Next Article in Journal
The Relationship of the Plasma Glycated CD59 Level with Microvascular Complications in Diabetic Patients and Its Evaluation as a Predictive Marker
Previous Article in Journal
Public Beliefs and Perception of Mental Disorders in Poland—A 2025 Nationwide Cross-Sectional Survey
Previous Article in Special Issue
Advances in Intracorneal Ring Segment (ICRS) Implantation for Keratoconus: A Comprehensive Literature Review, Clinical Insights, and Future Prospects
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Keratoconus: The Local Manifestation of a Systemic Disease?

by
Matteo Pederzolli
1,2,
Federico Procopio
2,3,
Beatrice Tombolini
4,
Simone Marra
2,3,
Massimo De Micheli
5,
Francesco Bandello
2,3 and
Giulio Ferrari
1,2,3,5,*
1
Eye Repair Lab, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
2
Cornea and Ocular Surface Disease Service, Ophthalmology Unit, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
3
Department of Ophthalmology, Vita-Salute San Raffaele University, 20132 Milan, Italy
4
Ophthalmology Department, Ospedale di Circolo e Fondazione Macchi, ASST Sette Laghi, 21100 Varese, Italy
5
Ophthalmology Unit, Ospedale Maggiore di Lodi, 21100 Lodi, Italy
*
Author to whom correspondence should be addressed.
J. Clin. Med. 2025, 14(13), 4587; https://doi.org/10.3390/jcm14134587
Submission received: 30 March 2025 / Revised: 16 June 2025 / Accepted: 21 June 2025 / Published: 28 June 2025
(This article belongs to the Special Issue Keratoconus: Current Status and Prospects)

Abstract

Keratoconus (KC) is the most common ectatic corneal disease. In this review, the systemic aspects of the disease are discussed, including patient age, genetics, systemic inflammatory status and immune system dysregulation, atopy and eye rubbing, systemic metabolism, the metabolism of micronutrients (including vitamin D), and hormonal balance. The association between KC and metabolic diseases, collagen diseases, and psychiatric conditions is also considered. The evidence that is currently available strongly suggests a systemic predisposition toward KC. The understanding that KC may be the local manifestation of a systemic disease could allow earlier detection/prevention and pave the way for research into new treatments addressing the pathogenetic foundations of KC, rather than limiting intervention to the corneal disease.

1. Introduction

Keratoconus (KC) is a bilateral ectatic corneal disorder that results in progressive thinning and steepening of the cornea, irregular astigmatism, and decreased visual acuity [1,2,3].
The 2015 Global Consensus on KC and Ectatic Diseases reached an agreement on considering KC a disease of many components: a genetic disorder, a biochemical disorder, a biomechanical disorder, and an environmental disorder [2].
In the past decades, evidence has accumulated that links this corneal disease to a series of systemic associated/predisposing factors. This is leading us to reconsider our paradigm of KC as an exclusively corneal disease. Interestingly, many of the diseases associated with KC show increased levels of systemic inflammation. In this vein, while KC used to be defined as a “non-inflammatory” corneal degeneration [3,4,5,6], such a concept has been revisited, as the role of inflammation, both local and systemic, has been acknowledged in the pathogenesis of KC [7,8,9,10].
In this review, we aim to review existing evidence supporting the hypothesis that KC is the ocular manifestation of a systemic disease.

2. Genetics

A first clue about KC not being—at least exclusively—caused by strictly local factors is the identification of various genetic alterations involving oxidative stress, extracellular matrix composition, and inflammation.
First of all, the existence of a genetic component in KC is supported by observations on family history: up to 20% of patients report a positive family history [11], and first-degree relatives of patients with KC have a much higher (up to 67%) risk of having KC [12]. In addition, consanguinity has been described as a risk factor for KC [13], and monozygotic twins have a similar predisposition toward the disease [14]. Sporadic, mitochondrial, autosomal dominant, and autosomal recessive traits have been described in association with KC; non-coding genes have also been hypothesized to play a role in KC pathogenesis [15,16,17,18]. Specific genes that have been associated with KC through candidate gene analysis include Visual System Homebox 1 (VSX1), Superoxide Dismutase 1 (SOD1), Zinc Finger Protein 469 (ZNF469), and collagen genes such as COL4A1/2/3/4 and COL8A1/2 [19,20,21,22,23,24,25,26,27,28,29,30,31]. Genome-wide analysis studies have found relevant associations of KC with many genetic variants, although with varying degrees of evidence, such as IL1B, TGF1B, MIR184, DOCK9, LOX, CAST, and single nucleotide polymorphisms of HGF and RAB3GAP1 [32,33,34,35,36,37,38,39,40,41,42,43,44,45,46,47,48,49,50,51,52].
Trisomy 21 (Down syndrome) is a striking example of how systemic predisposition on a genetic basis may interact with environmental factors in promoting KC. The reported prevalence of KC in patients with Down syndrome is variable due to differences in patient age, ethnicity, sample size, diagnostic criteria, and the type of technology used for diagnosis [53,54]. In general, the corneas of patients with Down syndrome appear thinner and steeper and present more aberrations [54,55]. Recently, KC was diagnosed in 26.3–26.9% of patients with Down syndrome based on corneal tomography [55]. When the proportions with KC and suspect KC are combined, the prevalence reaches 71.3% [54]. A nationwide claims database in the USA noted that individuals with Down syndrome had a sixfold increase in odds of having KC [56].
A strong eye-rubbing habit has been related to an increased risk of developing KC in patients with Down syndrome [55,57]. However, other studies have found no association between eye-rubbing and the severity of ectatic changes in the cornea in patients with Down syndrome [58,59].
Chromosome 21 hosts both collagen VI genes (COL6A1 and A2) and SOD1; these genes have been implicated in collagen matrix alterations and increased oxidative stress in patients with Down syndrome [60,61,62,63,64].
Consequently, it is suspected that eye rubbing, collagen structure, and oxidative stress all contribute to the higher risk of KC in patients with Down syndrome, even though their relative weight cannot be determined at the moment.

3. Systemic Inflammatory Pathways and Innate Immunity

Systemic inflammation seems to play a pivotal role in the pathogenesis of KC.
From an epidemiological point of view, several systemic inflammatory diseases have been found to be associated with KC, such as rheumatoid arthritis, ulcerative colitis, autoimmune chronic active hepatitis, and irritable bowel syndrome [65,66], although evidence is not unequivocal [67].
Numerous studies have found a correlation between systemic markers of inflammation and KC.
Altered redox metabolism has been demonstrated in the cornea of patients with KC [68,69,70,71], which induces mitochondrial DNA damage [72]. It is also known that, in the cornea of patients with KC, there are reduced levels of nonenzymatic (glutathione) [71,73,74] and enzymatic antioxidants [73,75,76,77].
The increased oxidative stress in the cornea is hypothesized to lead to oxidative damage, with increased extracellular matrix degradation eventually inducing corneal thinning [78,79]. Interestingly, the oxidative metabolism of patients with KC has been found to be altered not only at the ocular surface level but also systemically, in terms of higher levels of ROS, reduced glutathione concentration, and reduced activity of antioxidant-acting enzymes [80,81,82].
The arachidonic acid pathway has also been studied in association with KC: elevated serum levels of prostaglandins (F2-alpha, A2, and E2) as well as 5-hydroxyeicosatetraenoic acid are found in patients with KC [83,84].
Other inflammatory mediators, such as IL-6, TNF-α, and MMP-9, are increased both in the tears and the serum of patients with KC [85,86,87,88,89]; elevated concentrations of interleukin-8 (IL-8) were found both in corneal cells and blood samples from patients with KC [90]; and an increased neutrophil-to-lymphocyte ratio (a marker of systemic inflammation) was found to be associated with progressive KC, once again prompting the suggestion that KC may be part of a systemic inflammatory disorder [91].
To explain such a pro-inflammatory status in patients with KC, a compelling line of research is currently focusing on innate immunity hyperactivation: toll-like receptors 2 and 4 (TLR2 and TLR4) show increased expression both in conjunctival cells and monocytes/neutrophils of patients with KC compared to healthy individuals [92,93]. Such overexpression also correlates with tear and serum levels of inflammatory mediators and parameters of KC severity and is inversely proportional to tear and serum lactoferrin levels [94].

4. Atopy and Eye Rubbing

On the other hand, the association between KC and atopy, a condition primarily characterized by a hyperactivation of acquired (rather than innate) immunity, has been studied extensively and is consisently corroborated by additional evidence [95,96,97]. A recent meta-analysis, for example, showed that patients with allergic rhinitis are more likely to develop KC [98]. Moreover, a causal role of atopic dermatitis in the development of KC has been reported [99]. Most importantly, allergic conjunctivitis is often found in patients with KC [100,101,102,103]; this points to the usefulness of screening for KC during the management of patients with allergic conjunctivitis. A comparison of the proteomic profile in the tears of patients with KC and allergic conjunctivitis pointed to an association between the two conditions at a molecular level [104].
The comorbidity of KC and atopy is further demonstrated by the association between vernal keratoconjunctivitis and KC [102,105]. A significant correlation has been shown between the severity of KC and vernal keratoconjunctivitis [102]. On the other hand, a study conducted in Germany recently failed to show an association between KC and atopy [106].
It is important to note that a state of chronic allergic reaction of the ocular surface—and thus ocular itching—may lead to eye rubbing. The association between eye rubbing and KC emerges clearly in almost all studies carried out on a wide variety of populations [107,108,109]. When tightly controlled, a significant proportion of patients show no disease progression at a three-year follow-up following cessation of eye rubbing, without any need for further intervention [110]. It should also be noted that eye rubbing sometimes represents an addictive behavior in patients with KC [111]. A Chinese study described gene–environment interactions between single nucleotide polymorphisms (rs26515, rs27991, and rs9314177) in the CAST gene (which has been associated with KC) and eye rubbing [35,112].
The exact mechanism by which eye rubbing leads to the development or progression of KC has not been entirely elucidated. It was thought that the cornea of these patients is structurally weaker than normal and rubbing easily promotes its deformation [113]. A recent study showed a strong association between KC, eye rubbing, and an elevated concentration of Langerhans cells in the central cornea [114]. Despite the evidence, KC patients seem to show little awareness of the importance of avoiding this conduct [115]. It is our opinion that the first approach to KC must be thorough patient information and the reduction of trigger stimuli at a local and potentially systemic level (notably, by treating atopic conditions).

