Abstract
Angiogenesis, disruption of the retinal barrier, leukocyte‐adhesion and oedema are cardinal signs of proliferative retinopathies that are associated with vision loss. Therefore, identifying factors that regulate these vascular dysfunctions is critical to target pathological angiogenesis. Given the conflicting role of bioactive lipids reported in the current literature, the goal of this review is to provide the reader a clear road map of what has been accomplished so far in the field with specific focus on the role of polyunsaturated fatty acids (PUFAs)‐derived metabolites in proliferative retinopathies. This necessarily entails a description of the different retina cells, blood retina barriers and the role of (PUFAs)‐derived metabolites in diabetic retinopathy, retinopathy of prematurity and age‐related macular degeneration as the most common types of proliferative retinopathies.
Abbreviations
- AA
arachidonic acid
- AMD
age‐related macular degeneration
- BRB
blood retinal barrier
- CYP
cytochrome P450
- DHA
decosahexaenoic acid
- DHETs
dihydroxyeicosatrienoic acids
- DR
diabetic retinopathy
- EETs
epoxyeicosatrienoic acids
- EPA
eicosapentanoic acid
- HETE
hydroxyeicosatrienoic acids
- IGF‐1
insulin‐like growth factor‐1
- LA
linoleic acid
- LO
lipoxygenases
- PUFAs
polyunsaturated fatty acids
- ROP
retinopathy of prematurity
- RPE
retinal pigment epithelium
Introduction
The purpose of this review is to provide the reader with a clear summary of what has been accomplished so far in the field of bioactive lipids in pathological retinal angiogenesis with specific focus on the role of polyunsaturated fatty acids (PUFAs)‐derived metabolites in proliferative retinopathies. This necessarily entails a description of different retinal cells, blood‐retina barriers and the role of PUFA‐derived metabolites in diabetic retinopathy (DR), retinopathy of prematurity (ROP) and age‐related macular degeneration (AMD)as the most common types of proliferative retinopathies.
Retinal cell layers
The retina is the light‐sensitive part at the back of the eye, which converts the light signals into electrical signals (Figure 1). The retina can be divided anatomically into two parts: (i) central retina (also known as the macula) which is located in adult human retina nearly 3 mm temporal to the optic disc and is about 1.5 mm in diameter with a central depression called the fovea representing the area of maximum visual acuity. The fovea is dependent on choriocapillaries for its blood supply as it is devoid of retinal capillaries, (ii) the remaining part of the retina is called the peripheral retina (Kolb, 1995b). The main cellular components of the retina are the retinal pigment epithelium, the photoreceptors, the interneurons, the ganglion cells, the glial cells and the vascular cells.
Figure 1.

Anatomy of the eye and organization of retinal layers. The eye is divided into anterior and posterior parts. The vitreous and the retina occupy the posterior portion of the eye. Retinal branches of the central retinal artery that enters the eye within the substance of the optic nerve ramify on the vitreal side of the retina to form the retinal circulation. The magnified part is to show that the retina is formed of more than one layer.
The retina is arranged into 10 layers, which are described from outside inward:
Retinal pigment epithelium (RPE): Those are hexagonal shaped cells forming a single layered cuboidal epithelium, which is separated from the choriocapillaries by Bruch's membrane. The tight junctions between this epithelial monolayer constitutes the outer blood‐retinal barrier (BRB), which selectively separates the retinal photoreceptors from choroidal blood (Bonilha, 2014; Rizzolo, 2014).
Photoreceptor layer: The light‐sensitive part of the retina in which the rods are responsible for dark vision (also known as scotopic vision) and the cones which are responsible for colour vision (also known as photopic vision); form a single layer of photoreceptors (Anderson and Fisher, 1976).
Outer limiting membrane: It is formed by junctional complexes of neighbouring Müller cells in addition to the junctional complexes between Müller and photoreceptor cells (Omri et al., 2010).
Outer nuclear layer: It is formed of the nuclei of the photoreceptor cells (Kolb, 1995b).
Outer plexiform layer: It is the site of the synaptic connections between photoreceptor cells and the connecting neurones including the bipolar and horizontal cells (Brzezinski and Reh, 2015).
Inner nuclear layer: It contains the nuclei of the horizontal, the bipolar, the amacrine, the inter‐plexiform and the Müller cells. They have a specific organization where the horizontal cells are closer to the outer plexiform layer; amacrine cells closer to the inner plexiform layer; while the bipolar, inter‐plexiform and Müller cells are in between (Kolb, 1995b).
