Skip to main content
Advertisement

Main menu

  • Home
  • Articles
    • Current Issue
    • Fast Forward
    • Latest Articles
    • Special Sections
    • Archive
  • Information
    • Instructions to Authors
    • Submit a Manuscript
    • FAQs
    • For Subscribers
    • Terms & Conditions of Use
    • Permissions
  • Editorial Board
  • Alerts
    • Alerts
    • RSS Feeds
  • Virtual Issues
  • Feedback
  • Submit
  • Other Publications
    • Drug Metabolism and Disposition
    • Journal of Pharmacology and Experimental Therapeutics
    • Molecular Pharmacology
    • Pharmacological Reviews
    • Pharmacology Research & Perspectives
    • ASPET

User menu

  • My alerts
  • Log in
  • My Cart

Search

  • Advanced search
Drug Metabolism & Disposition
  • Other Publications
    • Drug Metabolism and Disposition
    • Journal of Pharmacology and Experimental Therapeutics
    • Molecular Pharmacology
    • Pharmacological Reviews
    • Pharmacology Research & Perspectives
    • ASPET
  • My alerts
  • Log in
  • My Cart
Drug Metabolism & Disposition

Advanced Search

  • Home
  • Articles
    • Current Issue
    • Fast Forward
    • Latest Articles
    • Special Sections
    • Archive
  • Information
    • Instructions to Authors
    • Submit a Manuscript
    • FAQs
    • For Subscribers
    • Terms & Conditions of Use
    • Permissions
  • Editorial Board
  • Alerts
    • Alerts
    • RSS Feeds
  • Virtual Issues
  • Feedback
  • Submit
  • Visit dmd on Facebook
  • Follow dmd on Twitter
  • Follow ASPET on LinkedIn
Review Article50th Anniversary Celebration Collection Special Section on New and Emerging Areas and Technologies in Drug Metabolism and Disposition, Part I—Minireview
Open Access

Challenges and Opportunities in P450 Research on the Eye

Irina A. Pikuleva
Drug Metabolism and Disposition October 2023, 51 (10) 1295-1307; DOI: https://doi.org/10.1124/dmd.122.001072
Irina A. Pikuleva
Department of Ophthalmology and Visual Sciences, Case Western Reserve University, Cleveland, Ohio
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • ORCID record for Irina A. Pikuleva
  • Article
  • Figures & Data
  • Info & Metrics
  • eLetters
  • PDF
Loading

Abstract

Of the 57 cytochrome P450 enzymes found in humans, at least 30 have ocular tissues as an expression site. Yet knowledge of the roles of these P450s in the eye is limited, in part because only very few P450 laboratories expanded their research interests to studies of the eye. Hence the goal of this review is to bring attention of the P450 community to the eye and encourage more ocular studies. This review is also intended to be educational for eye researchers and encourage their collaborations with P450 experts. The review starts with a description of the eye, a fascinating sensory organ, and is followed by sections on ocular P450 localizations, specifics of drug delivery to the eye, and individual P450s, which are grouped and presented based on their substrate preferences. In sections describing individual P450s, available eye-relevant information is summarized and concluded by the suggestions on the opportunities in ocular studies of the discussed enzymes. Potential challenges are addressed as well. The conclusion section outlines several practical suggestions on how to initiate eye-related research.

SIGNIFICANCE STATEMENT This review focuses on the cytochrome P450 enzymes in the eye to encourage their ocular investigations and collaborations between P450 and eye researchers.

Introduction

What people see is responsible for approximately 80% of what they learn as well as what they remember (Peterson, 2019). Currently, about 36 million people around the world are blind, another 216.6 million have moderate to severe visual impairment, and 188.5 million have mild visual impairment (Bourne et al., 2017). Blindness is the most feared condition in the US adult population, which ranks losing eyesight as potentially having the greatest effect on their day-to-day life than loss of limb, memory, hearing, or speech (Scott et al., 2016). Yet the role of different cytochrome P450 enzymes (P450s or CYPs) in the structure and function of the eye under normal and pathologic conditions is not yet well understood, in part because only very few P450 laboratories have expanded their research interests to studies of the eye. Other reasons include the complexity of the eye (Fig. 1), the need to learn additional eye-specific in vivo and in vitro characterizations, the cost and difficulty of acquisition of fresh human eyes/eye tissues through eye banks, and the very small size of the eyes of a mouse, the most common laboratory species, thus requiring a large animal colony.

Fig. 1.
  • Download figure
  • Open in new tab
  • Download powerpoint
Fig. 1.

Human eye anatomy. Each eyeball is an oblate spheroid with the mean perimeter of 75 mm and the diameters varying from 24 mm (anterio-posterio) to 23.5 mm (horizontal) and 23 mm (vertical diameter). The average eyeball weighs 7 g and has a volume of 6.5 ml. The eyeball has two segments, anterior (or front) and posterior (or back), which are fused in the middle and communicate through the pupil. The anterior segment includes (from anterior to posterior): cornea, anterior chamber (filled with ∼0.25 ml of the aqueous humor), iris-ciliary body, and lens. The posterior chamber is formed by the vitreous humor (a gel-like material, which fills the space behind the lens), retina, choroid, and optic disc. The eye has three coats or tunics: outer (fibrous), middle (vascular), and inner (nervous). The fibrous coat protects intraocular content and is comprised of the transparent anterior part (cornea) and the opaque posterior part (sclera). The vascular coat supplies nutrition to the various structures of the eyeball and is composed of the iris-ciliary body in the anterior segment and choroid in the posterior segment. The nervous coat (retina) is a sensory tissue in the posterior segment, which receives visual stimuli and initiates the visual process by relaying visual sensations to the to the optic nerve and then to the brain (occipital cortex) (Khurana et al., 2015). This figure was licensed from Carlson Stock Art and is used with permission.

Human eye, the second most powerful and complex organ in the human body after the brain, is composed of >2 million operational parts, including >107 million cells (Peterson, 2019). Hence, studies of P450s in the eye are always a challenge for non-eye researchers. Nevertheless, they are possible, as exemplified by the author of this article who started as a biochemist conducting structure and functional studies on purified cholesterol-metabolizing P450s in vitro and then extended her research expertise to in vivo investigations of cholesterol-metabolizing P450s in the brain and retina.

It has been some time since a publication of the excellent reviews on the P450s in eye (Duvvuri et al., 2003, 2004; Choudhary et al., 2006; Nakano et al., 2014). Therefore, this review provides an updated summary of the P450-releated research in the eye and indicates existing opportunities and challenges in ocular studies of P450s. Perhaps not all the opportunities are mentioned, only several, which reflect the expertise of this article author.

Expression of Different P450s in the Eye

Currently, cytochromes P450 are represented by more than 300,000 members found in all the biologic phyla so far examined (Nelson, 2018). These enzymes are typically monooxygenases, i.e., they activate molecular oxygen and incorporate one of its atoms in the substrate while reducing the other atom to water. Carbon hydroxylation is a very common P450 reaction. Other common reactions include heteroatom oxygenation, heteroatom release (dealkylation), and epoxidation. The less common reactions are reductions, desaturation, ester cleavage, and others (Guengerich, 2001). A summary of the ocular P450 localizations in different species is provided in Table 1, which, however, does not encompass all P450s that can be present in the eye. This is because several enzymes (e.g., CYP2U1, CYP21A2, and CYP51) have not yet been specifically investigated for ocular location; nevertheless, their ocular importance is suggested by the clinical manifestations of the human gene variants or a phenotype of knockout mice (Xie et al., 2000; Incorvaia et al., 2003; Leonardi et al., 2016; Huang et al., 2019; El Matri et al., 2021; Berry et al., 2022; Gong et al., 2022; Zenteno et al., 2022). Accordingly, it appears that of the 57 P450s found in humans (Nelson et al., 2004), 30 have the eye as an expression site. Ten of these P450s (1A1, 1A2, 2A6, 2B6, 2C9, 2C19, 2D6, 2E1, 3A4, and 3A5) participate in xenobiotic/drug metabolism in extraocular organs; nine P450s (1B1, 11A1, 17A1, 19A1, 21A2, 27A1, 39A1, 46A1, and CYP51) are involved in metabolism of sterols; six P450s are the fatty acid hydroxylases (2C8, 2J2, 2U1, 4A, 4B1, and 4V2), four P450s (26A1, 26B1, 26C1, and 27C1) metabolize vitamin A; and one P450 (4F8) acts on eicosanoids (Guengerich, 2005). The provided P450 grouping (based on the major P450 substrate) is not ideal, as some P450s metabolize more than one major substrate (Guengerich, 2005). Nevertheless, this classification is common and is used in the following sections.

View this table:
  • View inline
  • View popup
TABLE 1

Distribution of P450s in the eye

Drug Delivery to the Eye

The unique structure of the eye and its partial exposure to the external environment determine distinctive routes of drug delivery to the eye, which include topical applications, periocular injections, and intraocular injections (Fig. 2A). In addition, drugs can enter the eye from the systemic circulation after oral intake, subcutaneous, intramuscular, or intravenous injections (Khurana et al., 2015). Different anatomic barriers restrict ocular drug delivery (Fig. 2B). The tear film, cornea, conjunctiva, and sclera limit drug penetration to the anterior segment, whereas the blood-ocular barriers (blood-aqueous barrier and blood-retinal barrier) restrict drug access to the posterior segment. Topical and systemic administrations are the least invasive routes for delivering medications to the eye. The former are also the most commonly employed mode of ocular drug administration with >90% of the marketed ophthalmic formulations being in the form of eye drops (Gaudana et al., 2009). Topical drugs largely penetrate through the cornea and, by avoiding first-pass hepatic metabolism, reduce the need for higher dosing associated with oral administration. However, only 1% to 7% of the topically administered drugs can reach the aqueous humor due to the tear film, corneal and conjunctival barriers, as well as lacrimation (tear secretion), tear dilution, reflex blinking, and nasolacrimal drainage, the latter accounting for 80% to 90% of drug elimination (Janagam et al., 2017; Arturo et al., 2019). In the case of a systemic administration, only 1% to 2% of the drug reaches the vitreous cavity (Gaudana et al., 2009).

Fig. 2.
  • Download figure
  • Open in new tab
  • Download powerpoint
Fig. 2.

Routes and anatomic barriers for ocular drug delivery. (A) Unique routes of drug delivery to the eye include topical applications, periocular injections, and intraocular injections. Topical applications are administered in the form of eyedrops, ointments, gels, or ocuserts (membranes) or via soft contact lenses. Periocular injections include subconjunctival, subtenon, retrobulbar, and peribulbar injections. Intraocular injections are represented by intracameral injections (into the anterior chamber) and intravitreal injections (into the vitreous cavity). (Khurana et al., 2015). (B) The tear film, cornea, conjunctiva, sclera, BAB, and BRB represent the anatomic barriers for drug delivery to the eye. The tear film is composed of the three layers, including the external lipid layer, which covers the water layer and mucin layer. The lipid layer of the tear film limits access of aqueous formulations to the cornea. In addition, the aqueous phase under the lipid layer is rich in enzymes, proteins, and mucins that can inactivate drugs by protein binding or enzymatic degradation, thus reducing their bioavailability. A high turnover rate of the tear film dilutes topical drugs, and the blink reflux washes them away within 15 to 30 seconds after instillation (Arturo et al., 2019; Agarwal et al., 2021). The cornea also has three major layers: epithelium, stroma, and endothelium. The corneal epithelium acts as a physical barrier to hydrophilic drugs, and the corneal stroma is a barrier to lipophilic drugs (Gaudana et al., 2010; Janagam et al., 2017). Overall, the cornea favors the absorption of lipophilic over hydrophilic drugs (Farkouh et al., 2016). The conjunctiva is a thin mucous membrane consisting of the conjunctival epithelium and an underlying vascularized connective tissue. The conjunctival epithelium is relatively leaky and hydrophilic, with intercellular spaces approximately 230-fold larger than those in the cornea, rendering it permeable even to large biomolecules, such as proteins and peptides. The conjunctiva is more permeable than the cornea for hydrophilic drugs and acts as a dynamic barrier to hydrophobic drugs (Dhahir et al., 2021). The sclera (the white of the eye) consists of collagen and elastin chains that create a fiber matrix, where the pore diameter and intracellular spaces determine the drug flow with smaller molecules having a better permeability (Arturo et al., 2019). Positively charged molecules have poor permeability through the sclera, presumably due to their binding to the negatively charged proteoglycan matrix (Gaudana et al., 2010). Both the sclera and conjunctiva are richly perfused with blood vessels, hence a large fraction of drugs absorbed via these routes may be lost to systemic circulation (Agarwal et al., 2021). As compared with the corneal route, the conjunctiva and sclera are considered to be the minor pathways for drug delivery (Dosmar et al., 2022). The blood-ocular barriers, BAB and BRB, restrict drug entry from the systemic circulation. The BAB is formed by the capillary endothelium in the iris and the ciliary epithelium, which both contain tight junctions. The BRB is formed by the retinal pigment epithelium (outer BRB) and the endothelial membrane of the retinal blood vessels (inner BRB), which also contain tight junctions (Janagam et al., 2017). The BAB is relatively inefficient as compared with the BRB, and small molecules can cross it by permeation through fenestrated capillaries in the ciliary processes (Janagam et al., 2017). The BRB limits drug passage more efficiently than BAB with small lipophilic molecules crossing the BRB comparatively better than hydrophilic or large molecular weight compounds (Ako-Adounvo and Karla, 2018). All panels for this figure were licensed from Carlson Stock Art and are used with permission. BAB, blood-aqueous barrier; BRB, blood-retinal barrier.

Periocular injections (subconjunctival, subtenon, retrobulbar, and peribulbar) represent a deposition of therapeutic agents into the areas adjacent to or surrounding the eye. Periocular injections are used less frequently than topical instillations and are more invasive. Nevertheless, they are still relatively safe for delivering drugs that cannot penetrate the cornea but can easily pass through the sclera. Periocular injections take advantage of the large sclera area for drug absorption (95% of the surface of the eyeball) and are partly used to overcome the inefficiency of topical and systemic dosing to deliver therapeutic drug concentrations to the posterior segment of the eyes. Periocular injections are commonly used to administer local anesthetics and corticosteroids (Ako-Adounvo and Karla, 2018) with subtenon injections resulting in the highest and more sustained vitreous concentration of drug molecules compared to other periocular injections (Arturo et al., 2019).

Intraocular injections and implants are used only in certain cases to deliver the maximum drug concentration to the target tissue. Intraocular injections include intracameral injections (into the anterior chamber) and intravitreal injections (into the vitreous cavity). Intravitreal drug implants are inserted in the vitreous cavity for sustained and slow release. Intravitreal injections are an invasive route of drug administration with adverse events including endophthalmitis, retinal detachment, intraocular hemorrhage, and particulate contamination. Additionally, the need for frequent administration results in a significant treatment burden to patients, a high-volume burden on providers, and an increased cumulative risk of adverse events. Nevertheless, because of the therapeutic benefits, intravitreal injections are the preferred route for ocular delivery to the retina, specifically of the antivascular endothelial growth factor agents, corticosteroids, and some of the antibiotics (Ako-Adounvo and Karla, 2018; Arturo et al., 2019).

Drug-Metabolizing P450s

At least 10 human drug-metabolizing P450s (1A1, 1A2, 2A6, 2B6, 2C9, 2C19, 2D6, 2E1, 3A4, and 3A5) seem to be expressed in the anterior part of the eye (Table 1), which is affected by administration of ocular topical mediations, the most common marketed ophthalmic formulations (Gaudana et al., 2009). Notably, some of these topical medications (substrates, inducers, or inhibitors of P450s) are known to elicit both ocular and systemic effects (Syed et al., 2021). For example, among the drugs for glaucoma treatment, a major cause of legal blindness (Quigley and Broman, 2006), these are timolol metabolized by CYP2D6; betaxolol, a substrate for CYP1A2 and CYP2D6; dorzolamide eliminated by CYP2B1, CYP2C9, CYP2E1, and CYP3A2; and pilocarpine, which inhibits CYP2A6, CYP2A13, and CYP2E1 (Kimonen et al., 1995). Particular caution is recommended when ophthalmic timolol or betaxolol are coadministered with paroxetine or other strong CYP2D6 inhibitors (Farkouh et al., 2016; Vaajanen and Vapaatalo, 2017).