5. Vitamin D: A Modulator of Systemic Inflammation

Vitamin D (Vit D) is a pleiotropic fat-soluble prohormone. Perhaps its most commonly known function is immune system modulation: Vit D can act on both innate and adaptive immune responses [116].
As discussed so far, both types of immune response have been implicated in KC pathogenesis, which makes Vit D a factor of high interest.
Furthermore, it has been shown that Vit D plays a central role in corneal homeostasis [117,118,119,120,121,122]. Briefly, Vit D supports epithelial barrier function and endothelial cell survival [117,118,119,120], and it modulates inflammation [121,122] by reducing inflammatory mediators (cytokines, prostaglandins) [123]. Vitamin D can enter the cornea either via tear fluid and aqueous humor [67,124], or it can be synthesized de novo in limbal epithelium after UVB radiation exposure [119,125].
Accumulating evidence supports the hypothesis that Vit D may participate in KC pathogenesis [3,11,126,127,128,129,130,131,132,133,134,135,136,137,138,139,140,141,142]. KC may coexist with conditions associated with Vit D deficiency, including systemic affections (i.e., atopy, asthma, obstructive sleep apnea, Down syndrome, and heart valve and thyroid diseases) [130,131,132,133,134], adolescence, and pregnancy [3,11,135,136,137,138,139]. A 1938 preclinical study on animal models (which had many limitations in light of current knowledge) showed that a Vit D-deficient diet may lead to KC-like ocular alterations [143]. A subsequent 1939 pilot study found that vitamin D supplementation could arrest and even reverse KC [126]. More recently, patients affected by KC were found to have lower serum Vit D levels (<10 ng/mL) [127,128,129]; interestingly, the same has occasionally been reported for patients with myopia [117,144]. Furthermore, Vit D levels have been found to be lower in progressive rather than nonprogressive KC, although there is no unanimous consensus on this point [141].
Our group evaluated the clinical and biological impact of the administration of Vit D in patients with KC and Vit D insufficiency (<30 ng/mL). The first version of the study [145] included twenty adolescent patients who were supplemented for 6 months and followed up for 12 months. Interestingly, 60% of patients (72% of eyes) showed topographic stabilization of KC development (Kmax progression < 1D) after 12 months. Overall, TCT, Kmax, and BSCVA rates remained constant over the observation period. This study confirmed that patients with KC show downregulated vitamin D transport and activation [141]; since Vit D upregulates its own metabolism, supplementation increased both Vit D transport (by VDBP) and Vit D activation (by CYP27A1 and MMP-9/TIMP-1) [145]. Systemic collagen degradation was also found to decrease in the study population.
In 2025, we updated our findings with data obtained by including 20 additional patients [146]. The mitigating effect of Vit D on KC progression and systemic collagen turnover was confirmed in this 40-patient study. Additionally, RNA sequencing on peripheral blood monocytic cells obtained at baseline and after Vit D supplementation revealed a modulation in systemic inflammation and changes in platelet count and/or activity. Notably, RNA transcription patterns were found to be relatively coherent among patients showing KC stabilization and incoherent among non-responders, which points to different systemic predispositions towards Vit D response.
Overall, such data suggest that vitamin D supplementation could be a useful intervention in KC. These preliminary findings will require further confirmation by randomized clinical trials.

6. Other Micronutrients with a Possible Role in KC

Vitamin A (Vit A), whose deficiency is associated with pathologies such as keratomalacia and xerophthalmia, has been found to play a role in the development of KC. A 1939 study supplemented Vit A to Vit A-deprived rats; after a few days, the development of KC was observed [147]. It has also been observed that retinoic acid supplementation promotes corneal cross-linking (CXL) by increasing the expression of transglutaminase 2 (TG-2) in the corneal epithelium [148].
In an integrated analysis conducted on mouse models, it was observed that a significantly higher percentage of mice with KC had Vit A deficiency (VAD) than controls, and among KC mice, VAD was significantly higher in females [149]. A transcriptomic analysis found that genes that were more upregulated in female mice with VAD compared to female controls were mainly related to the immune response and were involved in the JAK-STAT pathway. In contrast, the differentially expressed genes between VAD and control male mice pertained to the estrogen signaling pathway [149].
Patients with KC have lower blood levels of riboflavin (B2) than healthy controls; this renders riboflavin a candidate risk factor for KC development. Blood levels of homocysteine, folic acid, and vitamin B12 have also been studied but showed no significant differences between patients with KC and the control group [150].
Serum levels of copper (CU), selenium (Se), and zinc (Zn) are lower in patients with KC compared to the general population [130,151,152]. In particular, copper (Cu) participates in collagen CXL and is known for its antioxidant activity [80,81,152,153,154]. Patients affected by KC display an altered copper metabolism [130,142,145,151,155], presenting reduced systemic levels of copper relative to healthy individuals [130,151]. Moreover, decreased copper levels have been detected in the central cornea of these patients [81,154,155,156]. Our group found enhanced copper availability after Vit D supplementation and hypothesized that copper metabolism alterations may contribute to KC progression despite normal Vit D levels [145].
Iron is also suspected to play a role in the pathogenesis of KC. High levels of iron are found (together with copper) in Fleischer’s corneal ring [155,156], even though no significant differences in serum iron levels have been observed in patients with KC [151]. Patients with KC exhibit reduced levels of iron-binding proteins, such as serotransferrin and lactoferrin, in both their tears and corneal epithelium, indicating local disruption in iron homeostasis within the KC cornea [141,157,158]. Polymorphisms of the transferrin gene have been identified as risk factors for KC. Altered iron metabolism may increase KC risk not only because it reduces the function of CXL enzymes but also because it generates reactive oxygen species through the Fenton reaction, leading to oxidative stress, which is known to be a risk factor for the development of KC [159,160]. Ferroptosis has also been implicated in KC, with recent studies identifying six ferroptosis-related genes differentially expressed in patients with KC using mathematical algorithms [161].

7. The Ocular Surface Microbiota

The gut–eye axis is an emerging concept that suggests that the gut microbiome can influence systemic inflammation and potentially affect the eyes [162]. While there is a lack of specific studies investigating intestinal microbiota alterations in patients with KC, a growing body of research is exploring the role of the ocular surface microbiota in this condition [162,163,164].
This initial body of evidence is mainly focused on the relative prevalence of different classes of bacteria in keratoconic and healthy corneas, but it remains ambiguous [165].

8. Does Systemic Metabolism Impact KC?

The Krebs cycle, or the tricarboxylic acid cycle, is believed to be involved in the development of KC, as increased lactate production has been found in KC both locally and systemically [166,167,168]. Abundant lactate production implies anaerobic respiration, which can then result in a reduction in intracellular pH, oxidative stress, and apoptosis [169].
In vitro models of KC found glucose metabolism to be upregulated, a finding that was later confirmed by an analysis carried out on the tears of patients with KC; anaerobic glycolysis appears to be the most affected pathway, as lactate concentrations higher than those of pyruvate are found [170,171]. In addition, it is well known that sex hormones can alter glucose metabolism (see later), and KC develops right at pubertal age [172,173].
Following in vitro CXL, there is a shift to aerobic respiration, with a significant increase in ATP production [166]. The lactate-to-malate ratio has also been proposed as an indicator of oxidative stress [71,168]. However, it is surprising to note that in the tears of patients with KC, there is increased production of ATP, malate, and malonyl-CoA [74].
Urea cycle metabolism has also been studied in relation to KC. Stimulation of the cells of patients with KC with dehydroepiandrosterone (DHEA), which is increased in the circulation of patients with KC, increases the urea cycle by altering the bioavailability of precursors necessary for collagen synthesis such as proline and hydroxyproline [84,173,174]. This finding is supported by the fact that reduced levels of ornithine and increased levels of aspartate have been found in the tear film of patients with KC [74].
Regarding acid metabolism, one study showed a significant reduction in both saturated and unsaturated fats in KC corneas [175]. This may be explained by the fact that patients with KC have reduced levels of malonyl CoA, a key precursor for fatty acid synthesis [74,176].
Docosahexaenoic acid (DHA) administration was found to significantly improve antioxidant capacity and reduce the expression of inflammatory cytokines. Such evidence, obtained through a preliminary study, corroborates the possible utility of DHA administration to target KC [177].

9. Association with Metabolic Disorders

Higher body mass index values are consistently associated with KC, suggesting that obesity is a risk factor for KC development [178,179,180]. Increased body mass index also correlates with an increased frequency of eye rubbing, which increases the risk of KC manifestation or progression [181]. KC is frequently noted in individuals with obstructive sleep apnea (OSA, a condition associated with obesity), possibly due to floppy eyelid syndrome, which is also strongly associated with KC [182,183]. A meta-analysis found that KC was associated with a 1.87 times higher risk of OSA compared to controls [184]. Furthermore, there is a significant increase in corneal steepening among patients diagnosed with severe OSA [185].
Diabetes could potentially serve as a protective factor in KC [186], as it has been proposed that it facilitates collagen CXL in the cornea [187]. Nonetheless, further studies indicated no significant correlation between KC risk and diabetes [188,189,190]. However, it was noticed that individuals with diabetes exhibited reduced odds of advancing their KC to a more severe stage [191].

10. Hormonal Balance and KC Risk

The role of thyroid hormones in the development of KC is still being investigated. A recent analysis demonstrated a higher prevalence of KC in patients with thyroid dysfunction [192], particularly among females [193]. Additionally, such patients showed increased maximum simulated keratometry value and thinner corneas [193]. This association has been confirmed by different authors, who have found a higher incidence of KC in the female sex and in association with hyperthyroidism [192,194]. On the contrary, no association was found between congenital hypothyroidism diagnosed and treated from newborn age and KC [195], and the severity of KC does not seem to change depending on the presence of hypothyroidism [196].
From a molecular perspective, elevated thyroxine values were described in the tears of patients with KC [197,198] and in the aqueous humor [199]. T4 receptor expression was also elevated in keratinocytes of patients with KC compared to controls, suggesting an active role of T4 in the development of KC [198]. In vitro, thyroxine was found to increase collagen I expression in fibroblasts of patients with KC but not in keratinocytes, while it had no effect on the production of transforming growth factor β1 (TGF-β1) or collagen V in either cell population, thus making it difficult to think that increased thyroxine concentration alone could play a role in the development of KC [200].
It should be noted that Hashimoto thyroiditis is an autoimmune disease. Therefore, regardless of the changes in thyroid hormone imbalance, it is possible that increased KC prevalence is a consequence of the dysregulated systemic immune response. In this sense, a statistically significant association of KC with Hashimoto’s thyroiditis, specifically, has also been described [201].
Data on the influence of sex hormones in KC are heterogeneous, and it is not yet certain how much impact they have on KC and which molecular pathways are involved [202]. Gender prevalence in KC is uncertain and may vary depending on geographical location [56,203,204,205]. A potential role of sex hormones in KC development has been postulated from the observation that KC typically develops during puberty, with stabilization around 40 years of age, when relevant hormonal changes occur [11,206]. Estrogen and androgen receptor expression is higher in keratinocytes of patients with KC compared with controls, while the level of progesterone receptor expression is lower [207,208]. Moreover, salivary and blood levels of estriol and estrone were found to be reduced in patients with KC, while dehydroepiandrosterone sulfate levels were increased [166,167,209]. Another study found reduced testosterone levels in the plasma of both male and female patients with KC [210]. A role for gonadotropins is also possible, as an altered luteinizing hormone/follicle-stimulating hormone ratio has been observed in patients with KC [211].
In women with KC, serum prolactin levels are found to be elevated [212]. A role of prolactin-induced protein as a marker of KC progression has also been proposed, as its levels are found to be altered in the tears, aqueous humor, plasma, and saliva of patients with KC [199,209].
Cases of development or progression of KC during or immediately after pregnancy have also been described [136,137]. Finally, it is interesting to highlight the case of KC progression following the administration of gender-affirming hormone therapy in a male-to-female transgender patient [213].
In conclusion, although the hypotheses revolving around a hormonal involvement in the pathogenesis of KC are intriguing, currently available data show an inconsistent level of evidence.