Inner plexiform layer: It is the site of the synaptic connections between bipolar and amacrine cells in the inner nuclear layer and ganglion cells in the ganglion cell layer (Kolb, 1995a, 1995b).
Ganglion cell layer: It contains ganglion cells along with other cells, including some amacrine cells, pericytes, endothelial cells and astrocytes. The ganglion cells collect the visual information from the retinal photoreceptors and project these data to the parvocellular and magnocellular layers of the lateral geniculate nucleus of the brain. Due to the long distance between the retina and the brain, the axonal transport of the ganglion cells is highly active and requires ATP which is supplied by the active axonal mitochondria (Kolb, 1995a, 1995b).
Nerve fibre layer: Axons of the ganglion cells collect in this layer travelling to the optic nerve head.
Inner limiting membrane: The inner processes of Müller cells expand on the vitreal surface of the retina to form this membrane in which the collagen fibres of the vitreous are inserted. The inner limiting membrane maintains the compartmentation between retina and vitreous and is perforated by new blood vessels that invade the vitreous in proliferative retinopathies (Kolb, 1995a, 1995b).
Retinal glial cells
There are different types of glial cells in the retina including Müller cells, astrocytes and micoglia. Müller cells are the most numerous type of glial cells that exist in the retina (Reichenbach and Bringmann, 2013). Microglial cells are the retinal resident macrophages. They are derived from progenitor cells found in the yolk sac during early embryonic development (Kierdorf et al., 2013; Elmore et al., 2014).
Glial cells are highly important for the proper function of the retina. They display several important functions. They actively participate in the retinal‐blood barrier as their processes surround the blood vessels controlling the movement of different materials across the BRB. Moreover, they form sheaths around neuronal cells of the retina ensuring their interaction and support of the neural retina (Vecino et al., 2016). The ability of Müller cells to regulate the homeostasis of extracellular pH and K+ ions enables them to play a critical role in maintaining the integrity of the BRB. Müller cells and astrocytes regulate the extracellular space volume, metabolites clearance, K+ buffering and calcium‐related signalling, via the aquaporins, particularly aquaporin 4, which are located in their cell membranes (Lan et al., 2017).
Interestingly, glial cells are also involved in lipid metabolism. Thus, Müller glial cells may have a role in the intra‐retinal transport of lipids (Tserentsoodol et al., 2006). Also, Müller cells take up docosahexaenoic acid (DHA) (Gordon and Bazan, 1990), which is incorporated into glial phospholipids and finally transported to the photoreceptors (Politi et al., 2001). In addition, glial cells produce extracellular matrix (ECM) which contributes to the inner limiting membrane, providing a significant support for the retina (Clark et al., 2011; Keenan et al., 2012). These ECM components have a role in retinal angiogenesis as well (Jiang et al., 1994; Keenan et al., 2012).
One of the most important factors released by Müller cells is VEGF. Although basal levels of VEGF are neuroprotective, higher levels of VEGF are associated with vascular disorders (Zheng et al., 2012). VEGF can induce neuronal regeneration and angiogenesis after ischaemia/reperfusion injury (Ma et al., 2011) via direct activation of both neurons and endothelial cells (Oosthuyse et al., 2001). Apart from VEGF, glial cells synthesize other neurotrophic factors including pigment epithelium‐derived factor (PEDF) (Zhou et al., 2009; Unterlauft et al., 2012), brain‐derived neurotrophic factor (Dai et al., 2012), ciliary neurotrophic factor (Escartin et al., 2007) and glial cell‐derived neurotrophic factor (Harada et al., 2003; Hauck et al., 2006).
Retinal microglia may contribute to retinal inflammation, cell death and cell survival via their released cytokines (Schafer et al., 2013). Microglial cells are the major cellular component of the innate immunity system in the retina, and their activation is strictly regulated by different inhibitory pathways (Copland et al., 2010; Chen et al., 2012). Activated retinal microglia secrete mediators that affect the neural retina leading to increased neuronal apoptosis (Altmann and Schmidt, 2018). Hyperglycaemia and glycated albumin are among the main factors that activate retinal microglia during diabetes (Ibrahim et al., 2011). Prolonged activation of microglial cells in diabetic retina tends to be proinflammatory rather than anti‐inflammatory and thus can generate a state of chronic inflammation that may be related to retinal affection during DR (Arroba and Valverde, 2017). Prolonged stimulation of the diabetic retina by damage‐associated molecular patterns leads to dysregulation of the retinal innate immune system, which may contribute to development of DR. In the progressive stages of DR, recruited immune cells infiltrating the retina can participate in retinal chronic inflammation with subsequent retinal vascular and neuronal damage (Xu and Chen, 2017).
