidouri, H.; McDermott, A.M. Vitamin D Activation and Function in Human Corneal Epithelial Cells During TLR-Induced Inflammation. Investig. Ophthalmol. Vis. Sci. 2015, 56, 7715. [Google Scholar] [CrossRef] [Green Version]
  39. Wimalawansa, S.J. Vitamin D Deficiency: Effects on Oxidative Stress, Epigenetics, Gene Regulation, and Aging. Biology 2019, 8, 30. [Google Scholar] [CrossRef] [Green Version]
  40. Lai, Y.T.; Cerquinho, R.G.; Perez, M.M.; da Alves, B.C.A.; Pereira, E.C.; Azzalis, L.A.; Junqueira, V.B.C.; Soares, L.R.; Fonseca, F.L.A. Determination of vitamin D in tears of healthy individuals by the electrochemiluminescence method. J. Clin. Lab. Anal. 2019, 33, 22830. [Google Scholar] [CrossRef]
  41. Lee, H.K.; Jung, E.H.; Cho, B.J. Epidemiological Association between Systemic Diseases and Keratoconus in a Korean Population: A 10-Year Nationwide Cohort Study. Cornea 2020, 39, 348–353. [Google Scholar] [CrossRef] [PubMed]
  42. Blackberg, S.N.; Knapp, A.A. Ocular Changes Accompanying Disturbances of Calcium-Phosphorus Metabolism. Arch. Ophthalmol. 1934, 11, 665. [Google Scholar] [CrossRef]
  43. Knapp, A.A. Results of vitamin-D-complex treatment of keratoconus. Preliminary study. Am. J. Ophthalmol. 1939, 22, 289–292. [Google Scholar] [CrossRef]
  44. Akkaya, S.; Ulusoy, D.M. Serum Vitamin D Levels in Patients with Keratoconus. Ocul. Immunol. Inflamm. 2020, 28, 348–353. [Google Scholar] [CrossRef] [PubMed]
  45. Aslan, M.G.; Fındık, H.; Okutucu, M.; Aydın, E.; Oruç, Y.; Arpa, M.; Uzun, F. Serum 25-Hydroxy Vitamin D, Vitamin B12, and Folic Acid Levels in Progressive and Nonprogressive Keratoconus. Cornea 2021, 40, 334–341. [Google Scholar] [CrossRef]
  46. Zarei-Ghanavati, S.; Yahaghi, B.; Hassanzadeh, S.; Mobarhan, M.G.; Hakimi, H.R.; Eghbali, P. Serum 25-hydroxyvitamin D, selenium, zinc and copper in patients with keratoconus. J. Curr. Ophthalmol. 2020, 32, 26–31. [Google Scholar] [CrossRef]
  47. López-López, M.; Regueiro, U.; Bravo, S.B.; del Chantada-Vázquez, M.P.; Varela-Fernández, R.; Ávila-Gómez, P.; Hervella, P.; Lema, I. Tear proteomics in keratoconus: A quantitative SWATH-MS analysis. Investig. Ophthalmol. Vis. Sci. 2021, 62, 30. [Google Scholar] [CrossRef]
  48. Saika, S.; Uenoyama, K.; Hiroi, K.; Tanioka, H.; Takase, K.; Hikita, M. Ascorbic acid phosphate ester and wound healing in rabbit corneal alkali burns: Epithelial basement membrane and stroma. Graefe’s Arch. Clin. Exp. Ophthalmol. 1993, 231, 221–227. [Google Scholar] [CrossRef]
  49. Guo, X.; Hutcheon, A.E.K.; Melotti, S.A.; Zieske, J.D.; Trinkaus-Randall, V.; Ruberti, J.W. Morphologic characterization of organized extracellular matrix deposition by ascorbic acid-stimulated human corneal fibroblasts. Investig. Ophthalmol. Vis. Sci. 2007, 48, 4050–4060. [Google Scholar] [CrossRef]
  50. Grobe, G.M.; Reichl, S. Characterization of Vitamin C-induced cell sheets formed from primary and immortalized human corneal stromal cells for tissue engineering applications. Cells Tissues Organs 2013, 197, 283–297. [Google Scholar] [CrossRef]