11. Association with Collagen Diseases

KC has been related to several congenital connective tissue diseases, notably mitral valve prolapse (MVP) and Ehlers–Danlos syndrome (EDS). Early studies observed the association between MVP and KC [214]. Further research revealed that 38–58% of individuals suffering from KC also exhibited MPV [214,215]. However, in later studies, much lower—but still notable—proportions of 11–13.4% of patients with MVP were found to have KC [216,217]. Interestingly, a study involving individuals with severe KC who also had corneal hydrops found that two-thirds of the patients had MVP [218]. A retrospective nationwide matched cohort study in Taiwan observed that patients over 40 years old with KC exhibited an almost twofold MPV risk compared to age-matched individuals [219].
Ehlers–Danlos syndrome (EDS) is characterized by skin hyperextensibility, joint hypermobility, and tissue fragility [220]. Metacarpophalangeal and wrist joint hypermobility is five times higher in patients with KC, emphasizing the potential risk of the disease in those with EDS [221]. Genetic variations were identified among several genes shared between the two disorders, such as COL5A1, TNXB, ZNF469, and COL12A1 [222].
Marfan syndrome is frequently mentioned as related to KC, but a large review of affected individuals did not confirm this finding [223].

12. Possible Association with Psychiatric Conditions

KC has been occasionally linked to Tourette’s disease, a psychiatric disorder that involves severe obsessive-compulsive behaviors and involuntary eye rubbing [224,225]. An association between ADHD and KC has also been described, although the severity of ADHD did not correlate with the severity of KC [226]. The correlation between KC and depression is debated [56,66,227,228,229].
In general, data on the relationship between KC and psychiatric conditions are limited, and, although compelling, these associations largely need to be proven by stronger evidence.

13. Conclusions

In conclusion, although many of the presented lines of study need to be consolidated, growing evidence seems to converge toward a systemic basis for KC (Figure 1). Based on the reviewed data, we suggest that the time is coming for a paradigm shift in the definition of KC. We propose that KC should be considered the corneal manifestation of a generalized disease or the final common corneal phenotype where constitutional (and, possibly, genetic) predisposition plays a relevant role, possibly exacerbated by local factors. In this vein, we could look at KC in the same way as we look at pseudo-exfoliation syndrome, which is mainly known to cause ocular complications but is associated with a generalized impairment of the extracellular matrix [230].
There are practical implications of this new definition of KC. First, patients with KC should be routinely investigated to search for systemic comorbidities, and second, efforts should be made to test treatments aimed at controlling inflammation and extracellular matrix impairment.
Further high-quality data are needed to confirm our interpretation.