Some of the systemically administered drugs also elicit serious ocular effects in the anterior and/or posterior parts of the eye (Syed et al., 2021). These include the antimalarial hydroxychloroquine and chloroquine (inhibit CYP2D6 and are metabolized by CYP2C8 and CYP3A4/5) (Rendic and Guengerich, 2020); the antiarrhythmic amiodarone (inhibits CYP1A2, CYP2C9, CYP2D6, and CYP3A4 and is metabolized by CYP3A4) (McDonald et al., 2015); the antiarrhythmic propranolol (metabolized by CYP1A2 and CYP2D6) (Johnson et al., 2000); the antiarrhythmic quinidine (inhibits CYP2D6 and is metabolized by CYP3A4) (Guengerich et al., 1986; Branch et al., 2000); the antidepressant fluoxetine (both an inhibitor and a substrate of CYP2D6) (Lynch and Price, 2007); the antidepressant fluvoxamine (inhibits CYP1A2) (Brøsen et al., 1993); cholesterol-lowering simvastatin, lovastatin, and atorvastatin (metabolized by CYP3A4) (Bellosta et al., 2004); the antituberculosis ethambutol (inhibits CYP1A2 and CYP2E1) (Lee et al., 2014); and the anticancer tamoxifen (metabolized by CYP2D6, CYP3A4, CYP2B6, and CYP2C19) (Singh et al., 2011).

A list of drugs with ocular side effects is much more extensive than that indicated here. Therefore, readers are referred to several review and experimental articles to learn more either about these drugs or specific manifestations of their ocular toxicity (Shichi and Nebert, 1982; Siu et al., 2008; Farkouh et al., 2016; Novack and Robin, 2016; Vaajanen and Vapaatalo, 2017; Morawski et al., 2020; Syed et al., 2021; Souza Monteiro de Araújo et al., 2022). The provided examples are meant to illustrate the importance of studies in the eye of drug pharmacokinetics and pharmacodynamics as well as drug–drug interactions and genetic differences in ocular drug metabolism. These areas of research represent both a challenge and an opportunity for the P450 community. A challenge is in the unique anatomy and complexity of the eye and the difficulty of obtaining samples of ocular tissues for drug quantifications. An opportunity is the multiple routes of drug administration to the eye, which can be used to improve ocular safety of the marketed drugs.

Sterol-Metabolizing P450s

CYP1B1

Of the nine sterol-metabolizing P450s that seem to be present in the eye (1B1, 11A1, 17A1, 19A1, 21A2, 27A1, 39A1, 46A1, and CYP51), CYP1B1 stands apart from the remaining eight enzymes (the classic endobiotic-metabolizing P450s) because it can use both endogenous and exogenous substrates. The former include 17β-estradiol, arachidonic acid, vitamin A, and melatonin (Jansson et al., 2001; Choudhary et al., 2004; Ma et al., 2005; Choudhary et al., 2006, 2008; Nakano et al., 2014). The latter are exemplified by procarcinogens, namely, polycyclic aromatic hydrocarbons, heterocyclic amines, and aromatic amines (Guengerich, 2005)]. In addition, CYP1B1 is perhaps one of the most well-known P450s among eye researchers as, in 1997, CYP1B1 was identified as a causative gene for primary congenital glaucoma (PCG), the major glaucoma type in the pediatric population. Later, CYP1B1 was found to be a modifier gene for primary open-angle glaucoma (POAG), the major glaucoma type in the adult population (Stoilov et al., 1997; Quigley and Broman, 2006; Vasiliou and Gonzalez, 2008; Aponte et al., 2010). Now more than 150 CYP1B1 mutations have been found in subjects with PCG (Rauf et al., 2016), and, of them, approximately two-thirds are missense mutations (Li et al., 2011). Of the latter, 23 of the most frequent CYP1B1 mutations were studied for the effect on enzyme activity in a cell-based system, and most variants were established to have reduced metabolism of 17β-estradiol and absent or increased metabolism of retinol (Banerjee et al., 2016). Intriguingly, the CYP1B1 mutation penetrance is variable in the human population (Bejjani et al., 1998, 2000), and it is currently unknown why.

Strong CYP1B1 expression in the fetal human and mouse eyes suggested the enzyme role in ocular development and function (Hakkola et al., 1997; Stoilov et al., 1997). Hence, CYP1B1 was proposed to mediate the metabolism of endogenous and exogenous compounds that are important for eye development (Stoilov et al., 1997; Choudhary et al., 2006; Vasiliou and Gonzalez, 2008). Also, CYP1B1 was suggested to coordinate the expression of some important genes relevant to the anterior chamber formation (Stoilov, 2001; Stoilov et al., 2001). These proposed roles of CYP1B1 were supported by the characterizations of Cyp1b1−/− mice, which were found to have developmental abnormalities similar to human PCG (Libby et al., 2003; Zhao et al., 2013; Teixeira et al., 2015). Recent studies also pointed to the CYP1B1 roles in the regulation of ocular iron homeostasis, oxidative stress, expression of the peroxisome proliferator-activated receptor (PPAR) γ target genes, and retinal neovascularization (Falero-Perez et al., 2018, 2019a, b; Song et al., 2022). Nevertheless, despite all these investigations, the key processes affected by CYP1B1 activity in the anterior and posterior parts of the eye and precise mechanisms by which CYP1B1 mutations underlie the development of PCG are currently not well understood. Accordingly, important areas in CYP1B1 research include uncovering CYP1B1 ocular significance and identification of both endogenous and exogenous CYP1B1 substrates that pertain to the PCG development. Both areas represent an opportunity for P450 investigators and simultaneously are a challenge as highlighted by the fact that these areas have remained a priority in the CYP1B1 ocular research for 25 years.

Steroidogenic P450s

CYPs 11A1, 17A1, 19A1, and 21A2 represent steroidogenic P450s (Fig. 3) as they catalyze the key steps in the production of different steroid hormones, namely glucocorticoids, mineralocorticoids, and sex hormones (androgens and estrogens) (Miller and Auchus, 2011). CYP11A1 converts cholesterol to pregnenolone, the precursor of all steroid hormones. Pregnenolone and its metabolite progesterone then could be hydroxylated by CYP17A1 to yield 17α-hydroxypregnenolone and 17α-hydroxyprogesterone, respectively, steroids that can be further metabolized by CYP17A1 to the androgens dehydroepiandrosterone and androstenedione, respectively. Dehydroepiandrosterone and androstenedione are precursors of the primary sex hormone testosterone, which can be converted to another primary sex hormone estradiol via the action of CYP19A1 that catalyzes the aromatization of androstenedione and testosterone. 17α-Hydroxyprogesterone produced by CYP17A1 can also serve as a substrate for CYP21A2 to ultimately generate the primary glucocorticoid cortisol. Progesterone, which is formed from pregnenolone, is another substrate for CYP21A2 and a precursor for aldosterone, the primary mineralocorticoid.

Fig. 3.
  • Download figure
  • Open in new tab
  • Download powerpoint
Fig. 3.

Sterol-metabolizing P450s expressed or implicated to be expressed in the eye. The P450 (in bold) grouping is by the biologic process (in bold italics). Not all the steps in the indicated three biologic processes are shown—only those involving the discussed P450 enzymes. Two or three arrows indicate multiple enzymatic reactions. The primary steroid hormones are in bold and underlined. See text for details.

The indicated P450-mediated reactions outline the principal pathways of steroidogenesis. Yet which of these pathways is operative and which types of steroid hormones are predominantly produced depends on a steroidogenic tissue and the cell type (Miller and Auchus, 2011). For example, corticosteroids (mineralocorticoids and glucocorticoids) are typically synthesized by the adrenal glands (in the zona glomerulosa and zona fasciculata, respectively), whereas sex hormones are mainly made in the gonads or placenta with some contribution from the zona reticularis of the adrenals, which produces androgens (Miller and Auchus, 2011). In addition, small amounts of steroid hormones are synthesized in the brain and constitute a local regulatory mechanism for different brain processes (Corpéchot et al., 1981; Baulieu, 1997).

Steroid hormones exert a significant influence on the health and well-being of the eye as receptors for steroid hormones are found in ocular structures in both the anterior (the lacrimal gland, meibomian gland, conjunctiva, goblet cells, cornea, anterior chamber, iris, ciliary body, and lens) and posterior parts of the eye (the vitreous humor and retina) (Sullivan, 2004). Accordingly, steroid hormones influence multiple structural and functional aspects of the eye. These include tissue morphology, epithelial cell dynamics (e.g., proliferation, maturation, transport, and secretion), aqueous tear output, lipid production, mucous secretion, tear film stability, corneal properties (e.g., thickness, curvature, sensitivity, and wetting time), goblet cell density, aqueous humor circulation and outflow, intraocular pressure, lens epithelial cell density, lens permeability, retinal thickness, optic cup area, as well as visual coordination and acuity (Sullivan, 2004; McKay et al., 2022). In addition, steroid hormones (mainly sex hormones) have been linked to the development, progression, and/or treatment of many ocular conditions, including dry eye syndromes, contact lens intolerance, allergic and vernal keratoconjunctivitis, allergic conjunctivitis, corneal angiogenesis, wound healing, transplant rejection, refractive errors, keratitis, myopia, keratoconus, cataracts, glaucoma, amblyopia, photophobia, optic neuritis, papilloedema, age-related macular degeneration (AMD), retinal vascular occlusion, retinal neuron apoptosis, and diabetic retinopathy (Sullivan, 2004).

However, it not clear whether hormonal ocular effects are solely due to the action of the hormones delivered from the systemic circulation or there is also some ocular steroidogenesis either from cholesterol or blood-borne steroid precursors. Several lines of evidence support the notion of local steroidogenesis in different oculars structures: first, detection of steroidogenic P450s in these structures (Table 1); second, capacity of some of these structures (e.g., the retina and cornea) to synthesize steroid hormones either ex vivo or in cultures of cells from these structures (Guarneri et al., 1994, 2003; Susarla et al., 2014; Cascio et al., 2015); and third, ocular manifestations of a steroidogenic P450 deficiency. The latter is exemplified by CYP21A2, which is necessary for mineralocorticoid and glucocorticoid production in the adrenal glands and whose deficiency leads to most cases of congenital adrenal hyperplasia (Miller and Auchus, 2011). Recently several pathogenic variants of CYP21A2 have been linked to autosomal dominant congenital cataracts (Berry et al., 2022), and earlier CYP21A2 was detected in cultures of human lens epithelial cells (Zhang et al., 2013). Collectively, these findings suggested that CYP21A2 could be important for biosynthesis of aldosterone and cortisol in the lens. In addition, CYP21A2 deficiency could be associated with keratoconus, a condition in which the cornea assumes a conical shape due to thinning of the corneal stroma (Incorvaia et al., 2003). Yet there are no reports on CYP21A2 expression in the cornea; rather, primary human corneal epithelial cells were shown to synthesize cortisol from cortisone (Susarla et al., 2014). Thus, establishing the major types and sources of steroid hormones in different ocular structures is an opportunity for P450 researchers, which will help to better control the physiology of ocular tissues and treat various disorders of the eye.

Cholesterol-Metabolizing P450s

In the eye, these include CYP11A1 (discussed in the previous section) as well as CYP27A1 and CYP46A1 that use cholesterol as the endogenous substrate along with CYP39A1 that acts on the cholesterol metabolite produced by CYP46A1 (Fig. 3). CYP27A1 sequentially hydroxylates cholesterol at C27 to yield 27-oxygenated sterols (27-hydroxycholesterol, 5-cholestenoic acid, and 7α-hydroxy-3-oxo-4-cholestenoic acid), whereas CYP46A1 hydroxylates cholesterol at C24 to generate 24-hydroxycholesterol (Wikvall, 1984; Pikuleva et al., 1998; Lund et al., 1999; Meaney et al., 2007). 24-Hydroxycholesterol could then be hydroxylated by CYP39A1 at C7 to produce 7α, 24-dihydroxycholesterol (Li-Hawkins et al., 2000).

So far, CYP27A1 and CYP46A1 have been mainly studied in the retina. This is because cholesterol is the major component of drusen and subretinal drusenoid deposits, two extracellular lesions and notable hallmarks of AMD (Curcio et al., 2005; Wang et al., 2010; Oak et al., 2014), a major cause of legal blindness in the elderly of industrialized countries (Wong et al., 2014). Also, evidence has linked dysregulation of the chorioretinal cholesterol homeostasis and AMD (Pikuleva and Curcio, 2014). Second, CYP27A1 deficiency in humans leads to cerebrotendinous xanthomatosis (Cali et al., 1991), a lipid storage disorder, which has ocular manifestations, including those in the retina (Koyama et al., 2021). These retinal manifestations are premature retinal senescence with drusen and retinal vessel sclerosis, cholesterol-like deposits along the vascular arcades, retinal pigment epithelium (RPE) defects on fluorescein angiography as well as optic disc pallor and neuritis (Cruysberg et al., 1995; Dotti et al., 2001; Miyamoto et al., 2019; Koyama et al., 2021). Third, CYP46A1 may play a role in glaucoma and diabetic retinopathy (Saadane et al., 2019; Zhang et al., 2021). A polymorphism in CYP46A1 was linked to increased risk of POAG (Fourgeux et al., 2009). However, this association was not confirmed in a subsequent study (Mossböck et al., 2011). Despite these conflicting reports, data suggest that 24-hydroxycholesterol could be an endogenous neuroprotectant under glaucomatous conditions (Ishikawa et al., 2016, 2018; Zhang et al., 2021). In addition, CYP46A1 could play a protective role in vascular damage in diabetic retinopathy (Saadane et al., 2019).

CYP27A1 and CYP46A1 have been immunolocalized to the retinal layers and cell types in different species (Lee et al., 2006; Bretillon et al., 2007; Ramirez et al., 2008; Zheng et al., 2012). The retinal abundance of these proteins and retinal levels of their metabolites were quantified by mass spectrometry (Liao et al., 2011; Mast et al., 2011; Wang et al., 2012). Retinal phenotype of Cyp27a1−/−, Cyp46a1−/−, and Cyp27a1−/−Cyp46a1−/− mice was extensively characterized and found to lead to retinal cholesterol accumulation and chorioretinal vascular abnormalities (Omarova et al., 2012; Saadane et al., 2014, 2019). CYP27A1 was discovered to be post-translationally modified in the AMD-affected human retina by isolevuglandins, arachidonate oxidation products, which diminished the enzyme activity (Charvet et al., 2011, 2013a). Importantly, in mice, pretreatment with pyridoxamine, a B6 vitamer and efficient scavenger of gamma-ketoaldehydes, reduced the levels of retinal isolevuglandin adducts and mitigated the isolevuglandin-associated retinal effects in animals exposed to bright light (Charvet et al., 2013b). CYP46A1 was studied as a pharmacologic target for enzyme inhibition and activation (Fourgeux et al., 2014; El-Darzi et al., 2022), and the latter was found to be beneficial in 5XFAD mice, a model of Alzheimer’s disease. Retinal CYP46A1 activation enhanced retinal cholesterol turnover and reduced more than fivefold retinal frequency of vascular lesions associated with deposits within the RPE and subretinal space (El-Darzi et al., 2022).