  51. Aghaei, N.; Ramin, S.; Aghaei, A. In Vitro Effects of Ascorbic Acid on Corneal Collagen Cross-Linking in Keratoconus. World Fam. Med. J./Middle East J. Fam. Med. 2017, 15, 133–140. [Google Scholar] [CrossRef]
  52. Snytnikova, O.A.; Yanshole, L.V.; Iskakov, I.A.; Yanshole, V.V.; Chernykh, V.V.; Stepakov, D.A.; Novoselov, V.P.; Tsentalovich, Y.P. Quantitative metabolomic analysis of the human cornea and aqueous humor. Metabolomics 2017, 13, 152. [Google Scholar] [CrossRef]
  53. Sharif, R.; Sejersen, H.; Frank, G.; Hjortdal, J.; Karamichos, D. Effects of collagen cross-linking on the keratoconus metabolic network. Eye 2018, 32, 1271. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Saijyothi, A.V.; Fowjana, J.; Madhumathi, S.; Rajeshwari, M.; Thennarasu, M.; Prema, P.; Angayarkanni, N. Tear fluid small molecular antioxidants profiling shows lowered glutathione in keratoconus. Exp. Eye Res. 2012, 103, 41–46. [Google Scholar] [CrossRef]
  55. Mutch, J.R.; Richards, M.B. Keratoconus Experimentally Produced in The Rat by Vitamin A Deficiency. Br. J. Ophthalmol. 1939, 23, 381–387. [Google Scholar] [CrossRef] [Green Version]
  56. Wu, J.; Wang, J.; Wang, L.; Huang, Y. Topical retinoic acid induces corneal strengthening by upregulating transglutaminase 2 in murine cornea. Exp. Eye Res. 2022, 214, 108850. [Google Scholar] [CrossRef]
  57. Yilmaz, M.; Arikan, S.; Türkön, H. Plasma homocysteine levels in patients with keratoconus. Clin. Exp. Optom. 2020, 103, 804–807. [Google Scholar] [CrossRef]
  58. Dudakova, L.; Liskova, P.; Jirsova, K. Is copper imbalance an environmental factor influencing keratoconus development? Med. Hypotheses 2015, 84, 518–524. [Google Scholar] [CrossRef]
  59. Bamdad, S.; Owji, N.; Bolkheir, A. Association between advanced keratoconus and serum levels of zinc, calcium, magnesium, iron, copper, and selenium. Cornea 2018, 37, 1306–1310. [Google Scholar] [CrossRef]
  60. Ortak, H.; Söǧüt, E.; Taş, U.; Mesci, C.; Mendil, D. The relation between keratoconus and plasma levels of MMP-2, zinc, and SOD. Cornea 2012, 31, 1048–1051. [Google Scholar] [CrossRef]
  61. Kiliç, R.; Bayraktar, A.C.; Bayraktar, S.; Kurt, A.; Kavutçu, M. Evaluation of serum superoxide dismutase activity, malondialdehyde, and zinc and copper levels in patients with keratoconus. Cornea 2016, 35, 1512–1515. [Google Scholar] [CrossRef] [PubMed]
  62. Avetisov, S.E.; Mamikonian, V.R.; Novikov, I.A. The role of tear acidity and Cu-cofactor of lysyl oxidase activity in the pathogenesis of keratoconus. Vestn. Oftalmol. 2011, 127, 3–8. [Google Scholar] [PubMed]
  63. Avetisov, S.E.; Mamikonyan, V.R.; Novikov, I.A.; Pateyuk, L.S.; Osipyan, G.A.; Kiryushchenkova, N.P. Abnormal distribution of trace elements in keratoconic corneas. Ann. Ophthalmol. Vestn. Oftal’Mologii 2015, 131, 34. [Google Scholar] [CrossRef] [PubMed]
  64. Hu, P.; Lin, L.; Wu, Z.; Jin, X.; Ni, H. Kayser-Fleischer ring with keratoconus: A coincidence? A case report. BMC Ophthalmol. 2020, 20, 190. [Google Scholar] [CrossRef] [PubMed]