Author Contributions

Conceptualization, G.F., M.P., M.D.M., and F.B.; methodology, G.F., M.P., M.D.M., and F.B.; data curation, G.F., M.P., F.P., B.T., and S.M.; writing—original draft preparation, G.F., M.P., F.P., B.T., and S.M.; writing—review and editing, G.F. and M.P.; supervision, G.F., M.D.M., and F.B. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Alió, J.L. (Ed.) Keratoconus; Springer International Publishing: Cham, Switzerland, 2017. [Google Scholar] [CrossRef]
  2. Gomes, J.A.P.; Tan, D.; Rapuano, C.J.; Belin, M.W.; Ambrósio, R.; Guell, J.L.; Malecaze, F.; Nishida, K.; Sangwan, V.; the Group of Panelists for the Global Delphi Panel of Keratoconus and Ectatic Diseases. Global consensus on keratoconus and ectatic diseases. Cornea 2015, 34, 359–369. [Google Scholar] [CrossRef] [PubMed]
  3. Santodomingo-Rubido, J.; Carracedo, G.; Suzaki, A.; Villa-Collar, C.; Vincent, S.J.; Wolffsohn, J.S. Keratoconus: An updated review. Cont. Lens Anterior Eye 2022, 45, 101559. [Google Scholar] [CrossRef]
  4. Smolin, G.; Thoft, R.; Foster, C.; Azar, D.; Dohlman, C. Smolin and Thoft’s the Cornea: Scientific Foundations and Clinical Practice; Lippincott Williams & Wilkins: Philadelphia, PA, USA, 2005. [Google Scholar]
  5. Krachmer, J.H.; Feder, R.S.; Belin, M.W. Keratoconus and related noninflammatory corneal thinning disorders. Surv. Ophthalmol. 1984, 28, 293–322. [Google Scholar] [CrossRef] [PubMed]
  6. Mannis, M.J.; Holland, E.J. Cornea: Fundamentals, diagnosis, and management. Am. J. Ophthalmol. 2022, 140, 779. [Google Scholar]
  7. Loh, I.P.; Sherwin, T. Is Keratoconus an Inflammatory Disease? The Implication of Inflammatory Pathways. Ocul. Immunol. Inflamm. 2022, 30, 246–255. [Google Scholar] [CrossRef]
  8. Bykhovskaya, Y.; Rabinowitz, Y.S. Update on the genetics of keratoconus. Exp. Eye Res. 2021, 202, 108398. [Google Scholar] [CrossRef]
  9. Rabinowitz, Y.S.; Galvis, V.; Tello, A.; Rueda, D.; García, J.D. Genetics vs chronic corneal mechanical trauma in the etiology of keratoconus. Exp. Eye Res. 2021, 202, 108328. [Google Scholar] [CrossRef]
  10. Ferrari, G.; Rama, P. The keratoconus enigma: A review with emphasis on pathogenesis. Ocul. Surf. 2020, 18, 363–373. [Google Scholar] [CrossRef]
  11. 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]
  12. Maher, E.R.; Yates, J.R.W.; Ferguson-Smith, M.A. Statistical analysis of the two stage mutation model in von Hippel-Lindau disease, and in sporadic cerebellar haemangioblastoma and renal cell carcinoma. J. Med. Genet. 1990, 27, 311–314. [Google Scholar] [CrossRef]
  13. Gordon-Shaag, A.; Millodot, M.; Essa, M.; Garth, J.; Ghara, M.; Shneor, E. Is consanguinity a risk factor for keratoconus? Optom. Vis. Sci. 2013, 90, 448–454. [Google Scholar] [CrossRef] [PubMed]
  14. Tuft, S.J.; Hassan, H.; George, S.; Frazer, D.G.; Willoughby, C.E.; Liskova, P. Keratoconus in 18 pairs of twins. Acta Ophthalmol. 2012, 90, e482–e486. [Google Scholar] [CrossRef] [PubMed]
  15. Valgaeren, H.; Koppen, C.; Van Camp, G. A new perspective on the genetics of keratoconus: Why have we not been more successful? Ophthalmic Genet. 2018, 39, 158–174. [Google Scholar] [CrossRef]
  16. Brancati, F.; Valente, E.M.; Sarkozy, A.; Fehèr, J.; Castori, M.; Del Duca, P.; Mingarelli, R.; Pizzuti, A.; Dallapiccola, B. A locus for autosomal dominant keratoconus maps to human chromosome 3p14-q13. J. Med. Genet. 2004, 41, 188–192. [Google Scholar] [CrossRef]
  17. Burdon, K.P.; Coster, D.J.; Charlesworth, J.C.; Mills, R.A.; Laurie, K.J.; Giunta, C.; Hewitt, A.W.; Latimer, P.; Craig, J.E. Apparent autosomal dominant keratoconus in a large Australian pedigree accounted for by digenic inheritance of two novel loci. Hum. Genet. 2008, 124, 379–386. [Google Scholar] [CrossRef]
  18. Jacobs, D.S.; Dohlman, C.H. Is keratoconus genetic? Int. Ophthalmol. Clin. 1993, 33, 249–260. [Google Scholar] [CrossRef]
  19. De Bonis, P.; Laborante, A.; Pizzicoli, C.; Stallone, R.; Barbano, R.; Longo, C.; Mazzilli, E.; Zelante, L.; Bisceglia, L. Mutational screening of VSX1, SPARC, SOD1, LOX, and TIMP3 in keratoconus. Mol. Vis. 2011, 17, 2482–2494. [Google Scholar]
  20. Saee-Rad, S.; Hashemi, H.; Miraftab, M.; Noori-Daloii, M.R.; Chaleshtori, M.H.; Raoofian, R.; Jafari, F.; Greene, W.; Fakhraie, G.; Rezvan, F.; et al. Mutation analysis of VSX1 and SOD1 in Iranian patients with keratoconus. Mol. Vis. 2011, 17, 3128–3136. [Google Scholar]
  21. Vincent, A.L.; Jordan, C.A.; Cadzow, M.J.; Merriman, T.R.; McGhee, C.N. Mutations in the zinc finger protein gene, ZNF469, contribute to the pathogenesis of keratoconus. Investig. Ophthalmol. Vis. Sci. 2014, 55, 5629–5635. [Google Scholar] [CrossRef]
  22. Lechner, J.; Porter, L.F.; Rice, A.; Vitart, V.; Armstrong, D.J.; Schorderet, D.F.; Munier, F.L.; Wright, A.F.; Inglehearn, C.F.; Black, G.C.; et al. Enrichment of pathogenic alleles in the brittle cornea gene, ZNF469, in keratoconus. Hum. Mol. Genet. 2014, 23, 5527–5535. [Google Scholar] [CrossRef]
  23. Karolak, J.A.; Gambin, T.; Rydzanicz, M.; Szaflik, J.P.; Polakowski, P.; Frajdenberg, A.; Mrugacz, M.; Podfigurna-Musielak, M.; Stankiewicz, P.; Gajecka, M. Evidence against ZNF469 being causative for keratoconus in Polish patients. Acta Ophthalmol. 2016, 94, 289–294. [Google Scholar] [CrossRef] [PubMed]
  24. Tang, Y.G.; Picornell, Y.; Su, X.; Li, X.; Yang, H.; Rabinowitz, Y.S. Three VSX1 gene mutations, L159M, R166W, and H244R, are not associated with keratoconus. Cornea 2008, 27, 189–192. [Google Scholar] [CrossRef] [PubMed]
  25. Barbaro, V.; Di Iorio, E.; Ferrari, S.; Bisceglia, L.; Ruzza, A.; De Luca, M.; Pellegrini, G. Expression of VSX1 in human corneal keratocytes during differentiation into myofibroblasts in response to wound healing. Investig. Ophthalmol. Vis. Sci. 2006, 47, 5243–5250. [Google Scholar] [CrossRef]
  26. Aldave, A.J.; Yellore, V.S.; Salem, A.K.; Yoo, G.L.; Rayner, S.A.; Yang, H.; Tang, G.Y.; Piconell, Y.; Rabinowitz, Y.S. No VSX1 gene mutations associated with keratoconus. Investig. Ophthalmol. Vis. Sci. 2006, 47, 2820–2822. [Google Scholar] [CrossRef]
  27. Štabuc-Šilih, M.; Stražišar, M.; Hawlina, M.; Glavač, D. Absence of pathogenic mutations in VSX1 and SOD1 genes in patients with keratoconus. Cornea 2010, 29, 172–176. [Google Scholar] [CrossRef]
  28. Liskova, P.; Ebenezer, N.D.; Hysi, P.G.; Gwilliam, R.; El-Ashry, M.F.; Moodaley, L.C.; . Hau, S.; Twa, M.; Tuft, S.J.; Bhatacharya, S.S. Molecular analysis of the VSX1 gene in familial keratoconus. Mol. Vis. 2007, 13, 1887–1891. [Google Scholar]
  29. Stabuc-Silih, M.; Ravnik-Glavac, M.; Glavac, D.; Hawlina, M.; Strazisar, M. Polymorphisms in COL4A3 and COL4A4 genes associated with keratoconus. Mol. Vis. 2009, 15, 2848–2860. [Google Scholar]
  30. Kokolakis, N.S.; Gazouli, M.; Chatziralli, I.P.; Koutsandrea, C.; Gatzioufas, Z.; Peponis, V.G.; Droutsas, K.D.; Kalogeropoulos, C.; Anagnou, N.; Miltsakakis, D.; et al. Polymorphism analysis of COL4A3 and COL4A4 genes in Greek patients with keratoconus. Ophthalmic Genet. 2014, 35, 226–228. [Google Scholar] [CrossRef]
  31. Lu, Y.; Vitart, V.; Burdon, K.P.; Khor, C.C.; Bykhovskaya, Y.; Mirshahi, A.; Hewitt, A.W.; Koehn, D.; Hysi, P.G.; Ramdas, W.D.; et al. Genome-wide association analyses identify multiple loci associated with central corneal thickness and keratoconus. Nat. Genet. 2013, 45, 155–163. [Google Scholar] [CrossRef]
  32. Hughes, A.E.; Bradley, D.T.; Campbell, M.; Lechner, J.; Dash, D.P.; Simpson, D.A.; Willoughby, C.E. Mutation altering the miR-184 seed region causes familial keratoconus with cataract. Am. J. Hum. Genet. 2011, 89, 628–633. [Google Scholar] [CrossRef]
  33. Iliff, B.W.; Amer Riazuddin, S.; Gottsch, J.D. A single-base substitution in the seed region of miR-184 causes EDICT syndrome. Investig. Ophthalmol. Vis. Sci. 2012, 53, 348–353. [Google Scholar] [CrossRef] [PubMed]
  34. Yanovitch, T.; Li, Y.J.; Metlapally, R.; Abbott, D.; Viet, K.N.T.; Young, T.L. Hepatocyte growth factor and myopia: Genetic association analyses in a Caucasian population. Mol. Vis. 2009, 15, 1028–1035. [Google Scholar] [PubMed]
  35. Li, X.; Bykhovskaya, Y.; Haritunians, T.; Siscovick, D.; Aldave, A.; Szczotka-flynn, L.; Iyengar, S.K.; Rotter, J.I.; Taylor, K.D.; Rabinowitz, Y.S. A genome-wide association study identifies a potential novel gene locus for keratoconus, one of the commonest causes for corneal transplantation in developed countries. Hum. Mol. Genet. 2012, 21, 421–429. [Google Scholar] [CrossRef]
  36. Wang, Y.; Wei, W.; Zhang, C.; Zhang, X.H.; Liu, M.; Zhu, X.; Xu, K. Association of Interleukin-1 Gene Single Nucleotide Polymorphisms with Keratoconus in Chinese Han Population. Curr. Eye Res. 2016, 41, 630–635. [Google Scholar] [CrossRef]
  37. Mikami, T.; Meguro, A.; Teshigawara, T.; Takeuchi, M.; Uemoto, R.; Kawagoe, T.; Nomura, E.; Asukata, Y.; Ishioka, M.; Iwasaki, M.; et al. Interleukin 1 beta promoter polymorphism is associated with keratoconus in a Japanese population. Mol. Vis. 2013, 19, 845–851. [Google Scholar]
  38. Kim, S.H.; Mok, J.W.; Kim, H.S.; Joo, C.K. Association of -31T>C and -511 C>T polymorphisms in the interleukin 1 beta (IL1B) promoter in Korean keratoconus patients. Mol. Vis. 2008, 14, 2109–2116. [Google Scholar]
  39. Palamar, M.; Onay, H.; Ozdemir, T.R.; Arslan, E.; Egrilmez, S.; Ozkinay, F.; Yagci, A. Relationship between IL1β-511C>T and ILRN VNTR polymorphisms and keratoconus. Cornea 2014, 33, 145–147. [Google Scholar] [CrossRef]
  40. Kannabiran, C. Genetics of corneal endothelial dystrophies. J. Genet. 2009, 88, 487–494. [Google Scholar] [CrossRef]