Blood supply of the retina
The retina has a double blood supply: the outer retina (RPE and the photoreceptors) is supplied by the choroidal circulation, while the inner retina is supplied by the retinal circulation. However, the fovea and the extreme peripheral retina are avascular and receive their nutrition via diffusion from the choroidal circulation (Delaey and Van De Voorde, 2000). The retinal capillaries are arranged in a laminar meshwork to guarantee satisfactory perfusion to all retinal cells. Retinal capillaries are formed of endothelial cells surrounded by pericytes at a ratio of 1:1 (Shin et al., 2014).
Retinal barriers
The BRB is divided into an inner and an outer BRB. The barrier is formed by tight junctions of endothelial cells, basal lamina and end‐feet of astrocytes (Yao et al., 2014). The inner BRB is formed by the tight junctions between the non‐fenestrated retinal endothelial cells, which are surrounded by the processes of pericytes and Müller cells. Retinal endothelium, pericytes and Müller cells together form the inner BRB which is critical for the nutrition and protection of the inner two‐thirds of the retina (Hosoya and Tachikawa, 2012). On the other hand, the outer BRB, which is important for homeostatic maintenance of the outer third of the retina, consists of the tight junctions of retinal pigment epithelial cells (Cunha‐Vaz et al., 2011; Campbell and Humphries, 2012). The barrier prevents water‐soluble molecules from entering the retina. The fenestrated choriocapillaris is permeable to macromolecules and does not appear to contribute importantly to the blood‐retinal barrier. Bruch's membrane, situated between the choriocapillaris and RPE, represents a diffusion barrier only to macromolecules. A greater oncotic pressure in the choroid than in the retina is generated due to the high protein permeability of the choriocapillaris which results in fluid absorption from the retinal extracellular spaces into the choroid. The blood retinal barrier is affected in retinal pathologies (Cunha‐Vaz et al., 2011; Campbell and Humphries, 2012).
Pathological states of retinal angiogenesis
A delicate balance between proangiogenic and antiangiogenic factors regulates retinal vascular haemostasis. Retinal hypoxia, ischaemia or inflammation, may disturb this balance to induce pathological neovascularization in DR, ROP and AMD (Campochiaro et al., 2016) (Figure 2). Pathological retinal and choroidal angiogenesis lead to vascular leakage, bleeding and fibrosis and ultimately significant visual impairment. Therefore, it is important to investigate the underlying cellular and molecular mechanisms of pathological retinal neovascularization (RNV) to identify new therapies to save and improve the vision in patients with DR, AMD or ROP (Dreyfuss et al., 2015; Campochiaro et al., 2016).
Figure 2.

General mechanisms of pathological RNV. Retinal hypoxia is a key factor in mediating pathological RNV in AMD, proliferative diabetic retinopathy (PDR) and ROP. Retinal hypoxia develops due to accumulation of drusen and RPE dysfunction in AMD, and capillary degeneration in PDR and ROP. Retinal hypoxia induces HIF1α‐dependent transcription of several proinflammatory and proangiogenic factors such as IL6, the chemokines CXCL12 and CCL2, inducible NO synthase (iNOS), VEGF and IGF‐1. This ultimately causes development of new blood vessels or RNV. HIF, hypoxia inducible factor.
Diabetic retinopathy
DR is a neurovascular complication affecting the retina of diabetic patients and remains one of the most common causes of blindness, worldwide. World Health Organization has added diabetic retinopathy on the priority list of eye conditions that should be prevented or treated. On a global level, there were approximately 285 million diabetes patients in 2010 and nearly 100 million of those exhibited signs of diabetic retinopathy (Lee et al., 2015).
There is a strong association between chronic hyperglycaemia during diabetes and the development of DR, although the underlying mechanisms of this association are still ill‐defined. A number of biochemical pathways have been suggested as possible links between hyperglycaemia and diabetic retinopathy, including dysregulation of the polyol pathway, accumulation of advanced glycation end products, activation of PKC, increased expression of growth factors as VEGF and insulin‐like growth factor‐1 (IGF‐1), haemodynamic alterations, oxidative stress, ER stress, activation of the renin‐angiotensin‐aldosterone system, chronic inflammation and increased adhesion of leucocytes to the endothelium (Cheung et al., 2010).
Clinically, DR is classified into two types: non‐proliferative and proliferative. Non‐proliferative DR is characterized by microaneurysms (the earliest sign, reflecting the pericyte loss and consequent weakness of the micro‐vessels walls), micro‐haemorrhage, macular oedema (the main cause of visual impairment), vascular tortuosity and beading. On the other hand, proliferative DR is characterized by retinal angiogenesis, vitreous neovascularization, haemorrhage or retinal detachment (Duh et al., 2017; Iwase et al., 2017).