  65. Kagan, H.M.; Trackman, P.C. Properties and function of lysyl oxidase. Am. J. Respir. Cell Mol. Biol. 1991, 5, 206–210. [Google Scholar] [CrossRef]
  66. Cantemir, A.; Alexa, A.I.; Ciobica, A.; Balmus, I.M.; Antioch, I.; Stoica, B.; Chiselita, D.; Costin, D. Evaluation of antioxidant enzymes in keratoconus. Rev. Chim. 2016, 67, 1538–1541. [Google Scholar]
  67. Tekin, S.; Seven, E. Assessment of serum catalase, reduced glutathione, and superoxide dismutase activities and malondialdehyde levels in keratoconus patients. Eye 2021. [Google Scholar] [CrossRef]
  68. Balasubramanian, S.A.; Pye, D.C.; Willcox, M.D.P. Levels of lactoferrin, secretory IgA and serum albumin in the tear film of people with keratoconus. Exp. Eye Res. 2012, 96, 132–137. [Google Scholar] [CrossRef]
  69. Chaerkady, R.; Shao, H.; Scott, S.G.; Pandey, A.; Jun, A.S.; Chakravarti, S. The keratoconus corneal proteome: Loss of epithelial integrity and stromal degeneration. J. Proteom. 2013, 87, 122–131. [Google Scholar] [CrossRef] [Green Version]
  70. Wójcik, K.A.; Synowiec, E.; Jiménez-García, M.P.; Kaminska, A.; Polakowski, P.; Blasiak, J.; Szaflik, J.; Szaflik, J.P. Polymorphism of the transferrin gene in eye diseases: Keratoconus and Fuchs endothelial corneal dystrophy. BioMed Res. Int. 2013, 2013, 247438. [Google Scholar] [CrossRef] [Green Version]
  71. Rouault, T.A. The role of iron regulatory proteins in mammalian iron homeostasis and disease. Nat. Chem. Biol. 2006, 2, 406–414. [Google Scholar] [CrossRef] [PubMed]
  72. Thanos, S.; Oellers, P.; Meyer Zu Hörste, M.; Prokosch, V.; Schlatt, S.; Seitz, B.; Gatzioufas, Z. Role of thyroxine in the development of keratoconus. Cornea 2016, 35, 1338–1346. [Google Scholar] [CrossRef] [PubMed]
  73. El-Massry, A.; Doheim, M.F.; Iqbal, M.; Fawzy, O.; Said, O.M.; Yousif, M.O.; Badawi, A.E.; Tawfik, A.; Abousamra, A. Association between keratoconus and thyroid gland dysfunction: A cross-sectional case-control study. J. Refract. Surg. 2020, 36, 253–257. [Google Scholar] [CrossRef] [Green Version]
  74. Meyer, J.J. Thyroid Gland Dysfunction and Keratoconus. Cornea 2018, 37, e3–e4. [Google Scholar] [CrossRef]
  75. AlHawari, H.H.; Khader, Y.S.; AlHawari, H.H.; Alomari, A.F.; Abbasi, H.N.; El-Faouri, M.S.; Al Bdour, M.D. Autoimmune Thyroid Disease and Keratoconus: Is There an Association? Int. J. Endocrinol. 2018, 2018, 7907512. [Google Scholar] [CrossRef] [PubMed]
  76. Flaskó, Z.; Zemova, E.; Eppig, T.; Módis, L.; Langenbucher, A.; Wagenpfeil, S.; Seitz, B.; Szentmáry, N. Hypothyroidism is Not Associated with Keratoconus Disease: Analysis of 626 Subjects. J. Ophthalmol. 2019, 2019, 3268595. [Google Scholar] [CrossRef]
  77. Kahán, I.L.; Varsányi-Nagy, M.; Tóth, M.; Nádrai, A. The possible role of tear fluid thyroxine in keratoconus development. Exp. Eye Res. 1990, 50, 339–343. [Google Scholar] [CrossRef]