  41. Guan, T.; Liu, C.; Ma, Z.; Ding, S. The point mutation and polymorphism in keratoconus candidate gene TGFBI in Chinese population. Gene 2012, 503, 137–139. [Google Scholar] [CrossRef]
  42. Karolak, J.A.; Polakowski, P.; Szaflik, J.; Szaflik, J.P.; Gajecka, M. Molecular screening of keratoconus susceptibility sequence variants in VSX1, TGFBI, DOCK9, STK24, and IPO5 genes in Polish patients and novel TGFBI variant identification. Ophthalmic Genet. 2016, 37, 37–43. [Google Scholar] [CrossRef]
  43. Udar, N.; Kenney, M.C.; Chalukya, M.; Anderson, T.; Morales, L.; Brown, D.; Nesburn, A.; Small, K. Keratoconus—No association with the transforming growth factor beta-induced gene in a cohort of American patients. Cornea 2004, 23, 13–17. [Google Scholar] [CrossRef] [PubMed]
  44. Gajecka, M.; Radhakrishna, U.; Winters, D.; Nath, S.K.; Rydzanicz, M.; Ratnamala, U.; Ewing, K.; Molinari, A.; Pitarque, J.A.; Kwanghyuk, L.; et al. Localization of a gene for keratoconus to a 5.6-Mb interval on 13q32. Investig. Ophthalmol. Vis. Sci. 2009, 50, 1531–1539. [Google Scholar] [CrossRef] [PubMed]
  45. Adzhubei, I.A.; Schmidt, S.; Peshkin, L.; Ramensky, V.E.; Gerasimova, A.; Bork, P.; Kondrashov, A.S.; Sunyaev, S.R. A method and server for predicting damaging missense mutations. Nat. Methods 2010, 7, 248–249. [Google Scholar] [CrossRef] [PubMed]
  46. Hasanian-Langroudi, F.; Saravani, R.; Validad, M.H.; Bahari, G.; Yari, D. Association of Lysyl oxidase (LOX) polymorphisms with the risk of keratoconus in an Iranian population. Ophthalmic Genet. 2015, 36, 309–314. [Google Scholar] [CrossRef]
  47. Hao, X.D.; Chen, P.; Chen, Z.L.; Li, S.X.; Wang, Y. Evaluating the association between keratoconus and reported genetic loci in a Han Chinese population. Ophthalmic Genet. 2015, 36, 132–136. [Google Scholar] [CrossRef]
  48. Dudakova, L.; Palos, M.; Jirsova, K.; Stranecky, V.; Krepelova, A.; Hysi, P.G.; Liskova, P. Validation of rs2956540:G>C and rs3735520:G>A association with keratoconus in a population of European descent. Eur. J. Hum. Genet. 2015, 23, 1581–1583. [Google Scholar] [CrossRef]
  49. Bykhovskaya, Y.; Li, X.; Taylor, K.D.; Haritunians, T.; Rotter, J.I.; Rabinowitz, Y.S. Linkage analysis of high-density SNPs confirms keratoconus locus at 5q chromosomal region. Ophthalmic Genet. 2016, 37, 109–110. [Google Scholar] [CrossRef]
  50. Li, X.; Bykhovskaya, Y.; Tang, Y.G.; Picornell, Y.; Haritunians, T.; Aldave, A.J.; Szczotka-Flynn, L.; Iyengar, S.K.; Rotter, J.I.; Taylor, K.D.; et al. An association between the calpastatin (CAST) gene and keratoconus. Cornea 2013, 32, 696–701. [Google Scholar] [CrossRef]
  51. Burdon, K.P.; Macgregor, S.; Bykhovskaya, Y.; Javadiyan, S.; Li, X.; Laurie, K.J.; Muszynska, D.; Lindsay, R.; Lechner, J.; Haritunians, T.; et al. Association of polymorphisms in the hepatocyte growth factor gene promoter with keratoconus. Investig. Ophthalmol. Vis. Sci. 2011, 52, 8514–8519. [Google Scholar] [CrossRef]
  52. Shetty, R.; Sathyanarayanamoorthy, A.; Ramachandra, R.A.; Arora, V.; Ghosh, A.; Srivatsa, P.R.; Pahuja, N.; Nuijts, R.M.M.A.; Sinha-Roy, A.; Mohan, R.R.; et al. Attenuation of lysyl oxidase and collagen gene expression in keratoconus patient corneal epithelium corresponds to disease severity. Mol. Vis. 2015, 21, 12–25. [Google Scholar]
  53. Kristianslund, O.; Drolsum, L. Prevalence of keratoconus in persons with Down syndrome: A review. BMJ Open Ophthalmol. 2021, 6, e000754. [Google Scholar] [CrossRef] [PubMed]
  54. 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] [PubMed]
  55. Mathan, J.J.; Gokul, A.; Simkin, S.K.; Meyer, J.J.; McGhee, C.N.J. Keratoconus in Down syndrome: Prevalence, risk factors, severity and corneal tomographic characteristics. Clin. Exp. Ophthalmol. 2024, 52, 22–30. [Google Scholar] [CrossRef]
  56. 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]
  57. Mathan, J.J.; Gokul, A.; Simkin, S.K.; Meyer, J.J.; Patel, D.V.; McGhee, C.N.J. Topographic screening reveals keratoconus to be extremely common in Down syndrome. Clin. Exp. Ophthalmol. 2020, 48, 1160–1167. [Google Scholar] [CrossRef]
  58. Hashemi, H.; Miraftab, M.; Amanzadeh, K.; Seyedian, M.A.; Vinciguerra, R.; Ambrósio, R.; Roberts, C.; Makateb, A.; Vinciguerra, P.; Asgari, S. Keratoconus detection by novel indices in patients with Down syndrome: A cohort population-based study. Jpn. J. Ophthalmol. 2020, 64, 285–291. [Google Scholar] [CrossRef]
  59. Vincent, A.L.; Weiser, B.A.; Cupryn, M.; Stein, R.M.; Abdolell, M.; Levin, A.V. Computerized corneal topography in a paediatric population with Down syndrome. Clin. Exp. Ophthalmol. 2005, 33, 47–52. [Google Scholar] [CrossRef]
  60. Gittenberger-de Groot, A.C.; Bartram, U.; Oosthoek, P.W.; Bartelings, M.M.; Hogers, B.; Poelmann, R.E.; Jongewaard, I.N.; Klewer, S.E. Collagen type VI expression during cardiac development and in human fetuses with trisomy 21. Anat. Rec. A Discov. Mol. Cell Evol. Biol. 2003, 275, 1109–1116. [Google Scholar] [CrossRef]
  61. Davies, G.E.; Howard, C.M.; Farrer, M.J.; Coleman, M.M.; Bennett, L.B.; Cullen, L.M.; Wyse, R.K.; Burn, J.; Williamson, R.; Kessling, A.M. Genetic variation in the COL6A1 region is associated with congenital heart defects in trisomy 21 (Down’s syndrome). Ann. Hum. Genet. 1995, 59, 253–269. [Google Scholar] [CrossRef]
  62. von Kaisenberg, C.S.; Brand-Saberi, B.; Christ, B.; Vallian, S.; Farzaneh, F.; Nicolaides, K.H. Collagen type VI gene expression in the skin of trisomy 21 fetuses. Obstet. Gynecol. 1998, 91, 319–323. [Google Scholar] [CrossRef]
  63. Quarello, E.; Guimiot, F.; Moalic, J.M.; Simoneau, M.; Ville, Y.; Delezoide, A.L. Quantitative evaluation of collagen type VI and SOD gene expression in the nuchal skin of human fetuses with trisomy 21. Prenat. Diagn. 2007, 27, 926–931. [Google Scholar] [CrossRef] [PubMed]
  64. Gulesserian, T.; Seidl, R.; Hardmeier, R.; Cairns, N.; Lubec, G. Superoxide dismutase SOD1, encoded on chromosome 21, but not SOD2 is overexpressed in brains of patients with Down syndrome. J. Investig. Med. 2001, 49, 41–46. [Google Scholar] [CrossRef] [PubMed]
  65. Nemet, A.Y.; Vinker, S.; Bahar, I.; Kaiserman, I. The association of keratoconus with immune disorders. Cornea 2010, 29, 1261–1264. [Google Scholar] [CrossRef]
  66. Chen, X.; Chen, L. Causal Links Between Systemic Disorders and Keratoconus in European Population. Am. J. Ophthalmol. 2024, 265, 189–199. [Google Scholar] [CrossRef]
  67. 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]
  68. 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]
  69. 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]
  70. 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]
  71. 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]
  72. Atilano, S.; Lee, D.; Fukuhara, P.; Chwa, M.; Nesburn, A.; Udar, N.; Kenney, M.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, 62–70. [Google Scholar]
  73. 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] [PubMed]
  74. 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] [PubMed]
  75. 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]
  76. 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]
  77. 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]
  78. Kenney, M.C.; Brown, D.J. The cascade hypothesis of keratoconus. Cont. Lens Anterior Eye. 2003, 26, 139–146. [Google Scholar] [CrossRef]
  79. Liu, X.; Zhang, X.; Li, Q.; Wei, C.; Guo, X.; Zhao, L.; Liu, T.; Bao, Q.; Dou, S.; Seitz, B.; et al. CYR61/CCN1: A novel mediator of redox response in corneal stromal cells of keratoconus. Exp. Eye Res. 2024, 248, 110093. [Google Scholar] [CrossRef]
  80. Cantemir, A.; Alexa, A.; Ciobica, A.; Balmus, I.; Antioch, I.; Stoica, B.; Chiselita, D.; Costin, D. Evaluation of antioxidant enzymes in keratoconus. Revista de Chimie 2016, 67, 1725–1727. [Google Scholar]
  81. Tekin, S.; Seven, E. Assessment of serum catalase, reduced glutathione, and superoxide dismutase activities and malondialdehyde levels in keratoconus patients. Eye 2022, 36, 2062–2066. [Google Scholar] [CrossRef]
  82. 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]
  83. 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]
  84. Galvis, V.; Sherwin, T.; Tello, A.; Merayo, J.; Barrera, R.; Acera, A. Keratoconus: An inflammatory disorder? Eye 2015, 29, 843–859. [Google Scholar] [CrossRef] [PubMed]
  85. Lema, I.; Durán, J.A. Inflammatory molecules in the tears of patients with keratoconus. Ophthalmology 2005, 112, 654–659. [Google Scholar] [CrossRef] [PubMed]
  86. Lema, I.; Sobrino, T.; Durán, J.A.; Brea, D.; Díez-Feijoo, E. Subclinical keratoconus and inflammatory molecules from tears. Br. J. Ophthalmol. 2009, 93, 820–824. [Google Scholar] [CrossRef]
  87. Pásztor, D.; Kolozsvári, B.L.; Csutak, A.; Berta, A.; Hassan, Z.; Ujhelyi, B.; Gogolák, P.; Fodor, M. Tear mediators in corneal ectatic disorders. PLoS ONE 2016, 11, e0153186. [Google Scholar] [CrossRef]
  88. Sorkhabi, R.; Ghorbanihaghjo, A.; Taheri, N.; Ahoor, M.H. Tear film inflammatory mediators in patients with keratoconus. Int. Ophthalmol. 2015, 35, 467–472. [Google Scholar] [CrossRef]
  89. Sobrino, T.; Regueiro, U.; Malfeito, M.; Vieites-Prado, A.; Pérez-Mato, M.; Campos, F.; Lema, I. Higher expression of Toll-like receptors 2 and 4 in blood cells of keratoconus patients. Sci. Rep. 2017, 7, 12975. [Google Scholar] [CrossRef]
  90. Marques, J.C.; Ladislau de Carvalho, K.I.; Xavier, R.; Nosé, W.; Rizzo, L.V. Inflammatory profile of keratoconic corneal epithelium. BMC Ophthalmol. 2023, 23, 326. [Google Scholar] [CrossRef]