The development of RNV in DR results from enhanced apoptosis of pericytes and endothelial cells and subsequent capillary degeneration that creates relative hypoxia. This leads to up‐regulation of the key angiogenic factor VEGF.
The current treatment modalities of DR include tight glycaemic control, anti‐VEGF therapies, laser photocoagulation, corticosteroids and surgical removal of the vitreous. Tight glycaemic control is effective in reducing incidence of DR and progression (Diabetes et al., 2015). The long‐term beneficial effect of glycaemic control has been examined by two large studies: The Diabetes Control and Complications Trial (DCCT) in Type 1 diabetes (Diabetes et al., 2015, 1993) and the United Kingdom Prospective Diabetes Study (UKPDS) in Type 2 diabetes (UKPDS Group, 1998). Both DCCT and the UKPDS have demonstrated that tight glycaemic control that maintains the levels of HbA1c ≤7% reduced development and progression of DR, with the beneficial effects of tight glycaemic control persisting up to 10–20 years.
The gold standard for the treatment of DR is still laser photocoagulation. This procedure is used to treat macular oedema in non‐proliferative DR. In proliferative DR, pan‐retinal photocoagulation is used where the laser is applied to the whole retina, except for the macula (Evans et al., 2014).
The introduction of intravitreal injection of anti‐VEGF therapies has changed the goal of DR treatment from stabilization to improvement of vision. Anti‐VEGF therapies significantly improve the visual acuity of patients with diabetic macular oedema. There are four FDA approved anti‐VEGF drugs that have been used in the treatment of DR. The first is bevacizumab (with the trade name Avastin, from Genentech). It is a recombinant humanized anti‐VEGF monoclonal antibody. The second drug is ranibizumab (trade name is Lucentis, also from Genentech), which is a recombinant antibody fragment (Fab) against VEGF‐A. The third anti‐VEGF drug is aflibercept (trade name is Eylea or known also as VEGF Trap‐Eye, from Regeneron), which is acting as a VEGF‐receptor capable of binding to both circulating VEGF‐A and VEGF‐B. It has VEGF‐binding domains of VEGF receptors 1 and 2. The fourth drug is pegaptanib (Macugen, from Valeant,), which binds specifically to the main pathological VEGF isomer in the eye, which is the VEGF‐A165 (Osaadon et al., 2014; Vaziri et al., 2015).
Another treatment, intravitreal injection of synthetic corticosteroids including triamcinolone acetonide (Bressler et al., 2013), dexamethasone (Callanan et al., 2013) and fluocinolone (Pearson et al., 2011) has been associated with reduced progression of DR, particularly when combined with other lines of treatment (photocoagulation or anti‐VEGF). However, this procedure was associated with adverse effects such as cataract formation or glaucoma progression. But they were less associated with vitreous haemorrhage, retinal detachment or possibility of endophthalmitis (Sampat and Garg, 2010).
Lastly, the surgical removal of the vitreous (vitrectomy) is used in proliferative DR when vitreous haemorrhage or retinal detachment occurs. Vitrectomy improved visual acuity in patients suffering from diabetic macular oedema, with a low rate of postoperative complications (Diabetic Retinopathy Clinical Research Network Writing et al., 2010).
Taken together, the pathogenesis of DR is complex and multifactorial, which provide a strong rationale to use more than one line of treatment in the same patient to achieve a better response. Studies have shown that when intravitreal corticosteroids (Avitabile et al., 2005; Kang et al., 2006; Lam et al., 2007; Gillies et al., 2011) or anti‐VEGF (Nguyen et al., 2009; Elman et al., 2011; Lee et al., 2011; Mitchell et al., 2011) were used as a combined therapy along with retinal laser photocoagulation, the outcome was better with a more improved visual acuity in those patients who received the combined therapy. However, these therapies are invasive and directed mainly towards late stages of DR, after significant and irreversible damage has occurred. Hence, preventative strategies that address early pathological events and neuronal dysfunction are highly desirable. These therapeutic limitations have generated the need for novel therapeutic interventions.
Retinopathy of prematurity
ROP is a multifactorial neovascular disease that affects premature infants and leads to childhood blindness worldwide. The disease was first described in the 1940s and was attributed to the use of oxygen to resuscitate preterm infants (Terry, 1946; Patz et al., 1953; Ashton et al., 1954). Several clinical studies showed a significant decrease in the severity and incidence of ROP by limiting the use of oxygen in premature infants (Tin et al., 2001; Tin and Wariyar, 2002; Chow, Wright, Sola, and Group, 2003; Anderson et al., 2004).