  78. Stachon, T.; Stachon, A.; Hartmann, U.; Seitz, B.; Langenbucher, A.; Szentmáry, N. Urea, Uric Acid, Prolactin and fT4 Concentrations in Aqueous Humor of Keratoconus Patients. Curr. Eye Res. 2017, 42, 842–846. [Google Scholar] [CrossRef]
  79. Karamichos, D.; Escandon, P.; Vasini, B.; Nicholas, S.E.; Van, L.; Dang, D.H.; Cunningham, R.L.; Riaz, K.M. Anterior pituitary, sex hormones, and keratoconus: Beyond traditional targets. Prog. Retin. Eye Res. 2021, 101016. [Google Scholar] [CrossRef]
  80. Fink, B.A.; Sinnott, L.T.; Wagner, H.; Friedman, C.; Zadnik, K. The influence of gender and hormone status on the severity and progression of keratoconus. Cornea 2010, 29, 65–72. [Google Scholar] [CrossRef]
  81. Kennedy, R.H.; Bourne, W.M.; Dyer, J.A. A 48-year clinical and epidemiologic study of keratoconus. Am. J. Ophthalmol. 1986, 101, 267–273. [Google Scholar] [CrossRef]
  82. Stock, R.A.; Thumé, T.; Bonamigo, E.L. Acute corneal hydrops during pregnancy with spontaneous resolution after corneal cross-linking for keratoconus: A case report. J. Med. Case Rep. 2017, 11, 53. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Bilgihan, K.; Hondur, A.; Sul, S.; Ozturk, S. Pregnancy-induced progression of keratoconus. Cornea 2011, 30, 991–994. [Google Scholar] [CrossRef]
  84. Yin, H.; Luo, C.; Tian, Y.; Deng, Y. Altered expression of sex hormone receptors in keratoconus corneas. Biomed. Res. 2017, 28, 5089–5092. [Google Scholar]
  85. Ayan, B.; Yuksel, N.; Carhan, A.; Gumuşkaya Ocal, B.; Akcay, E.; Cagil, N.; Asik, M.D. Evaluation estrogen, progesteron and androgen receptor expressions in corneal epithelium in keratoconus. Contact Lens Anterior Eye 2019, 42, 492–496. [Google Scholar] [CrossRef] [PubMed]
  86. Sharif, R.; Bak-Nielsen, S.; Sejersen, H.; Ding, K.; Hjortdal, J.; Karamichos, D. Prolactin-Induced Protein is a novel biomarker for Keratoconus. Exp. Eye Res. 2019, 179, 55–63. [Google Scholar] [CrossRef]
  87. McKay, T.B.; Hjortdal, J.; Sejersen, H.; Asara, J.M.; Wu, J.; Karamichos, D. Endocrine and Metabolic Pathways Linked to Keratoconus: Implications for the Role of Hormones in the Stromal Microenvironment. Sci. Rep. 2016, 6, 25534. [Google Scholar] [CrossRef] [Green Version]
  88. Karamichos, D.; Barrientez, B.; Nicholas, S.; Ma, S.; Van, L.; Bak-Nielsen, S.; Hjortdal, J. Gonadotropins in Keratoconus: The Unexpected Suspects. Cells 2019, 8, 1494. [Google Scholar] [CrossRef] [Green Version]
  89. Sies, H.; Jones, D. Oxidative Stress. In Encyclopedia of Stress; Academic Press: Cambridge, MA, USA, 2007; pp. 45–48. ISBN 9780123739476. [Google Scholar]
  90. 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]
  91. Sies, H. Oxidative stress: A concept in redox biology and medicine. Redox Biol. 2015, 4, 180–183. [Google Scholar] [CrossRef] [Green Version]