Despite extensive studies of CYP27A1 and CYP46A1 in the retina, opportunities still exist for expanding our knowledge of ocular significance of these P450s. For example, we still do not know the quantitative contributions of the CYP27A1- and CYP46A1-mediated retinal cholesterol removal to the total retinal cholesterol output or whether these contributions are similar in different species (e.g., mice, hamsters, and humans). Also, Cyp27a1−/− mice do not recapitulate all the features of CYP27A1 deficiency in humans and therefore do not develop cerebrotendinous xanthomatosis (Rosen et al., 1998; Dubrac et al., 2005). Accordingly, we likely underestimate ocular roles of CYP27A1 in humans based on studies of Cyp27a1−/− mice. Another opportunity is to characterize CYP27A1 and CYP46A1 in ocular structures other than the retina where cholesterol is abundant. Indeed, the content of cholesterol in the membranes of human lens is the highest of any known biologic membrane (Li et al., 1985; Cenedella, 1996). Yet little is known how cholesterol homeostasis is maintained in the lens. What is known, however, is that CYP27A1 deficiency in humans leads to juvenile bilateral cataracts (Morgan et al., 1989; Cruysberg et al., 1995; Dotti et al., 2001), and an SNP in CYP46A1 is associated with senile cataracts (Raza et al., 2017), the most common cause of blindness worldwide (Resnikoff et al., 2004). Thus, an opportunity is to delineate the role of lenticular cholesterol metabolism in juvenile and senile cataract formation. Perhaps the cornea and nonpigmented ciliary body epithelium could be studied as well, because CYP27A1 was found to be expressed in the cell lines of human corneal endothelium and nonpigmented ciliary body epithelium (Alsalem et al., 2014).

CYP51

CYP51 is the only P450 involved in the biosynthesis of cholesterol, where it catalyzes lanosterol and dihydrolanosterol 14α-demethylation (Fig. 3) (Debeljak et al., 2003; Lepesheva and Waterman, 2004). In the retina, local cholesterol biosynthesis accounts for the majority of retinal cholesterol input (Lin et al., 2016). Lens and cornea can also synthesize cholesterol (Cenedella, 1982; Hitchener and Cenedella, 1985; Cenedella and Fleschner, 1989), and studies show that the origin of cholesterol in the cornea is of importance. The latter is highlighted by corneal opacification in different genetic diseases (e.g., LCAT and APOA1 deficiencies, Tangier disease, and Schnyder corneal dystrophy) due to cholesterol accumulation (Cenedella and Fleschner, 1989; Cogan et al., 1992; Gaynor et al., 1996; Flores et al., 2019). Conversely, in the lens, reduction in cholesterol content due to inherited defects in the enzymes involved in cholesterol biosynthesis (7-dehydrocholesterol reductase or lanosterol synthase) or use of drugs (lovastatin and simvastatin) that inhibit lens cholesterol biosynthesis can be associated with cataracts in both animals and humans (Cenedella, 1996; Mori et al., 2006; Zhao et al., 2015; Widomska and Subczynski, 2019). Remarkably, the CYP51 substrate lanosterol was found to be a key molecule in the prevention of lens protein aggregation and was suggested to represent a novel strategy for cataract prevention and treatment (Mori et al., 2006; Zhao et al., 2015). Thus, an opportunity for P450 researchers is to study the role of CYP51 in the retina, cornea, and lens as CYP51 should be definitely expressed in these ocular structures. However, currently there does not seem to be any published studies on CYP51 in the eye.

Fatty Acid-Hydroxylating P450s

Currently, the activities of six fatty acid-hydroxylating P450s (2C8, 2J2, 2U1, 4A, 4B1, and 4V2) seem to be of importance for normal and pathologic processes in the eye. These activities are epoxygenation and/or hydroxylation of long-chain polyunsaturated fatty acids (LPUFAs)—ω-6 [arachidonic acid (AA)] and ω-3 [docosahexaenoic acid (DHA) and eicosapentaenoic acid (EPA)] (Fig. 4) as well as ω/ω-1 hydroxylation of short- to medium-chain saturated fatty acids. Many of these activities produce the biologically active metabolites (Fleming, 2014; Ni and Liu, 2021).

Fig. 4.
  • Download figure
  • Open in new tab
  • Download powerpoint
Fig. 4.

Some of the cytochrome P450 metabolites generated from the long-chain polyunsaturated fatty acids. Not all the P450s (in bold), the long-chain polyunsaturated fatty acids (in bold and underlined), and the products of their metabolism are indicated—only those discussed in the text. The non-P450 enzymes are also not indicated. See text for details. EDP, epoxydocosapentaenoic acid; EEQ, epoxyeicosatetraenoic acid; EET, epoxyeicosatrienoic acid; HETE, hydroxyeicosatetraeonic acid; HETrE, hydroxyeicosatrienoic acid.

Specifically, of the P450s from family 2, CYP2C8 can epoxygenate AA, DHA, and EPA at comparable rates to generate 14,15- and 11,12-epoxyeicosatrienoic acids (EETs) from AA; 19,20-, 16,17-, 13,14-, and 10,11-epoxydocosapentaenoic acid (EDPs) from DHA; and 17,18-, 14,15-, and 11,12-epoxyeicosa-tetraenoic acids (EEQs) from EPA with a minor formation of hydroxylated metabolites (Fer et al., 2008; Arnold et al., 2010). In addition, CYP2C8 can metabolize non-fatty acid substrates, which include all-trans-retinol and all-trans-retinoic acid and some drugs (e.g., paclitaxel, troglitazone, verapamil, rosiglitazone, cerivastatin, amiodarone, dapsone, and others) (Leo et al., 1989; Rendic, 2002). However, the contribution of CYP2C8 to the metabolism of clinically relevant drugs is not as significant as of CYP2C9 and other major drug-metabolizing P450s (Johnson et al., 2021). CYP2J2 catalyzes the epoxidation of the all four olefin bonds in AA to produce 14,15-, 11,12-, 8,9- and 5,6-EETs (Wu et al., 1996). However, EPA and DHA are much better substrates for CYP2J2 than AA, which mostly undergo epoxidation to 17,18-EEQ and 19,20-EDP, respectively (Fer et al., 2008; Arnold et al., 2010). CY2U1 catalyzes the ω/ω-1 hydroxylation of both saturated (palmitic and stearic acids but not lauric acid) and unsaturated fatty acids, including AA, EPA, and DHA. 19-Hydroxyeicosatetraeonic acid (HETE), 20-HETE, and either 22- and 21-hydroxyDHAs or 19,20-EDP are some of the identified CY2U1 metabolites (Chuang et al., 2004).

Of the P450s from family 4, the CYP4A isoforms hydroxylate medium-chain saturated fatty acids (e.g., lauric acid) as well as AA at the ω/ω-1 positions to generate 12- and 11-hydroxylauric acids and 20- and 19-HETEs, respectively (Okita and Okita, 2001). The CYP4A isoforms are the predominant fatty acid ω/ω-1 hydroxylases in most mammalian tissues (Capdevila et al., 2000). The substrate preferences of CYP4B1 are broader with the typical endogenous substrates being short- to medium-chain saturated fatty acids, which are ω-hydroxylated, and also AA, which was proposed to be C12-hydroxylated to yield 12-HETE and 12-hydroxy-5,8,14-eicosatrienoic acid (12-HETrE) (Mastyugin et al., 1999; Baer and Rettie, 2006; Nakano et al., 2014). In addition, CYP4B1 bioactivates a range of xenobiotic protoxins that often exert tissue-specific effects (Baer and Rettie, 2006). CYP4V2 was shown to be a selective ω-hydroxylase of the medium-chain saturated fatty acid and also have the ω-hydroxylase activity toward DHA and EPA to produce 19-hydroxyEPA, 20-hydroxyEPA, 21-hydroxyDHA, and 22-hydroxyDHA. Overall, CYP4V2 seems to be an efficient ω-hydroxylase of both saturated and unsaturated fatty acids (Nakano et al., 2009, 2012).

The DHA and EPA metabolites of CYP2C8 (19,20-EDP and 14,15-EEQ) were found to be proangiogenic and induce retinal neovascularization in mice, counteracting in part the overall antiangiogenic effects of DHA and EPA (Shao et al., 2014). Subsequent studies of CYP2C8-overexpressing mice fed the DHA+EPA-enriched diet and treated with montelukast as selective CYP2C8 inhibitor (Walsky et al., 2005b) showed a significant reduction in the plasma levels of the CYP2C8 products and suppression of pathologic angiogenesis in oxygen-induced retinopathy and laser-induced choroidal neovascularization (CNV) (Gong et al., 2016a). Also, fenofibrate, a PPARα agonist and drug that reduces progression of diabetic retinopathy in type 2 diabetes patients independent of its PPARα effects, suppressed retinal and choroidal neovascularization in mice overexpressing CYP2C8 in endothelial cells and reduced plasma levels of the CYP2C8 metabolite 19,20-EDP. As fenofibrate is a modest CYP2C8 inhibitor (Walsky et al., 2005a), this study suggested that the fenofibrate neovascular effects are mediated via both the PPARα agonist activity and CYP2C inhibition (Gong et al., 2016b). The PPARα-mediated effect on pathologic neovascularization was confirmed in a different study in mouse models that recapitulate some of the features of AMD (Qiu et al., 2017).

Conversely, a different group found that 19,20-EDP and 17,18-EEQ suppressed laser-induced CNV in mice (Yanai et al., 2014) and that animals overexpressing CYP2C8 in endothelial cells and fed the DHA+EPA-enriched diet had markedly attenuated neovascular lesions (Hasegawa et al., 2017). Apparently, the individual effects of 19,20-EDP, 14,15-EET, and 17,18-EEQ on ocular neovascularization need to be investigated to resolve the conflicting data on the CYP2C8 ocular significance. In the meantime, studies in cell culture and rat model of oxygen-induced retinopathy suggested that the AA metabolite 11,12-EET may have a proangiogenic effect in the retina (Capozzi et al., 2014).

As compared with CYP2C8, ocular studies of CYP2J2 are not as extensive but are also conflicting. Endothelium-specific overexpression of CYP2J2 in rats was shown to prevent vascular endothelial senescence and thereby attenuate retinal ganglion cell loss induced by retinal ischemia-reperfusion injury (Huang et al., 2019). In a different study, the laser-induced CNV was exacerbated in the CYP2J2-overexpressing mice and was associated with increased plasma levels of 19,20-EDP and 14,15-EET in animals fed the DHA+EPA diet but not the AA-enriched diet. CYP2J2 inhibition in the CYP2J2-overexpressing mice with flunarizine suppressed pathologic choroidal angiogenesis after the laser CNV induction, and cotreatment with montelukast inhibiting CYP2C8 further enhanced the effect. Thus, inhibition of P450s from family 2 was suggested to be a viable approach for suppression of CNV in AMD (Gong et al., 2022).

Attention to the ocular significance of CYP2U1 was brought about by the ocular phenotype of patients with spastic paraplegia 56, an autosomal recessive neurodegeneration characterized by early-onset progressive lower-limb spasticity and weakness due to mutations in CYP2U1 (Tesson et al., 2012). Pathogenic CYP2U mutations were found to cause ocular manifestations with or without neurologic symptoms. The spectrum of these manifestations included macular degeneration associated or not associated with bilateral macular telangiectasia (abnormalities of the macular retinal vasculature), fibrotic CNV, and macular yellowish punctuate deposits in both eyes (Leonardi et al., 2016; El Matri et al., 2021; Zenteno et al., 2022). Therefore, CYP2U1 was suggested to be included in the panels of genes tested for macular dystrophies, especially in the presence of macular telangiectasia and/or neurologic manifestations (El Matri et al., 2021).

There seem to be only one study on ocular CYP4A significance. In rats, inhibition of CYP4A activity with two different inhibitors, N-hydroxy-N’-(4-butyl-2-methylphenyl) formamidine and dibromododecenyl methylsulfonimide, decreased angiogenic response in the cornea. Accordingly, a CYP4A product, possibly 20-HETE, was suggested to play a critical role in the regulation of corneal angiogenesis and serve as a useful target for reduction of pathologic angiogenesis (Chen et al., 2005).

CYP4B1 was investigated for ocular significance in multiple studies and was shown to mediate hypoxia-induced corneal 12-HETE and 12-HETrE production (Mastyugin et al., 1999, 2001, 2004). Also, all-trans and 9-cis retinoic acids were found to increase the CYP4B1 expression and enhance the production of the inflammatory 12-hydroxyeicosanoids in the corneal epithelium (Ashkar et al., 2004). To confirm CYP4B1 involvement in corneal neovascularization, CYP4B1 was transfected into rabbit cornea in vivo, which led to increased corneal 12-HETrE production and neovascularization (Mezentsev et al., 2005). Conversely, in rabbits, subconjunctival injection of CYP4B1 siRNA decreased corneal 12-HETrE production and neovascularization (Seta et al., 2007). Collectively, these data suggested CYP4B1 as a component of the inflammatory and neovascular cascade initiated by the corneal injury and that the CYP4B1-12-HETrE system could be a new therapeutic target for preventing corneal neovascularization (Mezentsev et al., 2005; Seta et al., 2007).

Pleiotropic effects and in particular angiogenesis modulation by the P450 metabolites generated from AA, DHA, and EPA provide P450 researchers with multiple golden opportunities. The most obvious is the identification of the enzymes with ocular significance that can serve as pharmacologic targets to contribute to development of new treatments for the most common causes of vision loss. These are retinopathy of prematurity in premature infants, diabetic retinopathy in working-age adults, AMD in the elderly in developed countries, and corneal neovascularization, which affects people of different ages (Gong et al., 2017; Sharif and Sharif, 2019). Yet studies of the individual contributions of the fatty acid-hydroxylating P450s is a challenge, as the five enzymes discussed so far in this section are not the only fatty acid-hydroxylating P450s in mammals (Capdevila et al., 2000; Okita and Okita, 2001; Fer et al., 2008; Arnold et al., 2010; Ni and Liu, 2021). In fact, there are multiple CYP2 and CYP4 isoforms that share extensive amino acid sequence homology, metabolize LPUFAs to similar products, and often have common immunologic determinants. In addition, not all human genes have mouse or rat orthologs (Nelson et al., 2004; Ni and Liu, 2021), making it difficult to study the function of human genes in animals. In addition, many fatty acid-hydroxylating P450s are induced by commonly used drugs, show sex-based differences in expression, and have polymorphic variants (Guengerich, 2005; Jarrar and Lee, 2019; Ni and Liu, 2021), factors that collectively may lead to significant interindividual variability in P450 ocular significance. Nevertheless, all of these challenges could be overcome, at least in part, by careful planning of experiments and more knowledge in the field about the fatty acid-hydroxylating P450s.

CYP4V2 stands apart from the P450s 2C8, 2J2, 2U1, 4A, and 4B1 as mutations in its gene cause Bietti crystalline dystrophy (BCD), a rare autosomal recessive disease (Lee et al., 2001). Currently more than 100 disease-causing mutations in CYP4V2 have been reported, mostly missense, deletion, insertion, splicing, and nonsense mutations, which are either shown or predicted to lead to the enzyme loss of function (Lee et al., 2001; Nakano et al., 2012; García-García et al., 2019). BCD is manifested by multiple glistening intraretinal crystals (most cases also have similar crystals at the corneal limbus), a characteristic degeneration of the retina, and sclerosis of the choroidal vessels, ultimately leading to progressive night blindness and constriction of the visual field (Li et al., 2004; García-García et al., 2019). The precise chemical composition of the crystals found in patients with BCD is unknown, although studies of human RPE cells generated from patient-induced pluripotent stem cells (iPSCs) provided some insights (Hata et al., 2018). These RPE cells showed the accumulation of glucosylceramide and free cholesterol, and the accumulation of the latter was suggested to cause cell damage and subsequent cell death via the induction of lysosomal dysfunction and impairment of autophagy flux (Hata et al., 2018). In addition, studies of other cell types cultured from patients with BCD (fibroblasts and lymphocytes) demonstrated the absence of two fatty acid-binding proteins, abnormally high triglycerides and cholesterol storage, and reduced conversion of fatty acid precursors into ω-3 LPUFAs (Lee et al., 1998, 2001). Cyp4v3−/− mice (the mouse ortholog of CYP4V2) were generated and found to recapitulate the characteristic features of corneoretinal crystal accumulation and systemic dyslipidemia seen in BCD (Lockhart et al., 2014).

Two treatment approaches for BCD have been investigated so far. In the first, the increased free cholesterol content in the BCD iPSC-RPE cells was reduced by the cyclodextrin or δ-tocopherol treatment, which rescued the BCD phenotypes. These data suggested that local cholesterol metabolism may play a role in the pathogenesis of BCD and that decreasing the intracellular free cholesterol content may have therapeutic efficacy in patients with BCD (Hata et al., 2018). In the second approach, multiple cell lines were evaluated for transduction with the CYP4V2-containing adeno-associated virus as the clinical characteristics of BCD are believed to be ideal for gene therapy through subretinal injections. The best protein expression and enzyme activity were found with the iPSC-RPE cells and the codon optimized CYP4V2, thus supporting the development of CYP4V2 gene therapy for BCD treatment (Wang et al., 2022).