Development of ROP occurs in two distinctive stages. The first stage begins with the oxygen‐induced impairment of normal retinal vascular development and capillary degeneration. This stage is accompanied by a significant decrease in retinal levels of VEGF and IGF‐1 suggesting that administration of VEGF or IGF1 during this stage could inhibit the development of the second neovascular stage (Smith, 2004; Villegas‐Becerril et al., 2006). Capillary degeneration and vaso‐obliteration that occurs during the first stage of the disease and the increased metabolic activity of the developing retina leads to a relative retinal hypoxia. The developing hypoxia prevents hypoxia inducible factors from degradation by prolyl hydroxylases leading to up‐regulation of number of downstream pro‐angiogenic genes including VEGF, angiopoietins and erythropoietin (Smith, 2004; Heckmann, 2008) (Figure 2). Thus, interruption of hypoxia‐induced signalling has been accepted as a therapeutic strategy to treat and prevent the progression of ROP. This includes the use of anti‐VEGF therapy.
To investigate the underlying molecular and cellular mechanisms of retinal neovascularization in ROP and, in turn, to identify potential therapeutic targets, several animal models have been developed. These models collectively known as oxygen‐induced retinopathy models (OIR). In a mouse model of OIR, newborn mice are exposed to high oxygen levels (75% O2) at postnatal day 7 (P7) for 5 days, during which retinal vaso‐obliteration occurs. This is followed by exposure to normal room oxygen level for additional 5 days during which pathological retinal neovascularization develops due to retinal hypoxia (Smith et al., 1994). In the rat model of OIR, pups are subjected to fluctuating levels of oxygen (Ricci, 1990; Ventresca et al., 1990; Zhang et al., 2000). The limitation of animal models of OIR is the use of pups but not preterm animals. Although the rat model of OIR is more representative of clinical changes in ROP, the mouse model of OIR is used more frequently than the rat model because it allows for the use of mice with specific knockout mutations.
Laser therapy to the avascular retina is the standard treatment for ROP. However, laser therapy has therapeutic limitations including, but not limited to, the risk of general anaesthesia to the premature infants (Filippi et al., 2013; Attachoo et al., 2014). Targeting angiogenic factors such as the VEGF (anti‐VEGF agents) either alone or combined with laser therapy carries the promise of optimizing the vision improvement in ROP. Bevacizumab is a recombinant humanized monoclonal antibody directed against the VEGF. The Bevacizumab Eliminates the Angiogenic Threat of Retinopathy of Prematurity (ROP) study was a prospective, randomized, stratified, controlled, multicenter study (Mintz‐Hittner et al., 2011; Stahl, 2018) and the results suggested that inhibition of VEGF with bevacizumab might be beneficial in preventing retinal neovascularization. However, more studies are needed to make sure there are no long‐term side effects, particularly as VEGF plays a role as retinal neurotrophic factor (Park et al., 2014; Li et al., 2016). It is important to know anti‐VEGF therapy has a wider effect on the pathophysiology of ROP and may account for the late failures.
There are several other emerging therapeutic approaches including and not limited to the use of antioxidants. Our research has demonstrated that inhibition of NADPH oxidase as an approach to inhibit oxidative stress could be helpful in prevention of retinal neovascularization (Al‐Shabrawey et al., 2005). Furthermore, our recent studies have focused on 12/15‐lipoxygenase as a potential therapeutic target to prevent retinal neovascularization in the mouse model of OIR (Al‐Shabrawey et al., 2011). Moreover, ω‐3 fatty acids that can reduce phase 1 and phase 2 in the mouse OIR model are being investigated in human infants (Fu et al., 2015; Malamas et al., 2017).
Age‐related macular degeneration
AMD is a disease that affects primarily the macula and accounts for 7% of legal blindness worldwide (Bressler et al., 2003; Bourne et al., 2018). The increased ageing of the population worldwide is a key factor in increasing the number of patients with AMD which is expected to reach 196 million and 288 million by 2020 and 2040 respectively (Wong et al., 2014). This significant number represents a major progressive burden on health care systems.
The early stage of AMD is characterized by accumulation of drusen, underneath the RPE (Garcia‐Layana et al., 2017) followed by progression to the advanced stages with geographic atrophy and choroidal neovascularization (CNV). The pathological features of geographic atrophy include gradual loss of RPE cells, photoreceptors and the choriocapillaris (Holz et al., 2014). The neovascular AMD is characterized by CNV in which aberrant vessels grow from the choriocapillaris and invade the retina. The new vessels are susceptible to vascular leakage, bleeding and fibrosis leading to formation of macular scars and rapid loss of central vision compared to the atrophic or geographic AMD (Ferris et al., 1984).