  92. Cabrera, M.P.; Chihuailaf, R.H. Antioxidants and the integrity of ocular tissues. Vet. Med. Int. 2011, 2011, 905153. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Cejka, C.; Cejkova, J. Oxidative stress to the cornea, changes in corneal optical properties, and advances in treatment of corneal oxidative injuries. Oxid. Med. Cell. Longev. 2015, 2015, 591530. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Gondhowiardjo, T.D.; Van Haeringen, N.J.; Volker-Dieben, H.J.; Beekhuis, H.W.; Kok, J.H.C.; Van Rij, G.; Pels, L.; Kijlstra, A. Analysis of corneal aldehyde dehydrogenase patterns in pathologic corneas. Cornea 1993, 12, 146–154. [Google Scholar] [CrossRef] [PubMed]
  95. Buddi, R.; Lin, B.; Atilano, S.R.; Zorapapel, N.C.; Kenney, M.C.; Brown, D.J. Evidence of oxidative stress in human corneal diseases. J. Histochem. Cytochem. 2002, 50, 341–351. [Google Scholar] [CrossRef] [PubMed]
  96. Chwa, M.; Atilano, S.R.; Reddy, V.; Jordan, N.; Kim, D.W.; Kenney, M.C. Increased stress-induced generation of reactive oxygen species and apoptosis in human keratoconus fibroblasts. Investig. Ophthalmol. Vis. Sci. 2006, 47, 1902–1910. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  97. Arnal, E.; Peris-Martínez, C.; Menezo, J.L.; Johnsen-Soriano, S.; Romero, F.J. Oxidative stress in keratoconus? Investig. Ophthalmol. Vis. Sci. 2011, 52, 8592–8597. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  98. Atilano, S.; Lee, D.; Fukuhara, P.; Chwa, M.; Nesburn, A.; Udar, N.; Kenney, C. Corneal Oxidative Damage in Keratoconus Cells due to Decreased Oxidant Elimination from Modified Expression Levels of SOD Enzymes, PRDX6, SCARA3, CPSF3, and FOXM1. J. Ophthalmic Vis. Res. 2019, 14, 62. [Google Scholar] [CrossRef]
  99. Karamichos, D.; Zieske, J.D.; Sejersen, H.; Sarker-Nag, A.; Asara, J.M.; Hjortdal, J. Tear metabolite changes in keratoconus. Exp. Eye Res. 2015, 132, 1–8. [Google Scholar] [CrossRef] [Green Version]
  100. Navel, V.; Malecaze, J.; Pereira, B.; Baker, J.S.; Malecaze, F.; Sapin, V.; Chiambaretta, F.; Dutheil, F. Oxidative and antioxidative stress markers in keratoconus: A systematic review and meta-analysis. Acta Ophthalmol. 2021, 99, e777–e794. [Google Scholar] [CrossRef]
  101. Behndig, A.; Karlsson, K.; Johansson, B.O.; Brännström, T.; Marklund, S.L. Superoxide dismutase isoenzymes in the normal and diseased human cornea. Investig. Ophthalmol. Vis. Sci. 2001, 42, 2293–2296. [Google Scholar]
  102. Karamichos, D.; Hutcheon, A.E.K.; Rich, C.B.; Trinkaus-Randall, V.; Asara, J.M.; Zieske, J.D. In vitro model suggests oxidative stress involved in keratoconus disease. Sci. Rep. 2014, 4, 4608. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Toprak, I.; Kucukatay, V.; Yildirim, C.; Kilic-Toprak, E.; Kilic-Erkek, O. Increased systemic oxidative stress in patients with keratoconus. Eye 2014, 28, 285–289. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Kenney, M.C.; Brown, D.J. The cascade hypothesis of keratoconus. Contact Lens Anterior Eye 2003, 26, 139–146. [Google Scholar] [CrossRef]
  105. Hanna, V.S.; Hafez, E.A.A. Synopsis of arachidonic acid metabolism: A review. J. Adv. Res. 2018, 11, 23–32. [Google Scholar] [CrossRef]