  91. Karaca, E.E.; Özmen, M.C.; Ekici, F.; Yüksel, E.; Türkoğlu, Z. Neutrophil-to-lymphocyte ratio may predict progression in patients with keratoconus. Cornea 2014, 33, 1168–1173. [Google Scholar] [CrossRef]
  92. Regueiro, U.; López-López, M.; Hervella, P.; Sobrino, T.; Lema, I. Corneal and conjunctival alteration of innate immune expression in first-degree relatives of keratoconus patients. Graefes Arch. Clin. Exp. Ophthalmol. 2021, 259, 459–467. [Google Scholar] [CrossRef]
  93. Regueiro, U.; López-López, M.; Varela-Fernández, R.; Sobrino, T.; Diez-Feijoo, E.; Lema, I. Immunomodulatory effect of human lactoferrin on Toll-like receptors 2 expression as therapeutic approach for keratoconus. Int. J. Mol. Sci. 2022, 23, 12350. [Google Scholar] [CrossRef] [PubMed]
  94. Ben-Shaul, O.; Segal, A.; Schwartz, S.; Stein, N.; Hyams, M.; Saliba, W.; Mimouni, M. Factors associated with keratoconus in Israel—A cross-sectional population-based study. Acta Ophthalmol. 2024, 102, e1011–e1017. [Google Scholar] [CrossRef] [PubMed]
  95. Alqasimi, N.A.; Aljohani, L.H.; Ambrósio, R.; AlQahtani, B.S.; Al Haydar, N.S.; Alanazi, B.R.; Alfurayhan, D.T.; Saber, L.S.H.; Alsalem, F.S.; Alqahtani, N.A.; et al. Assessment of awareness of keratoconus and its relation to eye rubbing among Saudi Arabia population. Front. Ophthalmol. 2025, 5, 1545030. [Google Scholar] [CrossRef]
  96. Shah, Z.; Purohit, D.; Danayak, P. Keratoconus characteristics and associations: A cross-sectional keratoconus study in western India (CKSWI). Indian J. Ophthalmol. 2024, 72, 704–711. [Google Scholar] [CrossRef]
  97. Lo, J.E.; Huang, Y.H.; Bhattacharyya, N.; Moulton, E.A.; Ma, K.S.K. Allergic rhinitis and keratoconus: A systematic review and meta-analysis. J. Allergy Clin. Immunol. Pract. 2024, 12, 3096–3104. [Google Scholar] [CrossRef]
  98. Chang, Y.; Huang, T.; Yang, S.; Li, Y.; Chen, D. Causal association between atopic dermatitis and keratoconus: A Mendelian randomization study. Transl. Vis. Sci. Technol. 2024, 13, 13. [Google Scholar] [CrossRef]
  99. Balogun, M.M.; Fashola, M.B. Association between keratoconus and allergic conjunctivitis in children attending a tertiary hospital in Nigeria. Rom. J. Ophthalmol. 2023, 67, 134–139. [Google Scholar] [CrossRef]
  100. Xu, H.; Wen, Y.; Zheng, H.; Jiang, D.; Chen, W. Allergic disease and keratoconus: A two-sample univariable and multivariable Mendelian randomization study. World Allergy Organ. J. 2024, 17, 100993. [Google Scholar] [CrossRef]
  101. Wang, Q.; Deng, Y.; Li, S.; Du, X.; Zhao, X.; Zhang, T.; Yuan, J. Corneal biomechanical changes in allergic conjunctivitis. Eye Vis. 2021, 8, 17. [Google Scholar] [CrossRef]
  102. Naderan, M.; Rajabi, M.T.; Zarrinbakhsh, P.; Bakhshi, A. Effect of allergic diseases on keratoconus severity. Ocul. Immunol. Inflamm. 2017, 25, 418–423. [Google Scholar] [CrossRef]
  103. Gijs, M.; Adelaar, T.I.; Vergouwen, D.P.C.; Visser, N.; Dickman, M.M.; Ollivier, R.C.I.; Berendschot, T.T.J.M.; Nuijts, R.M.M.A. Tear fluid inflammatory proteome analysis highlights similarities between keratoconus and allergic conjunctivitis. Investig. Ophthalmol. Vis. Sci. 2023, 64, 9. [Google Scholar] [CrossRef] [PubMed]
  104. Merdler, I.; Hassidim, A.; Sorkin, N.; Shapira, S.; Gronovich, Y.; Korach, Z. Keratoconus and allergic diseases among Israeli adolescents between 2005 and 2013. Cornea 2005, 34, 525–529. [Google Scholar] [CrossRef] [PubMed]
  105. Wajnsztajn, D.; Solomon, A. Vernal keratoconjunctivitis and keratoconus. Curr. Opin. Allergy Clin. Immunol. 2021, 21, 507–514. [Google Scholar] [CrossRef] [PubMed]
  106. Marx-Gross, S.; Fieß, A.; Münzel, T.; Wild, P.S.; Beutel, M.E.; Schmidtmann, I.; Lackner, K.J.; Pfeiffer, N.; Schuster, A.K.G. Much higher prevalence of keratoconus than announced results of the Gutenberg Health Study (GHS). Graefes Arch. Clin. Exp. Ophthalmol. 2023, 261, 3241–3247. [Google Scholar] [CrossRef]
  107. Guo, X.H.; Bian, J.; Yang, K.; Liu, X.; Sun, Y.; Liu, M.; Qi, X.; Ren, S.; Dong, Y.; Gao, H. Eye Rubbing in Chinese Patients With Keratoconus: A Multicenter Analysis. J. Refract. Surg. 2023, 39, 712–718. [Google Scholar] [CrossRef]
  108. AlSomali, A.; Almithn, D.; Alamer, A.; Al-Omair, A.; Almuhaysin, F.; Almulhim, N. Awareness of Keratoconus and Its Relationship With Eye Rubbing Among the Population of the Eastern Province of Saudi Arabia. Cureus 2024, 16, e51627. [Google Scholar] [CrossRef]
  109. Duarte-Bueno, M.L.; Tello, A.; Diaz-Martínez, A.L.; Escobar, S.; Galvis, V. Evaluating the Influence of Eye Rubbing and Genetic Predisposition on Keratoconus in Bucaramanga (Colombia): A Case-control Study. Cesk Slov. Oftalmol. 2025, 81, 1–10. [Google Scholar] [CrossRef]
  110. Mazharian, A.; Flamant, R.; Elahi, S.; Panthier, C.; Rampat, R.; Gatinel, D. Medium to long term follow up study of the efficacy of cessation of eye-rubbing to halt progression of keratoconus. Front. Med. 2023, 10, 1152266. [Google Scholar] [CrossRef]
  111. Hage, A.; Knoeri, J.; Leveziel, L.; Majoulet, A.; Blanc, J.V.; Buffault, J.; Labbé, A.; Baudouin, C. EYERUBBICS: The Eye Rubbing Cycle Study. J. Clin. Med. 2023, 12, 1529. [Google Scholar] [CrossRef]
  112. Yin, S.; Xu, L.; Yang, K.; Fan, Q.; Gu, Y.; Yin, C.; Zang, Y.; Wang, Y.; Yuan, Y.; Chang, A.; et al. Gene–Environment Interaction Between CAST Gene and Eye-Rubbing in the Chinese Keratoconus Cohort Study: A Case-Only Study. Investig. Ophthalmol. Vis. Sci. 2024, 65, 36. [Google Scholar] [CrossRef]
  113. McMonnies, C.W. Mechanisms of rubbing-related corneal trauma in keratoconus. Cornea 2009, 28, 607–615. [Google Scholar] [CrossRef] [PubMed]
  114. Ferrini, E.; Posarelli, C.; Figus, M.; Lisi, D.; Gabbriellini, G. A pilot study on Langerhans cells in keratoconus patients by in vivo confocal microscopy before and after corneal cross-linking and correlation with eye rubbing. Cont. Lens Anterior Eye 2024, 47, 102170. [Google Scholar] [CrossRef] [PubMed]
  115. Alamri, A.; Alrizqi, A.A.; Aljohani, A.A.; Alzahrani, D.A.; Alassaf, O.M.; Hamzi, Y.A.; Alharbi, N.M.; Alharbi, B.A.; Taha, M., Sr. Awareness of Keratoconus and Its Association With Eye Rubbing Among the Population in Aseer Province. Cureus 2023, 15, e41271. [Google Scholar] [CrossRef]
  116. Bikle, D.D. Vitamin D regulation of immune function. Vitam. Horm. 2011, 86, 1–21. [Google Scholar] [CrossRef]
  117. 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]
  118. 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]
  119. 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]
  120. 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]
  121. 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, 7715–7727. [Google Scholar] [CrossRef]
  122. Wimalawansa, S.J. Vitamin D Deficiency: Effects on Oxidative Stress, Epigenetics, Gene Regulation, and Aging. Biology 2019, 8, 30. [Google Scholar] [CrossRef]
  123. Liu, W.; Zhang, L.; Xu, H.J.; Li, Y.; Hu, C.M.; Yang, J.Y.; Sun, M.Y. The Anti-Inflammatory Effects of Vitamin D in Tumorigenesis. Int. J. Mol. Sci. 2018, 19, 2736. [Google Scholar] [CrossRef] [PubMed]
  124. Lai, Y.T.; Cerquinho, R.G.; Perez, M.M.; Alves Bda, C.A.; Pereira, E.C.; Azzalis, L.A.; Campos Junqueira, V.B.; 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, e22830. [Google Scholar] [CrossRef] [PubMed]
  125. Alsalem, J.A.; Patel, D.; Susarla, R.; Coca-Prados, M.; Bland, R.; Walker, E.A.; Rauz, S.; Wallace, G.R. Characterization of vitamin D production by human ocular barrier cells. Investig. Ophthalmol. Vis. Sci. 2014, 55, 2140–2147. [Google Scholar] [CrossRef] [PubMed]
  126. Knapp, A.A. Results of vitamin-D-complex treatment of keratoconus. Preliminary study. Am. J. Ophthalmol. 1939, 22, 289–292. [Google Scholar] [CrossRef]
  127. Akkaya, S.; Ulusoy, D.M. Serum Vitamin D Levels in Patients with Keratoconus. Ocul. Immunol. Inflamm. 2020, 28, 348–353. [Google Scholar] [CrossRef]
  128. Aslan, M.G.; Findik, H.; Okutucu, M.; Aydin, 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]
  129. 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]
  130. Mackawy, A.M.H.; Al-Ayed, B.M.; Al-Rashidi, B.M. Vitamin D deficiency and its association with thyroid disease. Int J Health Sci (Qassim). 2013, 7, 267–275. [Google Scholar] [CrossRef]
  131. Stagi, S.; Lapi, E.; Romano, S.; Bargiacchi, S.; Brambilla, A.; Giglio, S.; Seminara, s.; De Martino, M. Determinants of vitamin D levels in children and adolescents with Down syndrome. Int. J. Endocrinol. 2015, 2015, 896758. [Google Scholar] [CrossRef]
  132. Eren, E.; Ellidag, H.Y.; Cekin, Y.; Ayoglu, R.U.; Sekercioglu, A.O.; Yilmaz, N. Heart valve disease: The role of calcidiol deficiency, elevated parathyroid hormone levels and oxidative stress in mitral and aortic valve insufficiency. Redox Rep. 2014, 19, 34–39. [Google Scholar] [CrossRef]
  133. Archontogeorgis, K.; Nena, E.; Papanas, N.; Steiropoulos, P. The role of vitamin D in obstructive sleep apnoea syndrome. Breathe (Sheff) 2018, 14, 206–215. [Google Scholar] [CrossRef] [PubMed]
  134. Hollams, E.M.; Hart, P.H.; Holt, B.J.; Serralha, M.; Parsons, F.; De Klerk, N.H.; Zhang, G.; Sly, P.D.; Holt, P.G. Vitamin D and atopy and asthma phenotypes in children: A longitudinal cohort study. Eur. Respir. J. 2011, 38, 1320–1327. [Google Scholar] [CrossRef] [PubMed]
  135. 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]
  136. Bilgihan, K.; Hondur, A.; Sul, S.; Ozturk, S. Pregnancy-induced progression of keratoconus. Cornea 2011, 30, 991–994. [Google Scholar] [CrossRef]