The pathogenesis of AMD is multifactorial in which RPE dysfunction, inflammation, oxidative stress and angiogenic factors play essential roles. Age‐dependent impairment of the phagocytic and barrier function of RPE leads to accumulation of cytotoxic lipofuscin, extracellular debris (drusen) and CNV (Figure 2). Therefore, current and future therapies are based on targeting the inflammatory, oxidative stress and angiogenic signalling that drive pathological and functional changes in AMD (Cabral et al., 2017; Hernandez‐Zimbron et al., 2018).
Antiangiogenic drugs, laser therapy, vitamins and surgery alone or in combination have all been adopted as therapeutic strategies to improve vision in AMD.
Treatment of dry non‐vascular AMD: according to the Age‐Related Eye Disease Study trial, antioxidant vitamins such as Vitamins C, E, A and lutein plus zinc were suggested for patients with dry non‐vascular AMD (Age‐Related Eye Disease Study Research, 2001a, 2001b).
Treatment of wet neovascular AMD: Effective treatment for wet neovascular AMD includes intravitreous injection of anti‐VEGF therapies, photodynamic therapy (PDT) and supplementation with antioxidant vitamins and zinc. Anti‐VEGF therapies such as bevacizumab, ranibizumab or aflibercept are strongly recommended for patients with neovascularization and PTD is suggested as an alternative for patients who cannot receive intravitreal injection with anti‐VEGF (Bandello et al., 2017; Hernandez‐Zimbron et al., 2018; Su et al., 2018).
Bioactive lipids
Lipids and their metabolites are not only components of cellular membranes but also they act as signalling molecules and are thus known as ‘bioactive lipids’. Among the key activities exerted by bioactive lipids are angiogenesis, regulation of inflammation and maintenance of homeostasis. Bioactive lipids cover a large number and variety of compounds and the most commonly ones are those derived from polyunsaturated fatty acids (PUFAs).
PUFA‐derived metabolites
These metabolites are produced in a two‐step reaction sequence. The first step is carried out by the action of PLA2 enzyme which releases PUFAs such as linoleic acid (LA), arachidonic acid (AA) which is the most common, DHA and eicosapentanoic acid (EPA) from the cell membrane lipid layers. Those released PUFAs undergo the second step to be metabolized either via COX enzymes, cytochrome P450 (CYP) enzyme or lipoxygenases (LO) enzymes. There is much evidence that metabolites of the ω‐6 PUFA, such as AA, via the three major enzymic pathways produce pro‐inflammatory and pro‐angiogenic effects. These effects are mostly opposed by those of the metabolites from the ω‐3 PUFAs, DHA and EPA (Figures 3 and 5).
COX‐derived metabolites
Figure 3.

(A) ω‐6 (arachidonic acid; AA)‐derived bioactive lipids or eicosanoids. Activation of PLA2 releases AA from the cell membrane phospholipids. AA is metabolized by COX to PGs, by CYP450 to EETs and by LO to HETEs (5, 12, 15‐HETEs), leukotrienes and lipoxins. (B) ω‐3‐derived bioactive lipids. PLA2 releases the ω‐3 polyunsaturated acids, DHA and EPA from the cell membrane phospholipids. DHA and EPA are converted by CYP450 to 19, 20‐EDP and 17, 18‐EEQ, by COX to PGE3, PGI3, TXA3 and LTB5 and by lipoxygenases to lipoxins, resolvins, neuroprotectins and LTB5.
Figure 5.

The pro‐angiogenic versus anti‐angiogenic effect of lipoxygenase and CYP450 metabolites. Metabolites of 12/15‐LO from AA such as 12‐ or 15‐HETE are primarily pro‐angiogenic while the metabolites from DHA and EPA such as lipoxins, resolvins and neuroprotectins are primarily anti‐angiogenic. Similarly, the metabolites formed by CYP450 from AA such as 11‐ and 12‐EET elicit pro‐angiogenic effects while its products from EPA or DHA, such as 17, 18‐EEQ or 19, 20‐EDP respectively, induce anti‐angiogenic effects.