  106. Yarla, N.S.; Bishayee, A.; Sethi, G.; Reddanna, P.; Kalle, A.M.; Dhananjaya, B.L.; Dowluru, K.S.V.G.K.; Chintala, R.; Duddukuri, G.R. Targeting arachidonic acid pathway by natural products for cancer prevention and therapy. Semin. Cancer Biol. 2016, 40–41, 48–81. [Google Scholar] [CrossRef]
  107. Radi, Z.A.; Render, J.A. The pathophysiologic role of cyclo-oxygenases in the eye. J. Ocul. Pharmacol. Ther. 2008, 24, 141–151. [Google Scholar] [CrossRef]
  108. Pouliquen, Y.; Bureau, J.; Mirshahi, M.; Mirshahi, S.S.; Assouline, M.; Lorens, G. Keratoconus and inflammatory processes. Bull. Soc. Belge Ophtalmol. 1996, 262, 25–28. [Google Scholar]
  109. Bureau, J.; Fabre, E.J.; Hecquet, C.; Pouliquen, Y.; Lorans, G. Modification of prostaglandin E2 and collagen synthesis in keratoconus fibroblasts, associated with an increase of interleukin 1α receptor number. Comptes Rendus l’Academie des Sci.-Ser. III 1993, 316, 425–430. [Google Scholar] [CrossRef]
  110. Bureau, J. Synthesis of t-PA, PAI, TGF-β1 in keratoconus cells. Vis. Res. 1995, 35, S178. [Google Scholar] [CrossRef]
  111. Daphne Teh, A.L.; Jayapalan, J.J.; Loke, M.F.; Wan Abdul Kadir, A.J.; Subrayan, V. Identification of potential serum metabolic biomarkers for patient with keratoconus using untargeted metabolomics approach. Exp. Eye Res. 2021, 211, 108734. [Google Scholar] [CrossRef]
  112. Galvis, V.; Sherwin, T.; Tello, A.; Merayo, J.; Barrera, R.; Acera, A. Keratoconus: An inflammatory disorder? Eye 2015, 29, 843–859. [Google Scholar] [CrossRef] [Green Version]
  113. Engelking, L.R. Leaks in the Tricarboxylic Acid (TCA) Cycle. In Textbook of Veterinary Physiological Chemistry; Academic Press: Cambridge, MA, USA, 2015; pp. 214–218. ISBN 978-0-12-391909-0. [Google Scholar]
  114. Rowan, S.; Jiang, S.; Chang, M.L.; Szymanski, J.; Korem, T.; Segal, E.; Cassalman, C.; McGuire, C.; Baleja, J.D.; Clish, C.B.; et al. Interaction of metabolome and microbiome contributes to dietary glycemia-induced age-related macular degeneration in aged C57BL/6J mice. Investig. Ophthalmol. Vis. Sci. 2016, 57, 5002. [Google Scholar]
  115. Hartong, D.T.; Dange, M.; McGee, T.L.; Berson, E.L.; Dryja, T.P.; Colman, R.F. Insights from retinitis pigmentosa into the roles of isocitrate dehydrogenases in the Krebs cycle. Nat. Genet. 2008, 40, 1230–1234. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  116. Blokhina, O.; Virolainen, E.; Fagerstedt, K.V. Antioxidants, Oxidative Damage and Oxygen Deprivation Stress: A Review. Ann. Bot. 2003, 91, 179. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  117. Akram, M. Mini-review on Glycolysis and Cancer. J. Cancer Educ. 2013, 28, 454–457. [Google Scholar] [CrossRef]
  118. Hatting, M.; Tavares, C.D.J.; Sharabi, K.; Rines, A.K.; Puigserver, P. Insulin regulation of gluconeogenesis. Ann. N. Y. Acad. Sci. 2018, 1411, 21–35. [Google Scholar] [CrossRef]
  119. McKay, T.B.; Lyon, D.; Sarker-Nag, A.; Priyadarsini, S.; Asara, J.M.; Karamichos, D. Quercetin Attenuates Lactate Production and Extracellular Matrix Secretion in Keratoconus. Sci. Rep. 2015, 5, 9003. [Google Scholar] [CrossRef]