CYP4V2 is expressed ubiquitously in human tissues, including brain, placenta, lung, liver, and kidney (Li et al., 2004; Nakano et al., 2012). Yet the disease phenotype seems to be restricted to the eye. Thus, some of the opportunities in studies of CYP4V2 are to ascertain the exact molecular mechanism(s) underlying the BCD ocular phenotype and to develop gene therapy or pharmacologic treatments for this currently incurable disease.

Vitamin-Metabolizing P450s

In the eye, these P450s are represented by the enzymes (26A1, 26B1, 26C1, and 27C1) that pertain to vitamin A, which is obtained from diet as it cannot be synthesized endogenously (von Lintig et al., 2021). Vitamin A is found at the highest concentrations in the eye (Luo et al., 2006), likely a reflection of its ocular significance. Indeed, vitamin A and its derivatives are essential for eye development as ocular malformations are the most sensitive indicators of fetal vitamin A deficiency and night blindness is an early sign of postnatal deficiency (Wilson et al., 1953). Also, after birth, the vitamin A derivative 11-cis-retinal is required for the visual cycle in the retina of humans and many other species and thus is critical for vision (Choi et al., 2021).

Of the ocular vitamin A-metabolizing P450s, CYPs 26A1, 26B1, and 26C1 are believed to be the main hydroxylases and degrading enzymes of all-trans-retinoic acid, the biologically active derivative of vitamin A1 (all-trans-retinol). In addition, CYPs 26A1, 26B1, and 26C1 can hydroxylate 4(S)- and 4(R)-hydroxy-all-trans-retinoic acid, 4-oxo-all-trans-retinoic acid, 9-cis-retinoic acid, and 13-cis-retinoic acid (Isoherranen and Zhong, 2019). CYP27C1, the forth vitamin A-metabolizing P450, was previously classified as an orphan P450 (a P450 whose endogenous or exogenous substates were unknown at the time of gene identification; Guengerich et al., 2005). However, recently it was shown to convert vitamin A1 into vitamin A2 (3,4-didehydroretinol) in vivo (zebrafish). In vitro, CYP27C1 was found to efficiently metabolize vitamin A1, retinal, and retinoic acid and had the highest catalytic efficiency with vitamin A1 (Enright et al., 2015).

Studies of vitamin A-metabolizing P450s in the eye are currently very limited and include only a few papers. Cyp26a1 and Cyp26c1 were shown to have coordinate expression in mouse retina during eye development; however, the expression of both enzymes became undetectable after postnatal day 14 (Sakai et al., 2004; Luo et al., 2006). In Xenopus laevis (the African clawed frog), CYP26A1 and CYP26B1 were found in both normal and regenerating corneas, and the expression of Cyp26a1, Cyp26b1, and Cyp26c1 was also detected in the lens, where the CYP26 activity was shown to be necessary for lens regeneration (Thomas and Henry, 2014). The ability of CYP27C1 to generate 3,4-didehydroretinol in zebrafish was discovered to underlie the unusual fish visual sensitivity beyond the range of human vision (a so-called red-shifted photosensitivity). This was because 3,4-didehydroretinol is the precursor of 11-cis-3,4-didehydroretinal, the visual chromophore in species with red-shifted photosensitivity as compared with 11-cis-retinal, the visual chromophore in humans and species with the range of human vision (Enright et al., 2015). Thus, an opportunity for P450 researchers is to ascertain the role of CYPs 26A1, 26B1, and 26C1 in the cornea and lens as well as to clarify whether these enzymes are important in the retina postnatally. As for CYP27C1, an opportunity is to establish its preferred endogenous substrates in different species and its ocular significance in humans.

Eicosanoid-Metabolizing P450 4F8

CYP4F8 was identified as the AA hydroxylase that mostly generates 18-HETE as well as a prominent prostaglandin H1 and H2 ω-2 hydroxylase that mostly yields 19(R)-hydroxylated products (Bylund et al., 2000). While originally discovered in the epithelium of human seminal vesicles (Bylund et al., 1999), this P450 was also immunolocalized to the corneal epithelium (Stark et al., 2003). However, the substrate preferences of CYP4F8 in the corneal epithelium and its corneal significance are currently unknown, representing an opportunity for P450 researchers.

Conclusions and Practical Suggestions

P450 expertise is needed in research efforts to prevent and combat blindness, the most feared condition in the US adult population, and numerous opportunities exist in the P450 research on the eye. Thus, a practical question is “How can eye-related research be initiated in my laboratory?” Multiple approaches are possible. If starting from scratch, first insights into the potential P450 function in the eye could be obtained by immunolocalizations of this P450 within the eye and its specific cell types, preferably in both human and mouse ocular tissues to ascertain any interspecies differences. If systemic or cell-specific P450 knockouts are available, these mutant mice can undergo ocular examination by the methods specific to the eye structure, where this P450 is expressed. Some of the challenges in this approach is that ocular examinations are not trivial and usually require a collaboration with an expert in the field. Specific equipment should also be available, which ranges from simple and relatively inexpensive (e.g., a direct ophthalmoscope for examining the eye fundus) to state-of-the-art and expensive (e.g., an ultra-high resolution spectral domain optical coherence tomograph) to assess retinal gross structure. Fortunately, in many cases, expensive and state-of-the-art ophthalmology equipment is accessible via the National Eye Institute-supported P30 Core facilities, which many universities have to facilitate eye-related research. Finally, it is always a possibility to wait until the ocular significance of a P450 is implicated by genetic and clinical studies or other approaches and then start addressing arising or remaining questions. Regardless of the approach, researchers should educate themselves about different ocular tissues, their structure and function, and initiate a collaboration with a basic scientist or a clinician scientist in the ophthalmology department. In addition to their ophthalmic expertise, these researchers can provide referrals to the private foundations and other organizations that support eye research and thus could be a source of funding. Overall, while challenging, it is possible for P450 investigators to study P450s in the eye, either as an independent research direction or as a collaborative study, as exemplified by the papers cited in this review.

Acknowledgments

The author thanks Natalia Mast for help with figure preparation.

Authorship Contributions

Wrote or contributed to the writing of the manuscript: Pikuleva.

Footnotes

    • Received August 14, 2022.
    • Accepted March 6, 2023.
  • Some of the studies described in this manuscript were supported in part by National Institutes of Health National Institute of Aging [Grant R01-AG067552] (to I.A.P.), National Eye Institute [Grants R01-EY018383 and P30-EY011373] (to I.A.P.) and [Grant R01-EY025383] (multiple principal investigators) as well as the unrestricted grant from the Cleveland Eye Bank Foundation.

  • The author does not have an actual or perceived conflict of interest with the contents of this article.

  • ↵*Previous position: Jules and Doris Stein Professor from Research to Prevent Blindness; current position: Carl F. Asseff Professor of Ophthalmology.

  • dx.doi.org/10.1124/dmd.122.001072.

ABBREVIATIONS

24HC
24-hydroxycholesterol
AA
arachidonic acid
AMD
age-related macular degeneration
BCD
Bietti crystalline dystrophy
CNV
choroidal neovascularization
CYP
cytochrome P450
DHA
docosahexaenoic acid
EDP
epoxydocosapentaenoic acid
EET
epoxyeicosatrienoic acid
EEQ
epoxyeicosatetraenoic acid
EPA
eicosapentaenoic acid
HETE
hydroxyeicosatetraeonic acid
HETrE
hydroxyeicosatrienoic acid
LPUFA
long-chain polyunsaturated fatty acid
iPSCs
induced pluripotent stem cells
PCG
primary congenital glaucoma
POAG
primary open-angle glaucoma
PPAR
peroxisome proliferator-activated receptor
RPE
retinal pigment epithelium
  • Copyright © 2023 The Author(s)

This is an open access article distributed under the CC BY Attribution 4.0 International license.