There are two main COX isoforms, COX‐1 and COX‐2. Both convert AA into PGH2, which is then converted by appropriate synthases, into PGs and TXs which are collectively known as prostanoids (Sales et al., 2008). The most important prostanoids are PGE2, PGF2α, PGD2, PGI2 and TxA2 (Figure 3). PGE2 is a key proinflammatory PG which has anti‐apoptotic properties with important functions in the immunity and cancer development. PGF2α is an autocrine growth factor related to endometrial carcinoma (Sales et al., 2008; Woodward et al., 2011). TxA2 (synthesized by platelets) and PGI2 (synthesized by endothelium) (Catella‐Lawson et al., 1999) have important roles in both the physiology and pathology of blood vessels. TxA2 causes platelet aggregation and vasoconstriction, while PGI2 has opposing effects (Caughey et al., 2001a, 2001b; Meyer‐Kirchrath et al., 2004). Interestingly, PG signalling is involved in chronic inflammatory diseases, for instance, asthma or Crohn's disease. This may happen via their ability to enhance the release of proinflammatory cytokines, their ability to activate proinflammatory T cells or macrophages (Claar et al., 2015; Wallace, 2018). In addition to COX‐derived proinflammatory metabolites from AA, COX converts ω‐3 PUFA, DHA and EPA to other anti‐inflammatory metabolites such as PGE3, and PGI3.
Cytochrome P450‐derived metabolites
In the linear metabolism of AA, cytochrome P450s (CYP450) metabolize this PUFA to oxidized eicosanoids such as epoxyeicosatrienoic acids (EETs; 5,6‐EET, 8,9‐EET, 11,12‐EET and 14,15‐EET), dihydroxyeicosatrienoic acids (DHETs) and 20‐hydroxyeicosatetraenoic acid (20‐HETE). EETs are further metabolized by soluble epoxide hydrolase enzyme (sEH) into DHETs (Huang et al., 2016). In addition to AA, CYP450 also metabolizes docosahexaenoic acid (DHA) and eicosapentaenoic acid (EPA) into epoxydocosapentaenoic acids (EDPs) and epoxyeicosatetraenoic acids (EEQs) respectively. CYP450‐derived eicosanoids are formed in a tissue and cell‐specific way with many biological functions. Interestingly, EETs and 20‐HETE have opposing actions within the vasculature. The vascular role of CYP‐derived metabolites has been initially linked to regulation of vascular tone via endothelium‐derived hyperpolarizing factor (Campbell et al., 1996). EETs have powerful vasodilatory effect while 20‐HETE is a strong vasoconstrictor. Both EDPs and EEQs exhibit potent vasodilatory and cardioprotective effects (Konkel and Schunck, 2011).
CYP450‐derived eicosanoids have been also linked to angiogenesis. However, the role of the various CYP450 isoforms and their metabolites is complicated by evidence that suggests metabolites of CYP450 from AA elicit pro‐angiogenic effects, while its metabolites from DHA and EPA are anti‐angiogenic (Yanai et al., 2014).
Lipoxygenase‐derived metabolites
The second arm of linear metabolism of PUFAs is carried out by lipoxygenases (LOs), which are non‐haem iron‐containing lipid peroxidizing enzymes which catalyse dioxygenation of PUFAs, forming a wide variety of bioactive lipids (Kuhn et al., 2015). The phylogenetic distribution of LOs demonstrates their wide distribution across both the plant and animal kingdoms. The human genome contains one functional LO gene located on chromosome 10 (ALOX5) and five genes located on chromosome 17 (ALOX15, ALOX15B, ALOX12, ALOX12B, ALOXE3), which encode for six different LO isoforms (Funk et al., 2002). The ALOX15 gene encodes for 12/15‐LO, the ALOX15B gene encodes for 15‐LO2, the ALOX12 gene encodes for the platelet‐type 12‐LO, the ALOX12B gene encodes for a 12R‐LO enzyme, the ALOXE3 gene encodes for two epidermal LO isoforms and finally, the ALOX5 gene encodes for 5‐LO enzyme. The corresponding mouse genes were located on chromosomes 6 (Alox5) and 11 (other Lo isoforms). These genes encode for slightly different LO enzymes in mice (Kinzig et al., 1999; Meruvu et al., 2005; Yu et al., 2006). Mammalian LO isoforms have diverse biological functions. Different LO isoforms are involved in the synthesis of RBCs, differentiation of epidermis and atherosclerosis, via their ability to oxidize esterified membrane lipids and thus to modify their structural and functional propertties (Chen et al., 1994; Sun and Funk, 1996; Kinzig et al., 1999; Capra et al., 2013). LOs affect different cellular functions via their ability to modify the cellular redox status. LOs form lipid peroxides which change the intracellular redox balance and are implicated in the induction of oxidative stress (Othman et al., 2013; Elmasry et al., 2018).