  120. Mckay, T.B.; Sarker-Nag, A.; Lyon, D.; Asara, J.M.; Karamichos, D. Quercetin modulates keratoconus metabolism in vitro. Cell Biochem. Funct. 2015, 33, 341–350. [Google Scholar] [CrossRef] [Green Version]
  121. Whelchel, A.E.; McKay, T.B.; Priyadarsini, S.; Rowsey, T.; Karamichos, D. Association between Diabetes and Keratoconus: A Retrospective Analysis. Sci. Rep. 2019, 9, 13808. [Google Scholar] [CrossRef] [Green Version]
  122. Gu, S.; Liu, Z.; Pan, S.; Jiang, Z.; Lu, H.; Amit, O.; Bradbury, E.M.; Hu, C.A.A.; Chen, X. Global investigation of p53-induced apoptosis through quantitative proteomic profiling using comparative amino acid-coded tagging. Mol. Cell. Proteom. 2004, 3, 998–1008. [Google Scholar] [CrossRef] [Green Version]
  123. McKay, T.B.; Hjortdal, J.; Sejersen, H.; Karamichos, D. Differential Effects of Hormones on Cellular Metabolism in Keratoconus in Vitro. Sci. Rep. 2017, 7, 42896. [Google Scholar] [CrossRef] [PubMed]
  124. McKay, T.B.; Priyadarsini, S.; Karamichos, D. Mechanisms of Collagen Crosslinking in Diabetes and Keratoconus. Cells 2019, 8, 1239. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  125. Matsumoto, S.; Häberle, J.; Kido, J.; Mitsubuchi, H.; Endo, F.; Nakamura, K. Urea cycle disorders—Update. J. Hum. Genet. 2019, 64, 833–847. [Google Scholar] [CrossRef] [PubMed]
  126. Kenchegowda, S.; Bazan, H.E.P. Significance of lipid mediators in corneal injury and repair. J. Lipid Res. 2010, 51, 879–891. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  127. Tschetter, R.T. Lipid Analysis of the Human Cornea with and without Arcus Senilis. Arch. Ophthalmol. 1966, 76, 403–405. [Google Scholar] [CrossRef]
  128. Wojakowska, A.; Pietrowska, M.; Widlak, P.; Dobrowolski, D.; Wylegała, E.; Tarnawska, D. Metabolomic signature discriminates normal human cornea from Keratoconus—A pilot GC/MS study. Molecules 2020, 25, 2933. [Google Scholar] [CrossRef]
  129. McKay, T.B.; Hjortdal, J.; Priyadarsini, S.; Karamichos, D. Acute hypoxia influences collagen and matrix metalloproteinase expression by human keratoconus cells in vitro. PLoS ONE 2017, 12, e0176017. [Google Scholar] [CrossRef] [Green Version]
  130. Fadó, R.; Rodríguez-Rodríguez, R.; Casals, N. The return of malonyl-CoA to the brain: Cognition and other stories. Prog. Lipid Res. 2021, 81, 101071. [Google Scholar] [CrossRef]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Lasagni Vitar, R.M.; Bonelli, F.; Rama, P.; Ferrari, G. Nutritional and Metabolic Imbalance in Keratoconus. Nutrients 2022, 14, 913. https://doi.org/10.3390/nu14040913

AMA Style

Lasagni Vitar RM, Bonelli F, Rama P, Ferrari G. Nutritional and Metabolic Imbalance in Keratoconus. Nutrients. 2022; 14(4):913. https://doi.org/10.3390/nu14040913

Chicago/Turabian Style

Lasagni Vitar, Romina Mayra, Filippo Bonelli, Paolo Rama, and Giulio Ferrari. 2022. "Nutritional and Metabolic Imbalance in Keratoconus" Nutrients 14, no. 4: 913. https://doi.org/10.3390/nu14040913

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