References

  1. ↵
    1. Agarwal P,
    2. Craig JP, and
    3. Rupenthal ID
    (2021) Formulation considerations for the management of dry eye disease. Pharmaceutics 13:207 10.3390/pharmaceutics13020207.
    OpenUrl
  2. ↵
    1. Patel JK,
    2. Sutariya V,
    3. Kanwar JR, and
    4. Pathak YV
    1. Ako-Adounvo A-M and
    2. Karla PK
    (2018) Transscleral drug delivery to retina and posterior segment disease, in Drug Delivery for the Retina and Posterior Segment Disease (Patel JK, Sutariya V, Kanwar JR, and Pathak YV, eds) pp 215–227, Springer International Publishing, Cham, Switzerland.
  3. ↵
    1. Alsalem JA,
    2. Patel D,
    3. Susarla R,
    4. Coca-Prados M,
    5. Bland R,
    6. Walker EA,
    7. Rauz S, and
    8. Wallace GR
    (2014) Characterization of vitamin D production by human ocular barrier cells. Invest Ophthalmol Vis Sci 55:2140–2147.
    OpenUrlAbstract/FREE Full Text
  4. ↵
    1. Aponte EP,
    2. Diehl N, and
    3. Mohney BG
    (2010) Incidence and clinical characteristics of childhood glaucoma: a population-based study. Arch Ophthalmol 128:478–482.
    OpenUrlCrossRefPubMed
  5. ↵
    1. Arnold C,
    2. Markovic M,
    3. Blossey K,
    4. Wallukat G,
    5. Fischer R,
    6. Dechend R,
    7. Konkel A,
    8. von Schacky C,
    9. Luft FC,
    10. Muller DN et al.
    (2010) Arachidonic acid-metabolizing cytochrome P450 enzymes are targets of omega-3 fatty acids. J Biol Chem 285:32720–32733.
    OpenUrlAbstract/FREE Full Text
  6. ↵
    1. Rajeev KT,
    2. Neeraj G,
    3. Rahul S, and
    4. Prakash Singh B
    1. Arturo S,
    2. Altamirano-Vallejo JC,
    3. Navarro-Partida J, and
    4. González-De la Rosa J
    (2019) Breaking down the barrier: topical liposomes as nanocarriers for drug delivery into the posterior segment of the eyeball, in Role of Novel Drug Delivery Vehicles in Nanobiomedicine (Rajeev KT, Neeraj G, Rahul S, and Prakash Singh B eds), IntechOpen, Rijeka, Croatia.
  7. ↵
    1. Ashkar S,
    2. Mesentsev A,
    3. Zhang WX,
    4. Mastyugin V,
    5. Dunn MW, and
    6. Laniado-Schwartzman M
    (2004) Retinoic acid induces corneal epithelial CYP4B1 gene expression and stimulates the synthesis of inflammatory 12-hydroxyeicosanoids. J Ocul Pharmacol Ther 20:65–74.
    OpenUrlPubMed
  8. ↵
    1. Baer BR and
    2. Rettie AE
    (2006) CYP4B1: an enigmatic P450 at the interface between xenobiotic and endobiotic metabolism. Drug Metab Rev 38:451–476.
    OpenUrlCrossRefPubMed
  9. ↵
    1. Banerjee A,
    2. Chakraborty S,
    3. Chakraborty A,
    4. Chakrabarti S, and
    5. Ray K
    (2016) Functional and structural analyses of CYP1B1 variants linked to congenital and adult-onset glaucoma to investigate the molecular basis of these diseases. PLoS One 11:e0156252.
    OpenUrl
  10. ↵
    1. Baulieu EE
    (1997) Neurosteroids: of the nervous system, by the nervous system, for the nervous system. Recent Prog Horm Res 52:1–32.
    OpenUrlPubMed
  11. ↵
    1. Bejjani BA,
    2. Lewis RA,
    3. Tomey KF,
    4. Anderson KL,
    5. Dueker DK,
    6. Jabak M,
    7. Astle WF,
    8. Otterud B,
    9. Leppert M, and
    10. Lupski JR
    (1998) Mutations in CYP1B1, the gene for cytochrome P4501B1, are the predominant cause of primary congenital glaucoma in Saudi Arabia. Am J Hum Genet 62:325–333.
    OpenUrlCrossRefPubMed
  12. ↵
    1. Bejjani BA,
    2. Stockton DW,
    3. Lewis RA,
    4. Tomey KF,
    5. Dueker DK,
    6. Jabak M,
    7. Astle WF, and
    8. Lupski JR
    (2000) Multiple CYP1B1 mutations and incomplete penetrance in an inbred population segregating primary congenital glaucoma suggest frequent de novo events and a dominant modifier locus. Hum Mol Genet 9:367–374.
    OpenUrlCrossRefPubMed
  13. ↵
    1. Bellosta S,
    2. Paoletti R, and
    3. Corsini A
    (2004) Safety of statins: focus on clinical pharmacokinetics and drug interactions. Circulation 109(23, Suppl 1):III50–III57.
    OpenUrlCrossRefPubMed
  14. ↵
    1. Berry V,
    2. Pontikos N,
    3. Ionides A,
    4. Kalitzeos A,
    5. Quinlan RA, and
    6. Michaelides M
    (2022) Pathogenic variants in the CYP21A2 gene cause isolated autosomal dominant congenital posterior polar cataracts. Ophthalmic Genet 43:218–223.
    OpenUrl
  15. ↵
    1. Bourne RRA,
    2. Flaxman SR,
    3. Braithwaite T,
    4. Cicinelli MV,
    5. Das A,
    6. Jonas JB,
    7. Keeffe J,
    8. Kempen JH,
    9. Leasher J,
    10. Limburg H et al
    ; Vision Loss Expert Group (2017) Magnitude, temporal trends, and projections of the global prevalence of blindness and distance and near vision impairment: a systematic review and meta-analysis. Lancet Glob Health 5:e888–e897.
    OpenUrlPubMed
  16. ↵
    1. Branch RA,
    2. Adedoyin A,
    3. Frye RF,
    4. Wilson JW, and
    5. Romkes M
    (2000) In vivo modulation of CYP enzymes by quinidine and rifampin. Clin Pharmacol Ther 68:401–411.
    OpenUrlCrossRefPubMed
  17. ↵
    1. Bretillon L,
    2. Diczfalusy U,
    3. Björkhem I,
    4. Maire MA,
    5. Martine L,
    6. Joffre C,
    7. Acar N,
    8. Bron A, and
    9. Creuzot-Garcher C
    (2007) Cholesterol-24S-hydroxylase (CYP46A1) is specifically expressed in neurons of the neural retina. Curr Eye Res 32:361–366.
    OpenUrlCrossRefPubMed
  18. ↵
    1. Brøsen K,
    2. Skjelbo E,
    3. Rasmussen BB,
    4. Poulsen HE, and
    5. Loft S
    (1993) Fluvoxamine is a potent inhibitor of cytochrome P4501A2. Biochem Pharmacol 45:1211–1214.
    OpenUrlCrossRefPubMed
  19. ↵
    1. Bylund J,
    2. Finnström N, and
    3. Oliw EH
    (1999) Gene expression of a novel cytochrome P450 of the CYP4F subfamily in human seminal vesicles. Biochem Biophys Res Commun 261:169–174.
    OpenUrlCrossRefPubMed
  20. ↵
    1. Bylund J,
    2. Hidestrand M,
    3. Ingelman-Sundberg M, and
    4. Oliw EH
    (2000) Identification of CYP4F8 in human seminal vesicles as a prominent 19-hydroxylase of prostaglandin endoperoxides. J Biol Chem 275:21844–21849.
    OpenUrlAbstract/FREE Full Text
  21. ↵
    1. Cali JJ,
    2. Hsieh CL,
    3. Francke U, and
    4. Russell DW
    (1991) Mutations in the bile acid biosynthetic enzyme sterol 27-hydroxylase underlie cerebrotendinous xanthomatosis. J Biol Chem 266:7779–7783.
    OpenUrlAbstract/FREE Full Text
  22. ↵
    1. Capdevila JH,
    2. Falck JR, and
    3. Harris RC
    (2000) Cytochrome P450 and arachidonic acid bioactivation. Molecular and functional properties of the arachidonate monooxygenase. J Lipid Res 41:163–181.
    OpenUrlAbstract/FREE Full Text
  23. ↵
    1. Capozzi ME,
    2. McCollum GW, and
    3. Penn JS
    (2014) The role of cytochrome P450 epoxygenases in retinal angiogenesis. Invest Ophthalmol Vis Sci 55:4253–4260.
    OpenUrlAbstract/FREE Full Text
  24. ↵
    1. Cascio C,
    2. Deidda I,
    3. Russo D, and
    4. Guarneri P
    (2015) The estrogenic retina: the potential contribution to healthy aging and age-related neurodegenerative diseases of the retina. Steroids 103:31–41.
    OpenUrlCrossRefPubMed
    1. Cascio C,
    2. Russo D,
    3. Drago G,
    4. Galizzi G,
    5. Passantino R,
    6. Guarneri R, and
    7. Guarneri P
    (2007) 17beta-estradiol synthesis in the adult male rat retina. Exp Eye Res 85:166–172.
    OpenUrlCrossRefPubMed
  25. ↵
    1. Cenedella RJ
    (1982) Sterol synthesis by the ocular lens of the rat during postnatal development. J Lipid Res 23:619–626.
    OpenUrlAbstract
  26. ↵
    1. Cenedella RJ
    (1996) Cholesterol and cataracts. Surv Ophthalmol 40:320–337.
    OpenUrlCrossRefPubMed
  27. ↵
    1. Cenedella RJ and
    2. Fleschner CR
    (1989) Cholesterol biosynthesis by the cornea. Comparison of rates of sterol synthesis with accumulation during early development. J Lipid Res 30:1079–1084.
    OpenUrlAbstract
  28. ↵
    1. Charvet C,
    2. Liao WL,
    3. Heo GY,
    4. Laird J,
    5. Salomon RG,
    6. Turko IV, and
    7. Pikuleva IA
    (2011) Isolevuglandins and mitochondrial enzymes in the retina: mass spectrometry detection of post-translational modification of sterol-metabolizing CYP27A1. J Biol Chem 286:20413–20422.
    OpenUrlAbstract/FREE Full Text
  29. ↵
    1. Charvet CD,
    2. Laird J,
    3. Xu Y,
    4. Salomon RG, and
    5. Pikuleva IA
    (2013a) Posttranslational modification by an isolevuglandin diminishes activity of the mitochondrial cytochrome P450 27A1. J Lipid Res 54:1421–1429.
    OpenUrlAbstract/FREE Full Text
  30. ↵
    1. Charvet CD,
    2. Saadane A,
    3. Wang M,
    4. Salomon RG,
    5. Brunengraber H,
    6. Turko IV, and
    7. Pikuleva IA
    (2013b) Pretreatment with pyridoxamine mitigates isolevuglandin-associated retinal effects in mice exposed to bright light. J Biol Chem 288:29267–29280.
    OpenUrlAbstract/FREE Full Text
  31. ↵
    1. Chen P,
    2. Guo M,
    3. Wygle D,
    4. Edwards PA,
    5. Falck JR,
    6. Roman RJ, and
    7. Scicli AG
    (2005) Inhibitors of cytochrome P450 4A suppress angiogenic responses. Am J Pathol 166:615–624.
    OpenUrlCrossRefPubMed
  32. ↵
    1. Choi EH,
    2. Daruwalla A,
    3. Suh S,
    4. Leinonen H, and
    5. Palczewski K
    (2021) Retinoids in the visual cycle: role of the retinal G protein-coupled receptor. J Lipid Res 62:100040.
    OpenUrl
  33. ↵
    1. Choudhary D,
    2. Jansson I,
    3. Sarfarazi M, and
    4. Schenkman JB
    (2006) Physiological significance and expression of P450s in the developing eye. Drug Metab Rev 38:337–352.
    OpenUrlCrossRefPubMed
  34. ↵
    1. Choudhary D,
    2. Jansson I,
    3. Sarfarazi M, and
    4. Schenkman JB
    (2008) Characterization of the biochemical and structural phenotypes of four CYP1B1 mutations observed in individuals with primary congenital glaucoma. Pharmacogenet Genomics 18:665–676.
    OpenUrlCrossRefPubMed
  35. ↵
    1. Choudhary D,
    2. Jansson I,
    3. Stoilov I,
    4. Sarfarazi M, and
    5. Schenkman JB
    (2004) Metabolism of retinoids and arachidonic acid by human and mouse cytochrome P450 1b1. Drug Metab Dispos 32:840–847.
    OpenUrlAbstract/FREE Full Text
  36. ↵
    1. Chuang SS,
    2. Helvig C,
    3. Taimi M,
    4. Ramshaw HA,
    5. Collop AH,
    6. Amad M,
    7. White JA,
    8. Petkovich M,
    9. Jones G, and
    10. Korczak B
    (2004) CYP2U1, a novel human thymus- and brain-specific cytochrome P450, catalyzes omega- and (omega-1)-hydroxylation of fatty acids. J Biol Chem 279:6305–6314.
    OpenUrlAbstract/FREE Full Text
  37. ↵
    1. Cogan DG,
    2. Kruth HS,
    3. Datilis MB, and
    4. Martin N
    (1992) Corneal opacity in LCAT disease. Cornea 11:595–599.
    OpenUrlCrossRefPubMed
    1. Corpéchot C,
    2. Robel P,
    3. Axelson M,
    4. Sjövall J, and
    5. Baulieu EE
    (1981) Characterization and measurement of dehydroepiandrosterone sulfate in rat brain. Proc Natl Acad Sci USA 78:4704–4707.
    OpenUrlAbstract/FREE Full Text
  38. ↵
    1. Cruysberg JR,
    2. Wevers RA,
    3. van Engelen BG,
    4. Pinckers A,
    5. van Spreeken A, and
    6. Tolboom JJ
    (1995) Ocular and systemic manifestations of cerebrotendinous xanthomatosis. Am J Ophthalmol 120:597–604.
    OpenUrlPubMed
  39. ↵
    1. Curcio CA,
    2. Presley JB,
    3. Malek G,
    4. Medeiros NE,
    5. Avery DV, and
    6. Kruth HS
    (2005) Esterified and unesterified cholesterol in drusen and basal deposits of eyes with age-related maculopathy. Exp Eye Res 81:731–741.
    OpenUrlCrossRefPubMed
  40. ↵
    1. Debeljak N,
    2. Fink M, and
    3. Rozman D
    (2003) Many facets of mammalian lanosterol 14alpha-demethylase from the evolutionarily conserved cytochrome P450 family CYP51. Arch Biochem Biophys 409:159–171.
    OpenUrlCrossRefPubMed
  41. ↵
    1. Dhahir RK,
    2. Al-Nima AM, and
    3. Al-Bazzaz FY
    (2021) Nanoemulsions as ophthalmic drug delivery systems. Turk J Pharm Sci 18:652–664.
    OpenUrl
    1. Doshi M,
    2. Marcus C,
    3. Bejjani BA, and
    4. Edward DP
    (2006) Immunolocalization of CYP1B1 in normal, human, fetal and adult eyes. Exp Eye Res 82:24–32.
    OpenUrlCrossRefPubMed
  42. ↵
    1. Dosmar E,
    2. Walsh J,
    3. Doyel M,
    4. Bussett K,
    5. Oladipupo A,
    6. Amer S, and
    7. Goebel K
    (2022) Targeting ocular drug delivery: an examination of local anatomy and current approaches. Bioengineering (Basel) 9:41.
    OpenUrl
  43. ↵
    1. Dotti MT,
    2. Rufa A, and
    3. Federico A
    (2001) Cerebrotendinous xanthomatosis: heterogeneity of clinical phenotype with evidence of previously undescribed ophthalmological findings. J Inherit Metab Dis 24:696–706.
    OpenUrlCrossRefPubMed
  44. ↵
    1. Dubrac S,
    2. Lear SR,
    3. Ananthanarayanan M,
    4. Balasubramaniyan N,
    5. Bollineni J,
    6. Shefer S,
    7. Hyogo H,
    8. Cohen DE,
    9. Blanche PJ,
    10. Krauss RM et al.
    (2005) Role of CYP27A in cholesterol and bile acid metabolism. J Lipid Res 46:76–85.
    OpenUrlAbstract/FREE Full Text
  45. ↵
    1. Duvvuri S,
    2. Majumdar S, and
    3. Mitra AK
    (2003) Drug delivery to the retina: challenges and opportunities. Expert Opin Biol Ther 3:45–56.
    OpenUrlCrossRefPubMed
  46. ↵
    1. Duvvuri S,
    2. Majumdar S, and
    3. Mitra AK
    (2004) Role of metabolism in ocular drug delivery. Curr Drug Metab 5:507–515.
    OpenUrlCrossRefPubMed
  47. ↵
    1. El Matri K,
    2. Falfoul Y,
    3. Habibi I,
    4. Chebil A,
    5. Schorderet D, and
    6. El Matri L
    (2021) Macular dystrophy with bilateral macular telangiectasia related to the CYP2U1 pathogenic variant assessed with multimodal imaging including OCT-angiography. Genes (Basel) 12:1795.
    OpenUrl
  48. ↵
    1. El-Darzi N,
    2. Mast N,
    3. Buchner DA,
    4. Saadane A,
    5. Dailey B,
    6. Trichonas G, and
    7. Pikuleva IA
    (2022) Low-dose anti-HIV drug efavirenz mitigates retinal vascular lesions in a mouse model of Alzheimer’s disease. Front Pharmacol 13:902254.
    OpenUrl
  49. ↵
    1. Enright JM,
    2. Toomey MB,
    3. Sato SY,
    4. Temple SE,
    5. Allen JR,
    6. Fujiwara R,
    7. Kramlinger VM,
    8. Nagy LD,
    9. Johnson KM,
    10. Xiao Y et al.
    (2015) Cyp27c1 red-shifts the spectral sensitivity of photoreceptors by converting vitamin A1 into A2. Curr Biol 25:3048–3057.
    OpenUrlCrossRefPubMed
  50. ↵
    1. Falero-Perez J,
    2. Larsen MC,
    3. Teixeira LBC,
    4. Zhang HF,
    5. Lindner V,
    6. Sorenson CM,
    7. Jefcoate CR, and
    8. Sheibani N
    (2019a) Targeted deletion of Cyp1b1 in pericytes results in attenuation of retinal neovascularization and trabecular meshwork dysgenesis. Trends Dev Biol 12:1–12.
    OpenUrl
  51. ↵
    1. Falero-Perez J,
    2. Song YS,
    3. Sorenson CM, and
    4. Sheibani N
    (2018) CYP1B1: a key regulator of redox homeostasis. Trends Cell Mol Biol 13:27–45.
    OpenUrl
  52. ↵
    1. Falero-Perez J,
    2. Sorenson CM, and
    3. Sheibani N
    (2019b) Cyp1b1-deficient retinal astrocytes are more proliferative and migratory and are protected from oxidative stress and inflammation. Am J Physiol Cell Physiol 316:C767–C781.
    OpenUrl
    1. Farkouh A,
    2. Frigo P, and
    3. Czejka M
    (2016) Systemic side effects of eye drops: a pharmacokinetic perspective. Clin Ophthalmol 10:2433–2441.
    OpenUrl
  53. ↵
    1. Fer M,
    2. Dréano Y,
    3. Lucas D,
    4. Corcos L,
    5. Salaün JP,
    6. Berthou F, and
    7. Amet Y
    (2008) Metabolism of eicosapentaenoic and docosahexaenoic acids by recombinant human cytochromes P450. Arch Biochem Biophys 471:116–125.
    OpenUrlCrossRefPubMed
  54. ↵
    1. Fleming I
    (2014) The pharmacology of the cytochrome P450 epoxygenase/soluble epoxide hydrolase axis in the vasculature and cardiovascular disease. Pharmacol Rev 66:1106–1140.pmid:25244930
    OpenUrlAbstract/FREE Full Text
  55. ↵
    1. Flores R,
    2. Jin X,
    3. Chang J,
    4. Zhang C,
    5. Cogan DG,
    6. Schaefer EJ, and
    7. Kruth HS
    (2019) LCAT, ApoD, and ApoA1 expression and review of cholesterol deposition in the cornea. Biomolecules 9:785.
    OpenUrl
  56. ↵
    1. Fourgeux C,
    2. Martine L,
    3. Acar N,
    4. Bron AM,
    5. Creuzot-Garcher CP, and
    6. Bretillon L
    (2014) In vivo consequences of cholesterol-24S-hydroxylase (CYP46A1) inhibition by voriconazole on cholesterol homeostasis and function in the rat retina. Biochem Biophys Res Commun 446:775–781.
    OpenUrlCrossRefPubMed
    1. Fourgeux C,
    2. Martine L,
    3. Bjorkhem I,
    4. Diczfalusy U,
    5. Joffre C,
    6. Acar N,
    7. Creuzot-Garcher C,
    8. Bron A, and
    9. Bretillon L
    (2009) Primary open-angle glaucoma: association with cholesterol 24S-hydroxylase (CYP46A1) gene polymorphism and plasma 24-hydroxycholesterol levels. Invest Ophthalmol Vis Sci 50:5712–5717.
    OpenUrlAbstract/FREE Full Text
  57. ↵
    1. García-García GP,
    2. Martínez-Rubio M,
    3. Moya-Moya MA,
    4. Pérez-Santonja JJ, and
    5. Escribano J
    (2019) Current perspectives in Bietti crystalline dystrophy. Clin Ophthalmol 13:1379–1399.
    OpenUrl
  58. ↵
    1. Gaudana R,
    2. Ananthula HK,
    3. Parenky A, and
    4. Mitra AK
    (2010) Ocular drug delivery. AAPS J 12:348–360.
    OpenUrlCrossRefPubMed
  59. ↵
    1. Gaudana R,
    2. Jwala J,
    3. Boddu SHS, and
    4. Mitra AK
    (2009) Recent perspectives in ocular drug delivery. Pharm Res 26:1197–1216.
    OpenUrlCrossRefPubMed
  60. ↵
    1. Gaynor PM,
    2. Zhang WY,
    3. Weiss JS,
    4. Skarlatos SI,
    5. Rodrigues MM, and
    6. Kruth HS
    (1996) Accumulation of HDL apolipoproteins accompanies abnormal cholesterol accumulation in Schnyder’s corneal dystrophy. Arterioscler Thromb Vasc Biol 16:992–999.
    OpenUrlAbstract/FREE Full Text
  61. ↵
    1. Gong Y,
    2. Fu Z,
    3. Edin ML,
    4. Liu CH,
    5. Wang Z,
    6. Shao Z,
    7. Fredrick TW,
    8. Saba NJ,
    9. Morss PC,
    10. Burnim SB et al.
    (2016a) Cytochrome P450 oxidase 2C inhibition adds to ω-3 long-chain polyunsaturated fatty acids protection against retinal and choroidal neovascularization. Arterioscler Thromb Vasc Biol 36:1919–1927.