  137. Manios, Y.; Moschonis, G.; Lambrinou, C.P.; Tsoutsoulopoulou, K.; Binou, P.; Karachaliou, A.; Breidenassel, C.; Gonzalez-Gross, M.; Kiely, M.; Cashman, K.D. A systematic review of vitamin D status in southern European countries. Eur. J. Nutr. 2018, 57, 2001–2036. [Google Scholar] [CrossRef]
  138. Huh, S.Y.; Gordon, C.M. Vitamin D deficiency in children and adolescents: Epidemiology, impact and treatment. Rev. Endocr. Metab. Disord. 2008, 9, 161–170. [Google Scholar] [CrossRef]
  139. Naderan, M.; Rajabi, M.T.; Zarrinbakhsh, P.; Farjadnia, M. Is keratoconus more severe in pediatric population? Int. Ophthalmol. 2017, 37, 1169–1173. [Google Scholar] [CrossRef]
  140. Naderan, M.; Jahanrad, A. Topographic, tomographic and biomechanical corneal changes during pregnancy in patients with keratoconus: A cohort study. Acta Ophthalmol. 2017, 95, e291–e296. [Google Scholar] [CrossRef]
  141. López-López, M.; Regueiro, U.; Bravo, S.B.; Chantada-Vázquez Mdel, 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]
  142. Gupta, P.; Pathak, M.; Thakur, B.; Fogla, R.; Agarwal, A.; Ram, J. Association of keratoconus with serum levels of 25-hydroxyvitamin D and antioxidant trace elements: A systematic review and meta-analysis. Indian J. Ophthalmol. 2022, 70, 2818–2824. [Google Scholar] [CrossRef]
  143. Knapp, A.A. Vitamin D Complex in Keratoconus: Etiology, Pathology and Treatment of Conical Cornea: Preliminary Report. J. Am. Med. Assoc. 1938, 110, 1993–1994. [Google Scholar] [CrossRef]
  144. Mutti, D.O.; Marks, A.R. Blood levels of vitamin D in teens and young adults with myopia. Optom. Vis. Sci. 2011, 88, 377–382. [Google Scholar] [CrossRef] [PubMed]
  145. Vitar, R.M.L.; Fonteyne, P.; Knutsson, K.A.; Bertuzzi, F.; Galli, L.; Rama, P.; Ferrari, G. Vitamin D Supplementation Impacts Systemic Biomarkers of Collagen Degradation and Copper Metabolism in Patients With Keratoconus. Transl. Vis. Sci. Technol. 2022, 11, 16. [Google Scholar] [CrossRef] [PubMed]
  146. Bartolomeo, N.; Pederzolli, M.; Palombella, S.; Fonteyne, P.; Suanno, G.; Tilaro, G.; de Pretis, S.; Borgo, F.; Bertuzzi, F.; Senni, C.; et al. The Effects of Vitamin D on Keratoconus Progression. Am. J. Ophthalmol. 2025, 276, 235–251. [Google Scholar] [CrossRef]
  147. 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]
  148. 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]
  149. Yu, H.; Wu, J.; Li, K.; Huang, Y.; Wu, T.; Wang, L.; Huang, Y. Integrated analysis of murine cornea identifies JAK/STAT signaling pathway upregulated specifically in female Vitamin A Deficient mice. Exp. Eye Res. 2023, 237, 109714. [Google Scholar] [CrossRef]
  150. Sozer, O.; Ozalp, O.; Atalay, E.; Demir, S.S.; Alatas, İ.O.; Yildirim, N. Comparison of blood levels of vitamin B12, folic acid, riboflavin, and homocysteine in keratoconus and healthy subjects. J. Cataract. Refract. Surg. 2023, 49, 589–594. [Google Scholar] [CrossRef]
  151. 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]
  152. 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]
  153. 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] [PubMed]
  154. Dudakova, L.; Liskova, P.; Jirsova, K. Is copper imbalance an environmental factor influencing keratoconus development? Med. Hypotheses 2015, 84, 518–524. [Google Scholar] [CrossRef]
  155. 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]
  156. 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. Vestn. Oftalmol. 2015, 131, 34–42. [Google Scholar] [CrossRef]
  157. 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]
  158. 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]
  159. 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]
  160. 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]
  161. Gao, J.F.; Dong, Y.Y.; Jin, X.; Dai, L.J.; Wang, J.R.; Zhang, H. Identification and Verification of Ferroptosis-Related Genes in Keratoconus Using Bioinformatics Analysis. J. Inflamm. Res. 2024, 17, 2383–2397. [Google Scholar] [CrossRef]
  162. Kumar, N.R.; Khamar, P.; Kannan, R.; Padmanabhan, A.; Shetty, R.; D’Souza, S.; Vaidya, T.; Sethu, S.; Ghosh, A. Distinct Ocular Surface Microbiome in Keratoconus Patients Correlate With Local Immune Dysregulation. Investig. Ophthalmol. Vis. Sci. 2025, 66, 60. [Google Scholar] [CrossRef]
  163. Rocha-de-Lossada, C.; Mazzotta, C.; Gabrielli, F.; Papa, F.T.; Gómez-Huertas, C.; García-López, C.; Urbinati, F.; Rachwani-Anil, R.; García-Lorente, M.; Sánchez-González, J.M.; et al. Ocular Surface Microbiota in Naïve Keratoconus: A Multicenter Validation Study. J. Clin. Med. 2023, 12, 6354. [Google Scholar] [CrossRef] [PubMed]
  164. Tunç, U.; Çelebi, A.C.; Ekren, B.Y.; Yıldırım, Y.; Kepez Yıldız, B.; Okullu, S.Ö.; Sezerman, O.U. Corneal bacterial microbiome in patients with keratoconus using next-generation sequencing-based 16S rRNA gene analysis. Exp. Eye Res. 2023, 228, 109402. [Google Scholar] [CrossRef] [PubMed]
  165. Kaur, P.; Moon, L.; Srikumaran, D.; Salzberg, S.L.; Lu, J.; Simner, P.J.; Soiberman, U.S. No Evidence of Chronic Infection in a Metagenomic Sequencing Study of the Keratoconus Corneal Epithelium. J. Clin. Med. 2024, 13, 3399. [Google Scholar] [CrossRef] [PubMed]
  166. Sharif, R.; Sejersen, H.; Frank, G.; Hjortdal, J.; Karamichos, D. Effects of collagen cross-linking on the keratoconus metabolic network. Eye 2018, 32, 1271–1281. [Google Scholar] [CrossRef]
  167. 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]
  168. 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]
  169. Blokhina, O.; Virolainen, E.; Fagerstedt, K.V. Antioxidants, oxidative damage and oxygen deprivation stress: A review. Ann. Bot. 2003, 91, 179–194. [Google Scholar] [CrossRef]
  170. 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]
  171. Whelchel, A.E.; McKay, T.B.; Priyadarsini, S.; Rowsey, T.; Karamichos, D. Association between Diabetes and Keratoconus: A Retrospective Analysis. Sci. Rep. 2019, 9, 18521. [Google Scholar] [CrossRef]
  172. 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]
  173. 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]
  174. 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]
  175. Wojakowska, A.; Pietrowska, A.; 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]
  176. 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] [PubMed]
  177. Peris-Martínez, C.; Piá-Ludeña, J.V.; Rog-Revert, M.J.; Fernández-López, E.; Domingo, J.C. Antioxidant and Anti-Inflammatory Effects of Oral Supplementation with a Highly-Concentrated Docosahexaenoic Acid (DHA) Triglyceride in Patients with Keratoconus: A Randomized Controlled Preliminary Study. Nutrients 2023, 15, 1300. [Google Scholar] [CrossRef]
  178. Pihlblad, M.S.; Schaefer, D.P. Eyelid laxity, obesity, and obstructive sleep apnea in keratoconus. Cornea 2013, 32, 1232–1236. [Google Scholar] [CrossRef]
  179. Eliasi, E.; Bez, M.; Megreli, J.; Avramovich, E.; Fischer, N.; Barak, A.; Levine, H. The Association Between Keratoconus and Body Mass Index: A Population-Based Cross-Sectional Study Among Half a Million Adolescents. Am. J. Ophthalmol. 2021, 224, 200–206. [Google Scholar] [CrossRef]
  180. Wang, J.; Liu, F.; Mo, J.; Gong, D.; Zheng, F.; Su, J.; Ding, S.; Yang, W.; Guo, P. Exploring the causal relationship between body mass index and keratoconus: A Mendelian randomization study. Front. Med. 2024, 11, 1402108. [Google Scholar] [CrossRef]
  181. Ren, S.; Tu, R.; Xu, L.; Gu, Y.; Fan, Q.; Wang, Q.; Zhu, M.; Yin, S.; Pang, C.; Zhao, D.; et al. A high body mass index strengthens the association between the time of eye rubbing and keratoconus in a Chinese population: A case control study. BMC Public Health 2023, 23, 2032. [Google Scholar] [CrossRef]
  182. Skorin, L.; Knutson, R. Ophthalmic Diseases in Patients With Obstructive Sleep Apnea. J. Am. Osteopath. Assoc. 2016, 116, 522–529. [Google Scholar] [CrossRef]
  183. Salinas, R.; Puig, M.; Fry, C.L.; Johnson, D.A.; Kheirkhah, A. Floppy eyelid syndrome: A comprehensive review. Ocul. Surf. 2020, 18, 31–39. [Google Scholar] [CrossRef] [PubMed]
  184. Bulloch, G.; Seth, I.; Zhu, Z.; Sukumar, S.; McNab, A. Ocular manifestations of obstructive sleep apnea: A systematic review and meta-analysis. Graefes Arch. Clin. Exp. Ophthalmol. 2024, 262, 19–32. [Google Scholar] [CrossRef] [PubMed]
  185. Karaca, U.; Akıncıoğlu, D.; Ayyildiz, O.; Dogan, D.; Ozge, G.; Usta, G.; Mutlu, F.M. Comparison of obstructive sleep apnea syndrome and keratoconus patients on elevation maps. Int. Ophthalmol. 2022, 42, 933–938. [Google Scholar] [CrossRef]
  186. Seiler, T.; Huhle, S.; Spoerl, E.; Kunath, H. Manifest diabetes and keratoconus: A retrospective case-control study. Graefes Arch. Clin. Exp. Ophthalmol. 2000, 238, 822–825. [Google Scholar] [CrossRef]
  187. Naderan, M.; Naderan, M.; Rezagholizadeh, F.; Zolfaghari, M.; Pahlevani, R.; Rajabi, M.T. Association between diabetes and keratoconus: A case-control study. Cornea 2014, 33, 1271–1273. [Google Scholar] [CrossRef]
  188. Naderan, M.; Shoar, S.; Rezagholizadeh, F.; Zolfaghari, M.; Naderan, M. Characteristics and associations of keratoconus patients. Cont. Lens Anterior Eye 2015, 38, 199–205. [Google Scholar] [CrossRef]
  189. Akowuah, P.K.; Arthur, C.; Otabil, F.A.; Ofori, C.A.; Osei-Poku, K.; Fummey, J.; Boadi, P.; Dadzie, E.E. Association between diabetes and keratoconus–a systematic review and meta-analysis. Eur. J. Ophthalmol. 2022, 32, 23–30. [Google Scholar] [CrossRef]
  190. Dong, X.X.; Liu, K.F.; Zhou, M.; Liang, G.; Pan, C.W. Diabetes Mellitus and Keratoconus: A Systematic Review and Meta-Analysis. Cornea 2022, 41, 1398–1404. [Google Scholar] [CrossRef]
  191. Kuo, I.C.; Broman, A.; Pirouzmanesh, A.; Melia, M. Is there an association between diabetes and keratoconus? Ophthalmology 2006, 113, 184–190. [Google Scholar] [CrossRef]