    OpenUrlAbstract/FREE Full Text
  62. ↵
    1. Gong Y,
    2. Fu Z,
    3. Liegl R,
    4. Chen J,
    5. Hellström A, and
    6. Smith LE
    (2017) ω-3 and ω-6 long-chain PUFAs and their enzymatic metabolites in neovascular eye diseases. Am J Clin Nutr 106:16–26.
    OpenUrlAbstract/FREE Full Text
  63. ↵
    1. Gong Y,
    2. Shao Z,
    3. Fu Z,
    4. Edin ML,
    5. Sun Y,
    6. Liegl RG,
    7. Wang Z,
    8. Liu CH,
    9. Burnim SB,
    10. Meng SS et al.
    (2016b) Fenofibrate inhibits cytochrome P450 epoxygenase 2C activity to suppress pathological ocular angiogenesis. EBioMedicine 13:201–211.
    OpenUrl
  64. ↵
    1. Gong Y,
    2. Tomita Y,
    3. Edin ML,
    4. Ren A,
    5. Ko M,
    6. Yang J,
    7. Bull E,
    8. Zeldin DC,
    9. Hellström A,
    10. Fu Z et al.
    (2022) Cytochrome P450 oxidase 2J inhibition suppresses choroidal neovascularization in mice. Metabolism 134:155266.
    OpenUrl
  65. ↵
    1. Guarneri P,
    2. Cascio C,
    3. Russo D,
    4. D’Agostino S,
    5. Drago G,
    6. Galizzi G,
    7. De Leo G,
    8. Piccoli F,
    9. Guarneri M, and
    10. Guarneri R
    (2003) Neurosteroids in the retina: neurodegenerative and neuroprotective agents in retinal degeneration. Ann N Y Acad Sci 1007:117–128.
    OpenUrlCrossRefPubMed
  66. ↵
    1. Guarneri P,
    2. Guarneri R,
    3. Cascio C,
    4. Pavasant P,
    5. Piccoli F, and
    6. Papadopoulos V
    (1994) Neurosteroidogenesis in rat retinas. J Neurochem 63:86–96.
    OpenUrlPubMed
  67. ↵
    1. Guengerich FP
    (2001) Common and uncommon cytochrome P450 reactions related to metabolism and chemical toxicity. Chem Res Toxicol 14:611–650.
    OpenUrlCrossRefPubMed
  68. ↵
    1. Ortiz de Montellano PR
    1. Guengerich FP
    (2005) Human cytochrome P450 enzymes, in Cytochrome P450 (Ortiz de Montellano PR ed), pp 377-530, Kluwer Academic/Plenum, New York.
  69. ↵
    1. Guengerich FP,
    2. Müller-Enoch D, and
    3. Blair IA
    (1986) Oxidation of quinidine by human liver cytochrome P-450. Mol Pharmacol 30:287–295.
    OpenUrlAbstract
  70. ↵
    1. Guengerich FP,
    2. Wu Z-L, and
    3. Bartleson CJ
    (2005) Function of human cytochrome P450s: characterization of the orphans. Biochem Biophys Res Commun 338:465–469.
    OpenUrlCrossRefPubMed
  71. ↵
    1. Hakkola J,
    2. Pasanen M,
    3. Pelkonen O,
    4. Hukkanen J,
    5. Evisalmi S,
    6. Anttila S,
    7. Rane A,
    8. Mäntylä M,
    9. Purkunen R,
    10. Saarikoski S et al.
    (1997) Expression of CYP1B1 in human adult and fetal tissues and differential inducibility of CYP1B1 and CYP1A1 by Ah receptor ligands in human placenta and cultured cells. Carcinogenesis 18:391–397.
    OpenUrlCrossRefPubMed
  72. ↵
    1. Hasegawa E,
    2. Inafuku S,
    3. Mulki L,
    4. Okunuki Y,
    5. Yanai R,
    6. Smith KE,
    7. Kim CB,
    8. Klokman G,
    9. Bielenberg DR,
    10. Puli N et al.
    (2017) Cytochrome P450 monooxygenase lipid metabolites are significant second messengers in the resolution of choroidal neovascularization. Proc Natl Acad Sci USA 114:E7545–E7553.
    OpenUrlAbstract/FREE Full Text
  73. ↵
    1. Hata M,
    2. Ikeda HO,
    3. Iwai S,
    4. Iida Y,
    5. Gotoh N,
    6. Asaka I,
    7. Ikeda K,
    8. Isobe Y,
    9. Hori A,
    10. Nakagawa S et al.
    (2018) Reduction of lipid accumulation rescues Bietti’s crystalline dystrophy phenotypes. Proc Natl Acad Sci USA 115:3936–3941.
    OpenUrlAbstract/FREE Full Text
  74. ↵
    1. Heo GY,
    2. Bederman I,
    3. Mast N,
    4. Liao WL,
    5. Turko IV, and
    6. Pikuleva IA
    (2011) Conversion of 7-ketocholesterol to oxysterol metabolites by recombinant CYP27A1 and retinal pigment epithelial cells. J Lipid Res 52:1117–1127.
    OpenUrlAbstract/FREE Full Text
    1. Hitchener WR and
    2. Cenedella RJ
    (1985) Absolute rates of sterol synthesis estimated from [3H]water for bovine lens epithelial cells in culture. J Lipid Res 26:1455–1463.
    OpenUrlAbstract
  75. ↵
    1. Huang J,
    2. Zhao Q,
    3. Li M,
    4. Duan Q,
    5. Zhao Y, and
    6. Zhang H
    (2019) The effects of endothelium-specific CYP2J2 overexpression on the attenuation of retinal ganglion cell apoptosis in a glaucoma rat model. FASEB J 33:11194–11209.
    OpenUrl
  76. ↵
    1. Iglesias-Osma MC,
    2. Blanco EJ,
    3. Carretero-Hernández M,
    4. Catalano-Iniesta L,
    5. García-Barrado MJ,
    6. Sánchez-Robledo V,
    7. Blázquez JL, and
    8. Carretero J
    (2022) The lack of Irs2 induces changes in the immunocytochemical expression of aromatase in the mouse retina. Ann Anat 239:151726.
    OpenUrl
    1. Ikeda H,
    2. Ueda M,
    3. Ikeda M,
    4. Kobayashi H, and
    5. Honda Y
    (2003) Oxysterol 7alpha-hydroxylase (CYP39A1) in the ciliary nonpigmented epithelium of bovine eye. Lab Invest 83:349–355.
    OpenUrlPubMed
    1. Incorvaia C,
    2. Parmeggiani F,
    3. Costagliola C,
    4. Perri P,
    5. Tittoni M, and
    6. Sebastiani A
    (2003) Congenital adrenal hyperplasia due to 21-hydroxylase deficiency associated with bilateral keratoconus. Am J Ophthalmol 135:557–559.
    OpenUrlPubMed
  77. ↵
    1. Ishikawa M,
    2. Yoshitomi T,
    3. Covey DF,
    4. Zorumski CF, and
    5. Izumi Y
    (2018) Neurosteroids and oxysterols as potential therapeutic agents for glaucoma and Alzheimer’s disease. Neuropsychiatry (London) 8:344–359.
    OpenUrl
    1. Ishikawa M,
    2. Yoshitomi T,
    3. Zorumski CF, and
    4. Izumi Y
    (2016) 24(S)-Hydroxycholesterol protects the ex vivo rat retina from injury by elevated hydrostatic pressure. Sci Rep 6:33886.
    OpenUrl
  78. ↵
    1. Isoherranen N and
    2. Zhong G
    (2019) Biochemical and physiological importance of the CYP26 retinoic acid hydroxylases. Pharmacol Ther 204:107400.
    OpenUrlCrossRefPubMed
  79. ↵
    1. Jaliffa CO,
    2. Howard S,
    3. Hoijman E,
    4. Salido E,
    5. Sarmiento MI,
    6. Arias P, and
    7. Rosenstein RE
    (2005) Effect of neurosteroids on the retinal gabaergic system and electroretinographic activity in the golden hamster. J Neurochem 94:1666–1675.
    OpenUrlCrossRefPubMed
    1. Janagam DR,
    2. Wu L, and
    3. Lowe TL
    (2017) Nanoparticles for drug delivery to the anterior segment of the eye. Adv Drug Deliv Rev 122:31–64.
    OpenUrl
  80. ↵
    1. Jansson I,
    2. Stoilov I,
    3. Sarfarazi M, and
    4. Schenkman JB
    (2001) Effect of two mutations of human CYP1B1, G61E and R469W, on stability and endogenous steroid substrate metabolism. Pharmacogenetics 11:793–801.
    OpenUrlCrossRefPubMed
  81. ↵
    1. Jarrar YB and
    2. Lee SJ
    (2019) Molecular functionality of cytochrome P450 4 (CYP4) genetic polymorphisms and their clinical implications. Int J Mol Sci 20:4274.
    OpenUrl
  82. ↵
    1. Johnson JA,
    2. Herring VL,
    3. Wolfe MS, and
    4. Relling MV
    (2000) CYP1A2 and CYP2D6 4-hydroxylate propranolol and both reactions exhibit racial differences. J Pharmacol Exp Ther 294:1099–1105.
    OpenUrlAbstract/FREE Full Text
  83. ↵
    1. Yan Z and
    2. Caldwell GW
    1. Johnson KM,
    2. Su D, and
    3. Zhang D
    (2021) Characteristics of major drug metabolizing cytochrome p450 enzymes, in Cytochrome P450: In Vitro Methods and Protocols (Yan Z and Caldwell GW eds), pp 27-54, Springer, New York.
  84. ↵
    1. Khurana AK,
    2. Khurana AK, and
    3. Khurana B
    (2015) Comprehensive Ophthalmology (A Free Companion: Review of Ophthalmology, Jaypee Brothers Medical Publishers [P] Ltd, New Delhi, India.
  85. ↵
    1. Kimonen T,
    2. Juvonen RO,
    3. Alhava E, and
    4. Pasanen M
    (1995) The inhibition of CYP enzymes in mouse and human liver by pilocarpine. Br J Pharmacol 114:832–836.
    OpenUrl
  86. ↵
    1. Kölln C and
    2. Reichl S
    (2016) Cytochrome P450 activity in ex vivo cornea models and a human cornea construct. J Pharm Sci 105:2204–2212.
    OpenUrl
    1. Koyama S,
    2. Sekijima Y,
    3. Ogura M,
    4. Hori M,
    5. Matsuki K,
    6. Miida T, and
    7. Harada-Shiba M
    (2021) Cerebrotendinous xanthomatosis: molecular pathogenesis, clinical spectrum, diagnosis, and disease-modifying treatments. J Atheroscler Thromb 28:905–925.
    OpenUrl
  87. ↵
    1. Lee J,
    2. Jiao X,
    3. Hejtmancik JF,
    4. Kaiser-Kupfer M, and
    5. Chader GJ
    (1998) Identification, isolation, and characterization of a 32-kDa fatty acid-binding protein missing from lymphocytes in humans with Bietti crystalline dystrophy (BCD). Mol Genet Metab 65:143–154.
    OpenUrlCrossRefPubMed
  88. ↵
    1. Lee J,
    2. Jiao X,
    3. Hejtmancik JF,
    4. Kaiser-Kupfer M,
    5. Gahl WA,
    6. Markello TC,
    7. Guo J, and
    8. Chader GJ
    (2001) The metabolism of fatty acids in human Bietti crystalline dystrophy. Invest Ophthalmol Vis Sci 42:1707–1714.
    OpenUrlAbstract/FREE Full Text
  89. ↵
    1. Lee JW,
    2. Fuda H,
    3. Javitt NB,
    4. Strott CA, and
    5. Rodriguez IR
    (2006) Expression and localization of sterol 27-hydroxylase (CYP27A1) in monkey retina. Exp Eye Res 83:465–469.
    OpenUrlCrossRefPubMed
  90. ↵
    1. Lee SY,
    2. Jang H,
    3. Lee JY,
    4. Kwon KI,
    5. Oh SJ, and
    6. Kim SK
    (2014) Inhibition of cytochrome P450 by ethambutol in human liver microsomes. Toxicol Lett 229:33–40.
    OpenUrlCrossRefPubMed
  91. ↵
    1. Leo MA,
    2. Lasker JM,
    3. Raucy JL,
    4. Kim CI,
    5. Black M, and
    6. Lieber CS
    (1989) Metabolism of retinol and retinoic acid by human liver cytochrome P450IIC8. Arch Biochem Biophys 269:305–312.
    OpenUrlCrossRefPubMed
    1. Leonardi L,
    2. Ziccardi L,
    3. Marcotulli C,
    4. Rubegni A,
    5. Longobardi A,
    6. Serrao M,
    7. Storti E,
    8. Pierelli F,
    9. Tessa A,
    10. Parisi V et al.
    (2016) Pigmentary degenerative maculopathy as prominent phenotype in an Italian SPG56/CYP2U1 family. J Neurol 263:781–783.
    OpenUrl
  92. ↵
    1. Lepesheva GI and
    2. Waterman MR
    (2004) CYP51—the omnipotent P450. Mol Cell Endocrinol 215:165–170.
    OpenUrlCrossRefPubMed
  93. ↵
    1. Li A,
    2. Jiao X,
    3. Munier FL,
    4. Schorderet DF,
    5. Yao W,
    6. Iwata F,
    7. Hayakawa M,
    8. Kanai A,
    9. Shy Chen M,
    10. Alan Lewis R et al.
    (2004) Bietti crystalline corneoretinal dystrophy is caused by mutations in the novel gene CYP4V2. Am J Hum Genet 74:817–826.
    OpenUrlCrossRefPubMed
  94. ↵
    1. Li LK,
    2. So L, and
    3. Spector A
    (1985) Membrane cholesterol and phospholipid in consecutive concentric sections of human lenses. J Lipid Res 26:600–609.
    OpenUrlAbstract
  95. ↵
    1. Li N,
    2. Zhou Y,
    3. Du L,
    4. Wei M, and
    5. Chen X
    (2011) Overview of cytochrome P450 1B1 gene mutations in patients with primary congenital glaucoma. Exp Eye Res 93:572–579.
    OpenUrlCrossRefPubMed
  96. ↵
    1. Liao WL,
    2. Heo GY,
    3. Dodder NG,
    4. Reem RE,
    5. Mast N,
    6. Huang S,
    7. Dipatre PL,
    8. Turko IV, and
    9. Pikuleva IA
    (2011) Quantification of cholesterol-metabolizing P450s CYP27A1 and CYP46A1 in neural tissues reveals a lack of enzyme-product correlations in human retina but not human brain. J Proteome Res 10:241–248.
    OpenUrlCrossRefPubMed
  97. ↵
    1. Libby RT,
    2. Smith RS,
    3. Savinova OV,
    4. Zabaleta A,
    5. Martin JE,
    6. Gonzalez FJ, and
    7. John SW
    (2003) Modification of ocular defects in mouse developmental glaucoma models by tyrosinase. Science 299:1578–1581.
    OpenUrlAbstract/FREE Full Text
  98. ↵
    1. Li-Hawkins J,
    2. Lund EG,
    3. Bronson AD, and
    4. Russell DW
    (2000) Expression cloning of an oxysterol 7alpha-hydroxylase selective for 24-hydroxycholesterol. J Biol Chem 275:16543–16549.
    OpenUrlAbstract/FREE Full Text
  99. ↵
    1. Lin JB,
    2. Mast N,
    3. Bederman IR,
    4. Li Y,
    5. Brunengraber H,
    6. Björkhem I, and
    7. Pikuleva IA
    (2016) Cholesterol in mouse retina originates primarily from in situ de novo biosynthesis. J Lipid Res 57:258–264.
    OpenUrlAbstract/FREE Full Text
  100. ↵
    1. Lockhart CM,
    2. Nakano M,
    3. Rettie AE, and
    4. Kelly EJ
    (2014) Generation and characterization of a murine model of Bietti crystalline dystrophy. Invest Ophthalmol Vis Sci 55:5572–5581.
    OpenUrlAbstract/FREE Full Text
    1. Lund EG,
    2. Guileyardo JM, and
    3. Russell DW
    (1999) cDNA cloning of cholesterol 24-hydroxylase, a mediator of cholesterol homeostasis in the brain. Proc Natl Acad Sci USA 96:7238–7243.
    OpenUrlAbstract/FREE Full Text
    1. Luo T,
    2. Sakai Y,
    3. Wagner E, and
    4. Dräger UC
    (2006) Retinoids, eye development, and maturation of visual function. J Neurobiol 66:677–686.
    OpenUrlCrossRefPubMed
  101. ↵
    1. Lynch T and
    2. Price A
    (2007) The effect of cytochrome P450 metabolism on drug response, interactions, and adverse effects. Am Fam Physician 76:391–396.
    OpenUrlPubMed
  102. ↵
    1. Ma X,
    2. Idle JR,
    3. Krausz KW, and
    4. Gonzalez FJ
    (2005) Metabolism of melatonin by human cytochromes p450. Drug Metab Dispos 33:489–494.
    OpenUrlAbstract/FREE Full Text
  103. ↵
    1. Martinez-Gil N,
    2. Flores-Bellver M,
    3. Atienzar-Aroca S,
    4. Lopez-Malo D,
    5. Urdaneta AC,
    6. Sancho-Pelluz J,
    7. Peris-Martínez C,
    8. Bonet-Ponce L,
    9. Romero FJ, and
    10. Barcia JM
    (2015) CYP2E1 in the human retinal pigment epithelium: expression, activity, and induction by ethanol. Invest Ophthalmol Vis Sci 56:6855–6863.
    OpenUrl
  104. ↵
    1. Mast N,
    2. Reem R,
    3. Bederman I,
    4. Huang S,
    5. DiPatre PL,
    6. Björkhem I, and
    7. Pikuleva IA
    (2011) Cholestenoic acid is an important elimination product of cholesterol in the retina: comparison of retinal cholesterol metabolism with that in the brain. Invest Ophthalmol Vis Sci 52:594–603.
    OpenUrlAbstract/FREE Full Text
  105. ↵
    1. Mastyugin V,
    2. Aversa E,
    3. Bonazzi A,
    4. Vafaes C,
    5. Mieyal P, and
    6. Schwartzman ML
    (1999) Hypoxia-induced production of 12-hydroxyeicosanoids in the corneal epithelium: involvement of a cytochrome P-4504B1 isoform. J Pharmacol Exp Ther 289:1611–1619.
    OpenUrlAbstract/FREE Full Text
  106. ↵
    1. Mastyugin V,
    2. Mezentsev A,
    3. Zhang WX,
    4. Ashkar S,
    5. Dunn MW, and
    6. Laniado-Schwartzman M
    (2004) Promoter activity and regulation of the corneal CYP4B1 gene by hypoxia. J Cell Biochem 91:1218–1238.
    OpenUrlCrossRefPubMed
    1. Mastyugin V,
    2. Mosaed S,
    3. Bonazzi A,
    4. Dunn MW, and
    5. Schwartzman ML
    (2001) Corneal epithelial VEGF and cytochrome P450 4B1 expression in a rabbit model of closed eye contact lens wear. Curr Eye Res 23:1–10.
    OpenUrlCrossRefPubMed
  107. ↵
    1. McDonald MG,
    2. Au NT, and
    3. Rettie AE
    (2015) P450-based drug-drug interactions of amiodarone and its metabolites: diversity of inhibitory mechanisms. Drug Metab Dispos 43:1661–1669.
    OpenUrlAbstract/FREE Full Text
  108. ↵
    1. McKay TB,
    2. Priyadarsini S, and
    3. Karamichos D
    (2022) Sex hormones, growth hormone, and the cornea. Cells 11:224.
    OpenUrl
    1. Meaney S,
    2. Heverin M,
    3. Panzenboeck U,
    4. Ekström L,
    5. Axelsson M,
    6. Andersson U,
    7. Diczfalusy U,
    8. Pikuleva I,
    9. Wahren J,
    10. Sattler W et al.
    (2007) Novel route for elimination of brain oxysterols across the blood-brain barrier: conversion into 7alpha-hydroxy-3-oxo-4-cholestenoic acid. J Lipid Res 48:944–951.
    OpenUrlAbstract/FREE Full Text
  109. ↵
    1. Mezentsev A,
    2. Mastyugin V,
    3. Seta F,
    4. Ashkar S,
    5. Kemp R,
    6. Reddy DS,
    7. Falck JR,
    8. Dunn MW, and
    9. Laniado-Schwartzman M
    (2005) Transfection of cytochrome P4504B1 into the cornea increases angiogenic activity of the limbal vessels. J Pharmacol Exp Ther 315:42–50.
    OpenUrlAbstract/FREE Full Text
  110. ↵
    1. Miller WL and
    2. Auchus RJ
    (2011) The molecular biology, biochemistry, and physiology of human steroidogenesis and its disorders. Endocr Rev 32:81–151.
    OpenUrlCrossRefPubMed
  111. ↵
    1. Miyamoto M,
    2. Ishii N,
    3. Mochizuki H,
    4. Shiomi K,
    5. Kaida T,
    6. Chuman H, and
    7. Nakazato M
    (2019) Optic neuropathy with features suggestive of optic neuritis in cerebrotendinous xanthomatosis. Case Rep Neurol Med 2019:2576826.
    OpenUrl
  112. ↵
    1. Morawski K,
    2. Klonowska A,
    3. Kubicka-Trzaska A,
    4. Woron J, and
    5. Romanowska-Dixon B
    (2020) Central serous chorioretinopathy induced by drugs metabolized by cytochrome P450 3A4. J Physiol Pharmacol DOI: 10.26402/jpp.2020.2.15 [published ahead of print].
  113. ↵
    1. Morgan SJ,
    2. McKenna P, and
    3. Bosanquet RC
    (1989) Case of cerebrotendinous xanthomatosis. I: unusual ophthalmic features. Br J Ophthalmol 73:1011–1014.
    OpenUrlAbstract/FREE Full Text
  114. ↵
    1. Mori M,
    2. Li G,
    3. Abe I,
    4. Nakayama J,
    5. Guo Z,
    6. Sawashita J,
    7. Ugawa T,
    8. Nishizono S,
    9. Serikawa T,
    10. Higuchi K et al.
    (2006) Lanosterol synthase mutations cause cholesterol deficiency-associated cataracts in the Shumiya cataract rat. J Clin Invest 116:395–404.
    OpenUrlCrossRefPubMed
  115. ↵
    1. Mossböck G,
    2. Weger M,
    3. Faschinger C,
    4. Schmut O,
    5. Renner W,
    6. Wedrich A,
    7. Zimmermann C, and
    8. El-Shabrawi Y
    (2011) Role of cholesterol 24S-hydroxylase gene polymorphism (rs754203) in primary open angle glaucoma. Mol Vis 17:616–620.
    OpenUrlPubMed
  116. ↵
    1. Nakamura K,
    2. Fujiki T, and
    3. Tamura HO
    (2005) Age, gender and region-specific differences in drug metabolising enzymes in rat ocular tissues. Exp Eye Res 81:710–715.
    OpenUrlCrossRefPubMed
  117. ↵
    1. Nakano M,
    2. Kelly EJ, and
    3. Rettie AE