  192. 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]
  193. Said, O.M.; Iqbal, M.; El-Massry, A.; Elgharieb, M.E.; Mady, M.; Sharawy, A.M.; Abdelaziz, K. Thyroid gland dysfunction and keratoconus. Med. Hypothesis Discov. Innov. Ophthalmol. 2024, 13, 104–111. [Google Scholar] [CrossRef] [PubMed]
  194. Chung, I.K.; Kim, B.S.; Han, K.D.; Yoo, Y.S.; Kim, H.; Jeong, C. Ten-year incidence of keratoconus in relation to sex, age, and thyroid gland dysfunction: A nationwide population-based cohort study (2009–2018). Ann. Transl. Med. 2024, 12, 45. [Google Scholar] [CrossRef] [PubMed]
  195. Roszkowska, A.M.; Alessandrello, F.; Waśniewska, M.; Tropeano, A.; Gargano, R.; Aragona, P. Is keratoconus associated to thyroid diseases? Assessment of the corneal parameters in patients with congenital hypothyroidism. Eur. J. Ophthalmol. 2022, 32, 31–35. [Google Scholar] [CrossRef]
  196. Razafimino, S.; Flockerzi, E.; Zemova, E.; Munteanu, C.; Seitz, B. Impact of Hypothyroidism on Tomography and Biomechanics in Keratoconus—Cross-Sectional and Longitudinal Assessment within the Homburg Keratoconus Center at the Time of Inclusion and after 1 Year. Klin. Monbl Augenheilkd 2023, 240, 1185–1191. [Google Scholar] [CrossRef]
  197. 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]
  198. 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]
  199. 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]
  200. Stachon, T.; Omar Ali, M.; Latta, L.; Huessein, G.H.; Mohamed, T.A.; Soliman, W.; Seitz, B.; Szentmary, N. Effect of Thyroxine on Transforming Growth Factor β1, Collagen I, and V Expression in Keratoconus Corneal Fibroblasts and Keratocytes, in Vitro. Curr. Eye Res. 2022, 47, 206–213. [Google Scholar] [CrossRef]
  201. Claessens, J.L.J.; Godefrooij, D.A.; Vink, G.; Frank, L.E.; Wisse, R.P.L. Nationwide epidemiological approach to identify associations between keratoconus and immune-mediated diseases. Br. J. Ophthalmol. 2022, 106, 1350–1354. [Google Scholar] [CrossRef]
  202. Karamichos, D.; Escandon, P.; Vasini, B.; Nicholas, S.E.; Van, L.; Dang, D.H.; Cunningham, R.L.; Riaz, K. Anterior pituitary, sex hormones, and keratoconus: Beyond traditional targets. Prog. Retin. Eye Res. 2022, 88, 101016. [Google Scholar] [CrossRef]
  203. Wagner, H.; Barr, J.T.; Zadnik, K. Collaborative Longitudinal Evaluation of Keratoconus (CLEK) Study: Methods and findings to date. Cont. Lens Anterior Eye. 2007, 30, 223–232. [Google Scholar] [CrossRef] [PubMed]
  204. Hashemi, H.; Beiranvand, A.; Khabazkhoob, M.; Asgari, S.; Emamian, M.H.; Shariati, M.; Fotouhi, A. Prevalence of keratoconus in a population-based study in Shahroud. Cornea 2013, 32, 1441–1445. [Google Scholar] [CrossRef] [PubMed]
  205. Hashemi, H.; Khabazkhoob, M.; Yazdani, N.; Ostadimoghaddam, H.; Norouzirad, R.; Amanzadeh, K.; Miraftab, M.; Derakhshan, A.; Yekta, A.A. The prevalence of keratoconus in a young population in Mashhad, Iran. Ophthalmic Physiol. Opt. 2014, 34, 519–527. [Google Scholar] [CrossRef]
  206. Rabinowitz, Y.S. Keratoconus. Surv. Ophthalmol. 1998, 42, 297–319. [Google Scholar] [CrossRef]
  207. Yin, H.; Luo, C.; Tian, Y.; Deng, Y. Altered expression of sex hormone receptors in keratoconus corneas. Biomed. Res.-India 2017, 28, 5089–5092. [Google Scholar]
  208. 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. Cont. Lens Anterior Eye. 2019, 42, 492–496. [Google Scholar] [CrossRef]
  209. 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]
  210. Zhao, X.; Yuan, Y.; Sun, T.; Zhang, Y.; Chen, Y. Associations Between Keratoconus and the Level of Sex Hormones: A Cross-Sectional Study. Front. Med. 2022, 9, 828233. [Google Scholar] [CrossRef]
  211. 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]
  212. Jamali, H.; Heydari, M.; Masihpour, N.; Khosravi, A.; Zare, M.; Shams, M.; Omrani, G.R. Serum androgens and prolactin levels in patients with keratoconus. Clin. Exp. Optom. 2023, 106, 484–488. [Google Scholar] [CrossRef]
  213. Deitel, C.M.; Chen, K.H.; Uber, I.C. Possible association of keratoconus progression with gender-affirming hormone therapy: A case report. Am. J. Ophthalmol. Case Rep. 2023, 30, 101850. [Google Scholar] [CrossRef]
  214. Beardsley, T.L.; Foulks, G.N. An association of keratoconus and mitral valve prolapse. Ophthalmology 1982, 89, 35–37. [Google Scholar] [CrossRef] [PubMed]
  215. 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] [PubMed]
  216. Lichter, H.; Loya, N.; Sagie, A.; Cohen, N.; Muzmacher, L.; Yassur, Y.; Weinberger, D. Keratoconus and mitral valve prolapse. Am. J. Ophthalmol. 2000, 129, 667–668. [Google Scholar] [CrossRef]
  217. Kalkan Akcay, E.; Akcay, M.; Uysal, B.S.; Kosekahya, P.; Aslan, A.N.; Caglayan, M.; Koseoglu, C.; Yulek, F.; Cagil, N. Impaired corneal biomechanical properties and the prevalence of keratoconus in mitral valve prolapse. J. Ophthalmol. 2014, 2014, 402193. [Google Scholar] [CrossRef]
  218. Rabbanikhah, Z.; Javadi, M.A.; Rostami, P.; Aghdaie, A.; Yaseri, M.; Yahyapour, F.; Katibeh, M. Association between acute corneal hydrops in patients with keratoconus and mitral valve prolapse. Cornea 2011, 30, 154–157. [Google Scholar] [CrossRef]
  219. Chang, Y.S.; Tai, M.C.; Weng, S.F.; Wang, J.J.; Tseng, S.H.; Jan, R.L. Risk of Mitral Valve Prolapse in Patients with Keratoconus in Taiwan: A Population-Based Cohort Study. Int. J. Environ. Res. Public Health 2020, 17, 6049. [Google Scholar] [CrossRef]
  220. Ritelli, M.; Colombi, M. Molecular Genetics and Pathogenesis of Ehlers-Danlos Syndrome and Related Connective Tissue Disorders. Genes 2020, 11, 547. [Google Scholar] [CrossRef]
  221. Woodward, E.G.; Morris, M.T. Joint hypermobility in keratoconus. Ophthalmic Physiol. Opt. 1990, 10, 360–362. [Google Scholar] [CrossRef]
  222. Fransen, E.; Valgaeren, H.; Janssens, K.; Sommen, M.; De Ridder, R.; Vandeweyer, G.; Bisceglia, L.; Soler, V.; Hoischen, A.; Mortier, G.; et al. Resequencing of candidate genes for Keratoconus reveals a role for Ehlers-Danlos Syndrome genes. Eur. J. Hum. Genet. 2021, 29, 1745–1755. [Google Scholar] [CrossRef]
  223. Maumenee, I.H. The eye in the Marfan syndrome. Trans. Am. Ophthalmol. Soc. 1981, 79, 684–733. [Google Scholar] [PubMed]
  224. Mashor, R.S.; Kumar, N.L.; Ritenour, R.J.; Rootman, D.S. Keratoconus caused by eye rubbing in patients with Tourette Syndrome. Can. J. Ophthalmol. 2011, 46, 83–86. [Google Scholar] [CrossRef]
  225. Knutsson, K.A.; Paganoni, G.; Ambrosio, O.; Ferrari, G.; Rama, P. Corneal collagen cross-linking for management of keratoconus in patients affected by Tourette syndrome. Eur. J. Ophthalmol. 2021, 31, 2233–2236. [Google Scholar] [CrossRef] [PubMed]
  226. Safir, M.; Hecht, I.; Heller, D.; Pras, E.; Lifshitz, M.; Einan-Lifshitz, A. Psychiatric Comorbidities Associated with Keratoconus. JAMA Ophthalmol. 2023, 141, 1145–1150. [Google Scholar] [CrossRef] [PubMed]
  227. Alfardan, F.; Alsanad, M.H.; Altoub, H.A. Prevalence of Psychiatric Illness Among Keratoconus Patients. Cureus 2023, 15, e42141. [Google Scholar] [CrossRef]
  228. Moschos, M.M.; Gouliopoulos, N.S.; Kalogeropoulos, C.; Androudi, S.; Kitsos, G.; Ladas, D.; Tsatsos, M.; Chatziralli, I. Psychological Aspects and Depression in Patients with Symptomatic Keratoconus. J. Ophthalmol. 2018, 2018, 7314308. [Google Scholar] [CrossRef]
  229. Moshfeghinia, R.; Arman, A.; Sobhi, N.; Mahmoudinezhad, G.; Molavi Vardanjani, H. Depression among keratoconus patients: A systematic review and meta-analysis. Front. Public Health. 2024, 12, 1477411. [Google Scholar] [CrossRef]
  230. Schlötzer Schrehardt, U.M.; Koca, M.R.; Naumann, G.O.H.; Volkholz, H. Pseudoexfoliation syndrome. Ocular manifestation of a systemic disorder? Arch. Ophthalmol. 1992, 110, 1752–1756. [Google Scholar] [CrossRef]
Figure 1. Summary of possible systemic alterations predisposing to keratoconus development. These factors may act independently or, as in the case of systemic inflammation, they may influence one another and work synergistically with local factors (such as eye rubbing), leading to the ectatic phenotype.
Figure 1. Summary of possible systemic alterations predisposing to keratoconus development. These factors may act independently or, as in the case of systemic inflammation, they may influence one another and work synergistically with local factors (such as eye rubbing), leading to the ectatic phenotype.
Jcm 14 04587 g001
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Pederzolli, M.; Procopio, F.; Tombolini, B.; Marra, S.; De Micheli, M.; Bandello, F.; Ferrari, G. Keratoconus: The Local Manifestation of a Systemic Disease? J. Clin. Med. 2025, 14, 4587. https://doi.org/10.3390/jcm14134587

AMA Style

Pederzolli M, Procopio F, Tombolini B, Marra S, De Micheli M, Bandello F, Ferrari G. Keratoconus: The Local Manifestation of a Systemic Disease? Journal of Clinical Medicine. 2025; 14(13):4587. https://doi.org/10.3390/jcm14134587

Chicago/Turabian Style

Pederzolli, Matteo, Federico Procopio, Beatrice Tombolini, Simone Marra, Massimo De Micheli, Francesco Bandello, and Giulio Ferrari. 2025. "Keratoconus: The Local Manifestation of a Systemic Disease?" Journal of Clinical Medicine 14, no. 13: 4587. https://doi.org/10.3390/jcm14134587

APA Style

Pederzolli, M., Procopio, F., Tombolini, B., Marra, S., De Micheli, M., Bandello, F., & Ferrari, G. (2025). Keratoconus: The Local Manifestation of a Systemic Disease? Journal of Clinical Medicine, 14(13), 4587. https://doi.org/10.3390/jcm14134587

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