    (2009) Expression and characterization of CYP4V2 as a fatty acid omega-hydroxylase. Drug Metab Dispos 37:2119–2122.
    OpenUrlAbstract/FREE Full Text
  118. ↵
    1. Nakano M,
    2. Kelly EJ,
    3. Wiek C,
    4. Hanenberg H, and
    5. Rettie AE
    (2012) CYP4V2 in Bietti’s crystalline dystrophy: ocular localization, metabolism of ω-3-polyunsaturated fatty acids, and functional deficit of the p.H331P variant. Mol Pharmacol 82:679–686.
    OpenUrlAbstract/FREE Full Text
    1. Nakano M,
    2. Lockhart CM,
    3. Kelly EJ, and
    4. Rettie AE
    (2014) Ocular cytochrome P450s and transporters: roles in disease and endobiotic and xenobiotic disposition. Drug Metab Rev 46:247–260.
    OpenUrlCrossRefPubMed
    1. Nelson DR
    (2018) Cytochrome P450 diversity in the tree of life. Biochim Biophys Acta Proteins Proteomics 1866:141–154.
    OpenUrl
  119. ↵
    1. Nelson DR,
    2. Zeldin DC,
    3. Hoffman SM,
    4. Maltais LJ,
    5. Wain HM, and
    6. Nebert DW
    (2004) Comparison of cytochrome P450 (CYP) genes from the mouse and human genomes, including nomenclature recommendations for genes, pseudogenes and alternative-splice variants. Pharmacogenetics 14:1–18.
    OpenUrlCrossRefPubMed
  120. ↵
    1. Ni KD and
    2. Liu JY
    (2021) The functions of cytochrome P450 ω-hydroxylases and the associated eicosanoids in inflammation-related diseases. Front Pharmacol 12:716801.
    OpenUrl
  121. ↵
    1. Novack GD and
    2. Robin AL
    (2016) Ocular pharmacology. J Clin Pharmacol 56:517–527.
    OpenUrl
    1. Oak AS,
    2. Messinger JD, and
    3. Curcio CA
    (2014) Subretinal drusenoid deposits: further characterization by lipid histochemistry. Retina 34:825–826.
    OpenUrlCrossRefPubMed
  122. ↵
    1. Okita RT and
    2. Okita JR
    (2001) Cytochrome P450 4A fatty acid omega hydroxylases. Curr Drug Metab 2:265–281.
    OpenUrlCrossRefPubMed
  123. ↵
    1. Omarova S,
    2. Charvet CD,
    3. Reem RE,
    4. Mast N,
    5. Zheng W,
    6. Huang S,
    7. Peachey NS, and
    8. Pikuleva IA
    (2012) Abnormal vascularization in mouse retina with dysregulated retinal cholesterol homeostasis. J Clin Invest 122:3012–3023.
    OpenUrlCrossRefPubMed
  124. ↵
    1. Peterson JA
    (2019) Shareable resource: ten nice-to-know facts about the eyes. ACSM’s Health Fit J 23:53.
    OpenUrl
  125. ↵
    1. Pikuleva IA,
    2. Babiker A,
    3. Waterman MR, and
    4. Björkhem I
    (1998) Activities of recombinant human cytochrome P450c27 (CYP27) which produce intermediates of alternative bile acid biosynthetic pathways. J Biol Chem 273:18153–18160.
    OpenUrlAbstract/FREE Full Text
  126. ↵
    1. Pikuleva IA and
    2. Curcio CA
    (2014) Cholesterol in the retina: the best is yet to come. Prog Retin Eye Res 41:64–89.
    OpenUrlCrossRefPubMed
  127. ↵
    1. Qiu F,
    2. Matlock G,
    3. Chen Q,
    4. Zhou K,
    5. Du Y,
    6. Wang X, and
    7. Ma JX
    (2017) Therapeutic effects of PPARα agonist on ocular neovascularization in models recapitulating neovascular age-related macular degeneration. Invest Ophthalmol Vis Sci 58:5065–5075.
    OpenUrl
  128. ↵
    1. Quigley HA and
    2. Broman AT
    (2006) The number of people with glaucoma worldwide in 2010 and 2020. Br J Ophthalmol 90:262–267.
    OpenUrlAbstract/FREE Full Text
    1. Ramirez DM,
    2. Andersson S, and
    3. Russell DW
    (2008) Neuronal expression and subcellular localization of cholesterol 24-hydroxylase in the mouse brain. J Comp Neurol 507:1676–1693.
    OpenUrlCrossRefPubMed
  129. ↵
    1. Rauf B,
    2. Irum B,
    3. Kabir F,
    4. Firasat S,
    5. Naeem MA,
    6. Khan SN,
    7. Husnain T,
    8. Riazuddin S,
    9. Akram J, and
    10. Riazuddin SA
    (2016) A spectrum of CYP1B1 mutations associated with primary congenital glaucoma in families of Pakistani descent. Hum Genome Var 3:16021.
    OpenUrl
  130. ↵
    1. Raza ST,
    2. Abbas S,
    3. Chandra A,
    4. Singh L,
    5. Rizvi S, and
    6. Mahdi F
    (2017) Association of angiotensin-converting enzyme, CYP46A1 genes polymorphism with senile cataract. Oman J Ophthalmol 10:21–25.
    OpenUrl
  131. ↵
    1. Rendic S
    (2002) Summary of information on human CYP enzymes: human P450 metabolism data. Drug Metab Rev 34:83–448.
    OpenUrlCrossRefPubMed
  132. ↵
    1. Rendic S and
    2. Guengerich FP
    (2020) Metabolism and interactions of chloroquine and hydroxychloroquine with human cytochrome P450 enzymes and drug transporters. Curr Drug Metab 21:1127–1135.
    OpenUrl
  133. ↵
    1. Resnikoff S,
    2. Pascolini D,
    3. Etya’ale D,
    4. Kocur I,
    5. Pararajasegaram R,
    6. Pokharel GP, and
    7. Mariotti SP
    (2004) Global data on visual impairment in the year 2002. Bull World Health Organ 82:844–851.
    OpenUrlCrossRefPubMed
  134. ↵
    1. Rosen H,
    2. Reshef A,
    3. Maeda N,
    4. Lippoldt A,
    5. Shpizen S,
    6. Triger L,
    7. Eggertsen G,
    8. Björkhem I, and
    9. Leitersdorf E
    (1998) Markedly reduced bile acid synthesis but maintained levels of cholesterol and vitamin D metabolites in mice with disrupted sterol 27-hydroxylase gene. J Biol Chem 273:14805–14812.
    OpenUrlAbstract/FREE Full Text
  135. ↵
    1. Saadane A,
    2. Mast N,
    3. Charvet CD,
    4. Omarova S,
    5. Zheng W,
    6. Huang SS,
    7. Kern TS,
    8. Peachey NS, and
    9. Pikuleva IA
    (2014) Retinal and nonocular abnormalities in Cyp27a1(-/-)Cyp46a1(-/-) mice with dysfunctional metabolism of cholesterol. Am J Pathol 184:2403–2419.
    OpenUrlCrossRefPubMed
  136. ↵
    1. Saadane A,
    2. Mast N,
    3. Trichonas G,
    4. Chakraborty D,
    5. Hammer S,
    6. Busik JV,
    7. Grant MB, and
    8. Pikuleva IA
    (2019) Retinal vascular abnormalities and microglia activation in mice with deficiency in cytochrome P450 46A1-mediated cholesterol removal. Am J Pathol 189:405–425.
    OpenUrlCrossRefPubMed
  137. ↵
    1. Sakai Y,
    2. Luo T,
    3. McCaffery P,
    4. Hamada H, and
    5. Dräger UC
    (2004) CYP26A1 and CYP26C1 cooperate in degrading retinoic acid within the equatorial retina during later eye development. Dev Biol 276:143–157.
    OpenUrlCrossRefPubMed
  138. ↵
    1. Schirra F,
    2. Suzuki T,
    3. Dickinson DP,
    4. Townsend DJ,
    5. Gipson IK, and
    6. Sullivan DA
    (2006) Identification of steroidogenic enzyme mRNAs in the human lacrimal gland, meibomian gland, cornea, and conjunctiva. Cornea 25:438–442.
    OpenUrlCrossRefPubMed
  139. ↵
    1. Scott AW,
    2. Bressler NM,
    3. Ffolkes S,
    4. Wittenborn JS, and
    5. Jorkasky J
    (2016) Public attitudes about eye and vision health. JAMA Ophthalmol 134:1111–1118.
    OpenUrl
  140. ↵
    1. Seta F,
    2. Patil K,
    3. Bellner L,
    4. Mezentsev A,
    5. Kemp R,
    6. Dunn MW, and
    7. Schwartzman ML
    (2007) Inhibition of VEGF expression and corneal neovascularization by siRNA targeting cytochrome P450 4B1. Prostaglandins Other Lipid Mediat 84:116–127.
    OpenUrlCrossRefPubMed
  141. ↵
    1. Shao Z,
    2. Fu Z,
    3. Stahl A,
    4. Joyal JS,
    5. Hatton C,
    6. Juan A,
    7. Hurst C,
    8. Evans L,
    9. Cui Z,
    10. Pei D et al.
    (2014) Cytochrome P450 2C8 ω3-long-chain polyunsaturated fatty acid metabolites increase mouse retinal pathologic neovascularization--brief report. Arterioscler Thromb Vasc Biol 34:581–586.
    OpenUrlAbstract/FREE Full Text
    1. Sharif Z and
    2. Sharif W
    (2019) Corneal neovascularization: updates on pathophysiology, investigations & management. Rom J Ophthalmol 63:15–22.
    OpenUrl
  142. ↵
    1. Shichi H and
    2. Nebert DW
    (1982) Genetic differences in drug metabolism associated with ocular toxicity. Environ Health Perspect 44:107–117.
    OpenUrlPubMed
  143. ↵
    1. Singh MS,
    2. Francis PA, and
    3. Michael M
    (2011) Tamoxifen, cytochrome P450 genes and breast cancer clinical outcomes. Breast 20:111–118.
    OpenUrlCrossRefPubMed
  144. ↵
    1. Siu TL,
    2. Morley JW, and
    3. Coroneo MT
    (2008) Toxicology of the retina: advances in understanding the defence mechanisms and pathogenesis of drug- and light-induced retinopathy. Clin Exp Ophthalmol 36:176–185.
    OpenUrlPubMed
  145. ↵
    1. Song YS,
    2. Annalora AJ,
    3. Marcus CB,
    4. Jefcoate CR,
    5. Sorenson CM, and
    6. Sheibani N
    (2022) Cytochrome P450 1B1: a key regulator of ocular iron homeostasis and oxidative stress. Cells 11:2930.
    OpenUrl
  146. ↵
    1. Souza Monteiro de Araújo D,
    2. Brito R,
    3. Pereira-Figueiredo D,
    4. Dos Santos-Rodrigues A,
    5. De Logu F,
    6. Nassini R,
    7. Zin A, and
    8. Calaza KC
    (2022) Retinal toxicity induced by chemical agents. Int J Mol Sci 23:8182.
    OpenUrl
  147. ↵
    1. Stark K,
    2. Törmä H,
    3. Cristea M, and
    4. Oliw EH
    (2003) Expression of CYP4F8 (prostaglandin H 19-hydroxylase) in human epithelia and prominent induction in epidermis of psoriatic lesions. Arch Biochem Biophys 409:188–196.
    OpenUrlCrossRefPubMed
  148. ↵
    1. Stoilov I
    (2001) Cytochrome P450s: coupling development and environment. Trends Genet 17:629–632.
    OpenUrlCrossRefPubMed
  149. ↵
    1. Stoilov I,
    2. Akarsu AN, and
    3. Sarfarazi M
    (1997) Identification of three different truncating mutations in cytochrome P4501B1 (CYP1B1) as the principal cause of primary congenital glaucoma (Buphthalmos) in families linked to the GLC3A locus on chromosome 2p21. Hum Mol Genet 6:641–647.
    OpenUrlCrossRefPubMed
  150. ↵
    1. Stoilov I,
    2. Jansson I,
    3. Sarfarazi M, and
    4. Schenkman JB
    (2001) Roles of cytochrome p450 in development. Drug Metabol Drug Interact 18:33–55.
    OpenUrlPubMed
  151. ↵
    1. Sullivan DA
    (2004) Tearful relationships? Sex, hormones, the lacrimal gland, and aqueous-deficient dry eye. Ocul Surf 2:92–123.
    OpenUrlCrossRefPubMed
  152. ↵
    1. Susarla R,
    2. Liu L,
    3. Walker EA,
    4. Bujalska IJ,
    5. Alsalem J,
    6. Williams GP,
    7. Sreekantam S,
    8. Taylor AE,
    9. Tallouzi M,
    10. Southworth HS et al.
    (2014) Cortisol biosynthesis in the human ocular surface innate immune response. PLoS One 9:e94913.
    OpenUrlCrossRefPubMed
  153. ↵
    1. Syed MF,
    2. Rehmani A, and
    3. Yang M
    (2021) Ocular side effects of common systemic medications and systemic side effects of ocular medications. Med Clin North Am 105:425–444.
    OpenUrl
  154. ↵
    1. Teixeira LB,
    2. Zhao Y,
    3. Dubielzig RR,
    4. Sorenson CM, and
    5. Sheibani N
    (2015) Ultrastructural abnormalities of the trabecular meshwork extracellular matrix in Cyp1b1-deficient mice. Vet Pathol 52:397–403.
    OpenUrlCrossRefPubMed
  155. ↵
    1. Tesson C,
    2. Nawara M,
    3. Salih MA,
    4. Rossignol R,
    5. Zaki MS,
    6. Al Balwi M,
    7. Schule R,
    8. Mignot C,
    9. Obre E,
    10. Bouhouche A et al.
    (2012) Alteration of fatty-acid-metabolizing enzymes affects mitochondrial form and function in hereditary spastic paraplegia. Am J Hum Genet 91:1051–1064.
    OpenUrlCrossRefPubMed
  156. ↵
    1. Thomas AG and
    2. Henry JJ
    (2014) Retinoic acid regulation by CYP26 in vertebrate lens regeneration. Dev Biol 386:291–301.
    OpenUrl
  157. ↵
    1. Tsao CC,
    2. Coulter SJ,
    3. Chien A,
    4. Luo G,
    5. Clayton NP,
    6. Maronpot R,
    7. Goldstein JA, and
    8. Zeldin DC
    (2001) Identification and localization of five CYP2Cs in murine extrahepatic tissues and their metabolism of arachidonic acid to regio- and stereoselective products. J Pharmacol Exp Ther 299:39–47.
    OpenUrlAbstract/FREE Full Text
  158. ↵
    1. Vaajanen A and
    2. Vapaatalo H
    (2017) A single drop in the eye—effects on the whole body? Open Ophthalmol J 11:305–314.
    OpenUrlPubMed
  159. ↵
    1. Vasiliou V and
    2. Gonzalez FJ
    (2008) Role of CYP1B1 in glaucoma. Annu Rev Pharmacol Toxicol 48:333–358.
    OpenUrlCrossRefPubMed
  160. ↵
    1. von Lintig J,
    2. Moon J, and
    3. Babino D
    (2021) Molecular components affecting ocular carotenoid and retinoid homeostasis. Prog Retin Eye Res 80:100864.
    OpenUrlPubMed
    1. Walsky RL,
    2. Gaman EA, and
    3. Obach RS
    (2005a) Examination of 209 drugs for inhibition of cytochrome P450 2C8. J Clin Pharmacol 45:68–78.
    OpenUrlCrossRefPubMed
  161. ↵
    1. Walsky RL,
    2. Obach RS,
    3. Gaman EA,
    4. Gleeson JP, and
    5. Proctor WR
    (2005b) Selective inhibition of human cytochrome P4502C8 by montelukast. Drug Metab Dispos 33:413–418.
    OpenUrlAbstract/FREE Full Text
  162. ↵
    1. Wang JH,
    2. Lidgerwood GE,
    3. Daniszewski M,
    4. Hu ML,
    5. Roberts GE,
    6. Wong RCB,
    7. Hung SSC,
    8. McClements ME,
    9. Hewitt AW,
    10. Pébay A et al.
    (2022) AAV2-mediated gene therapy for Bietti crystalline dystrophy provides functional CYP4V2 in multiple relevant cell models. Sci Rep 12:9525.
    OpenUrl
  163. ↵
    1. Wang L,
    2. Clark ME,
    3. Crossman DK,
    4. Kojima K,
    5. Messinger JD,
    6. Mobley JA, and
    7. Curcio CA
    (2010) Abundant lipid and protein components of drusen. PLoS One 5:e10329.
    OpenUrlCrossRefPubMed
  164. ↵
    1. Wang M,
    2. Heo GY,
    3. Omarova S,
    4. Pikuleva IA, and
    5. Turko IV
    (2012) Sample prefractionation for mass spectrometry quantification of low-abundance membrane proteins. Anal Chem 84:5186–5191.
    OpenUrlCrossRefPubMed
  165. ↵
    1. Widomska J and
    2. Subczynski WK
    (2019) Why is very high cholesterol content beneficial for the eye lens but negative for other organs? Nutrients 11:1083.
    OpenUrl
  166. ↵
    1. Wikvall K
    (1984) Hydroxylations in biosynthesis of bile acids. Isolation of a cytochrome P-450 from rabbit liver mitochondria catalyzing 26-hydroxylation of C27-steroids. J Biol Chem 259:3800–3804.
    OpenUrlAbstract/FREE Full Text
  167. ↵
    1. Wilson JG,
    2. Roth CB, and
    3. Warkany J
    (1953) An analysis of the syndrome of malformations induced by maternal vitamin A deficiency. Effects of restoration of vitamin A at various times during gestation. Am J Anat 92:189–217.
    OpenUrlCrossRefPubMed
  168. ↵
    1. Wong WL,
    2. Su X,
    3. Li X,
    4. Cheung CM,
    5. Klein R,
    6. Cheng CY, and
    7. Wong TY
    (2014) Global prevalence of age-related macular degeneration and disease burden projection for 2020 and 2040: a systematic review and meta-analysis. Lancet Glob Health 2:e106–e116.
    OpenUrlCrossRefPubMed
  169. ↵
    1. Wu S,
    2. Moomaw CR,
    3. Tomer KB,
    4. Falck JR, and
    5. Zeldin DC
    (1996) Molecular cloning and expression of CYP2J2, a human cytochrome P450 arachidonic acid epoxygenase highly expressed in heart. J Biol Chem 271:3460–3468.
    OpenUrlAbstract/FREE Full Text
  170. ↵
    1. Xie Q,
    2. Zhang QY,
    3. Zhang Y,
    4. Su T,
    5. Gu J,
    6. Kaminsky LS, and
    7. Ding X
    (2000) Induction of mouse CYP2J by pyrazole in the eye, kidney, liver, lung, olfactory mucosa, and small intestine, but not in the heart. Drug Metab Dispos 28:1311–1316.
    OpenUrlAbstract/FREE Full Text
  171. ↵
    1. Yanai R,
    2. Mulki L,
    3. Hasegawa E,
    4. Takeuchi K,
    5. Sweigard H,
    6. Suzuki J,
    7. Gaissert P,
    8. Vavvas DG,
    9. Sonoda KH,
    10. Rothe M et al.
    (2014) Cytochrome P450-generated metabolites derived from ω-3 fatty acids attenuate neovascularization. Proc Natl Acad Sci USA 111:9603–9608.
    OpenUrlAbstract/FREE Full Text
  172. ↵
    1. Zenteno JC,
    2. Arce-Gonzalez R,
    3. Matsui R,
    4. Lopez-Bolaños A,
    5. Montes L,
    6. Martinez-Aguilar A, and
    7. Chacon-Camacho OF
    (2023) Clinical-genetic findings in a group of subjects with macular dystrophies due to mutations in rare inherited retinopathy genes. Graefes Arch Clin Exp Ophthalmol 261:353–365.
    OpenUrl
  173. ↵
    1. Zhang T,
    2. Xiang CD,
    3. Gale D,
    4. Carreiro S,
    5. Wu EY, and
    6. Zhang EY
    (2008) Drug transporter and cytochrome P450 mRNA expression in human ocular barriers: implications for ocular drug disposition. Drug Metab Dispos 36:1300–1307.
    OpenUrlAbstract/FREE Full Text
  174. ↵
    1. Zhang X,
    2. Alhasani RH,
    3. Zhou X,
    4. Reilly J,
    5. Zeng Z,
    6. Strang N, and
    7. Shu X
    (2021) Oxysterols and retinal degeneration. Br J Pharmacol 178:3205–3219.
    OpenUrl
  175. ↵
    1. Zhang Y,
    2. Yao K,
    3. Yu Y,
    4. Ni S,
    5. Zhang L,
    6. Wang W, and
    7. Lai K
    (2013) Effects of 1.8 GHz radiofrequency radiation on protein expression in human lens epithelial cells. Hum Exp Toxicol 32:797–806.
    OpenUrlCrossRefPubMed
    1. Zhao C,
    2. Schwartzman ML, and
    3. Shichi H
    (1996) Immunocytochemical study of cytochrome P450 4A induction in mouse eye. Exp Eye Res 63:747–751.
    OpenUrlCrossRefPubMed
    1. Zhao C and
    2. Shichi H
    (1995) Immunocytochemical study of cytochrome P450 (1A1/1A2) induction in murine ocular tissues. Exp Eye Res 60:143–152.
    OpenUrlCrossRefPubMed
  176. ↵
    1. Zhao L,
    2. Chen XJ,
    3. Zhu J,
    4. Xi YB,
    5. Yang X,
    6. Hu LD,
    7. Ouyang H,
    8. Patel SH,
    9. Jin X,
    10. Lin D et al.
    (2015) Lanosterol reverses protein aggregation in cataracts. Nature 523:607–611.
    OpenUrlCrossRefPubMed
  177. ↵
    1. Zhao Y,
    2. Wang S,
    3. Sorenson CM,
    4. Teixeira L,
    5. Dubielzig RR,
    6. Peters DM,
    7. Conway SJ,
    8. Jefcoate CR, and
    9. Sheibani N
    (2013) Cyp1b1 mediates periostin regulation of trabecular meshwork development by suppression of oxidative stress. Mol Cell Biol 33:4225–4240.
    OpenUrlAbstract/FREE Full Text
  178. ↵
    1. Zheng W,
    2. Reem RE,
    3. Omarova S,
    4. Huang S,
    5. DiPatre PL,
    6. Charvet CD,
    7. Curcio CA, and
    8. Pikuleva IA
    (2012) Spatial distribution of the pathways of cholesterol homeostasis in human retina. PLoS One 7:e37926.
    OpenUrlCrossRefPubMed
PreviousNext
Back to top

In this issue

Drug Metabolism and Disposition: 51 (10)
Drug Metabolism and Disposition
Vol. 51, Issue 10
1 Oct 2023
  • Table of Contents
  • Table of Contents (PDF)
  • About the Cover
  • Index by author
  • Editorial Board (PDF)
  • Front Matter (PDF)
Download PDF
Article Alerts
Sign In to Email Alerts with your Email Address
Email Article

Thank you for sharing this Drug Metabolism & Disposition article.

NOTE: We request your email address only to inform the recipient that it was you who recommended this article, and that it is not junk mail. We do not retain these email addresses.

Enter multiple addresses on separate lines or separate them with commas.
Challenges and Opportunities in P450 Research on the Eye
(Your Name) has forwarded a page to you from Drug Metabolism & Disposition
(Your Name) thought you would be interested in this article in Drug Metabolism & Disposition.
CAPTCHA
This question is for testing whether or not you are a human visitor and to prevent automated spam submissions.
Citation Tools
Review Article50th Anniversary Celebration Collection Special Section on New and Emerging Areas and Technologies in Drug Metabolism and Disposition, Part I—Minireview

P450s in the Eye

Irina A. Pikuleva
Drug Metabolism and Disposition October 1, 2023, 51 (10) 1295-1307; DOI: https://doi.org/10.1124/dmd.122.001072

Citation Manager Formats

  • BibTeX
  • Bookends
  • EasyBib
  • EndNote (tagged)
  • EndNote 8 (xml)
  • Medlars
  • Mendeley
  • Papers
  • RefWorks Tagged
  • Ref Manager
  • RIS
  • Zotero

Share
Review Article50th Anniversary Celebration Collection Special Section on New and Emerging Areas and Technologies in Drug Metabolism and Disposition, Part I—Minireview

P450s in the Eye

Irina A. Pikuleva
Drug Metabolism and Disposition October 1, 2023, 51 (10) 1295-1307; DOI: https://doi.org/10.1124/dmd.122.001072
Reddit logo Twitter logo Facebook logo Mendeley logo
  • Tweet Widget
  • Facebook Like
  • Google Plus One

Jump to section

  • Article
    • Abstract
    • Introduction
    • Conclusions and Practical Suggestions
    • Acknowledgments
    • Authorship Contributions
    • Footnotes
    • ABBREVIATIONS
    • References
  • Figures & Data
  • Info & Metrics
  • eLetters
  • PDF

Related Articles

Cited By...

More in this TOC Section

  • Mass Spectrometry-Based Imaging of Drug Disposition
  • High-Throughput Assays for Investigating CYP-Mediated DDIs
Show more 50th Anniversary Celebration Collection Special Section on New and Emerging Areas and Technologies in Drug Metabolism and Disposition, Part I—Minireview

Similar Articles

Advertisement
  • Home
  • Alerts
Facebook   Twitter   LinkedIn   RSS

Navigate

  • Current Issue
  • Fast Forward by date
  • Fast Forward by section
  • Latest Articles
  • Archive
  • Search for Articles
  • Feedback
  • ASPET

More Information

  • About DMD
  • Editorial Board
  • Instructions to Authors
  • Submit a Manuscript
  • Customized Alerts
  • RSS Feeds
  • Subscriptions
  • Permissions
  • Terms & Conditions of Use

ASPET's Other Journals

  • Journal of Pharmacology and Experimental Therapeutics
  • Molecular Pharmacology
  • Pharmacological Reviews
  • Pharmacology Research & Perspectives
ISSN 1521-009X (Online)

Copyright © 2023 by the American Society for Pharmacology and Experimental Therapeutics