A number of LO derived‐bioactive lipids and some LO isoforms have been associated with the pathogenesis of multiple metabolic disorders including diabetes both type 1 and type 2. Interestingly, 12‐hydroxyeicosatrienoic acids (12‐HETE) derived from either ALOX15 or ALOX12 and ALOX5 derived‐leukotriene B4 were elevated in diabetic patients (Issan et al., 2013). Particularly for ALOX15, there is good evidence to connect the enzyme to the pathogenesis of diabetes (Bleich et al., 1998, 1999; Laybutt et al., 2002; Chen et al., 2005a; Nunemaker et al., 2008; Sears et al., 2009). Expression of ALOX15 was significantly increased in both cell culture and animal models of diabetes. This may be related in part to LO‐induced oxidative stress (Dobrian et al., 2011; Zhang et al., 2013). In general, 12/15‐LO and 5‐LO convert AA to pro‐inflammatory and pro‐angiogenic metabolites such as 12‐ and 15‐HETEs and leukotrienes (Lee et al., 2004; Al‐Shabrawey et al., 2011; Faulkner et al., 2015; Ibrahim et al., 2017).
Linoleic acid (LA), arachidonic acid (AA), EPA and DHA are possible substrates for ALOX15. The enzyme binding site for the fatty acid substrates is a hydrophobic pocket close to the non‐haem iron into which the substrate slides with its methyl end in front (Kuhn et al., 1986; Rickert and Klinman, 1999). Phospholipids (Schewe et al., 1975) and cholesterol esters (Belkner et al., 1991) containing polyunsaturated fatty acids are also possible substrates to ALOX15. The ability of ALOX15 to oxygenate phospholipids was not expected, because the phospholipid molecule is too big to fit in the binding site of ALOX15. This can only be explained if the enzyme shows a high degree of structural flexibility, allowing the active site to rearrange, in order to facilitate phospholipid binding. Moreover, membrane lipids, ester lipids of low‐density lipoproteins and lipoproteins were reported to be additional substrates for ALOX15 (Kuhn et al., 1990; Belkner et al., 1993). This activity of ALOX15 suggested a mechanism for explaining how the bio‐membranes are reformed during the process of maturational degradation of cellular organelles (van Leyen et al., 1998). When LA is the ALOX15 substrate, the main products of the enzymic reaction are 13‐HODE or 9‐HODE. When arachidonic acid is the substrate, ALOX15 produces 15‐HETE or 12‐HETE. These different products are due to the dual positional specificity of ALOX15. Furthermore, 12/15‐LO metabolizes EPA and DHA to lipoxins and resolvins that exhibt anti‐inflammatory and anti‐angiogenic effects (Jin et al., 2009).
Bioactive lipids and retinopathies
Bioactive lipids such as PUFAs and their derivatives were reported to have a role in the pathogenesis of retinopathies including ROP, AMD and DR.
A significant reduction of retinal angiogenesis in ROP has been reported by dietary uptake of ω‐3 PUFAs which was associated with reduced retinal‐ER stress (Fu et al., 2015). Additionally, 12/15‐LO (ALOX15) and CYP are involved in pathological RNV in the OIR and hence, inhibition of 12/15‐LO or CYP significantly reduced RNV in an OIR model (Al‐Shabrawey et al., 2011; Capozzi et al., 2014). Additional results were obtained with bovine retinal endothelial cells, highlighting the role of CYP450 and its EET derivatives in retinal angiogenesis (Michaelis et al., 2008). CYP450‐derived EETs are further metabolized by sEH. Interestingly, mice lacking sEH showed defective angiogenesis via dysregulating Notch signalling, particularly in Müller cells (Hu et al., 2014).
In early stages of AMD, lipid‐containing deposits, called drusen, appear in Bruch's membrane underlying the RPE, suggesting a role of lipids in the pathogenesis of AMD (Shen et al., 2016). Earlier studies demonstrated that ω‐3 PUFAs, such as EPA, reduced CNV associated with AMD. Additionally, ω‐3 PUFAs such as EPA and DHA protected photoreceptors against oxidative stress (Sala et al., 2010). These studies highlight the protective role of ω‐3 PUFAs against AMD development and other pathological RNV as well (Connor et al., 2007; Koto et al., 2007). The protective effect was suggested to be related to 5‐LO‐dependent oxidation of DHA to 4‐hydroxy‐decosahexaenoic acid which inhibits endothelial proliferation and angiogenesis in a PPARγ‐dependent mechanism (Sapieha et al., 2011). Interestingly, in patients with wet‐AMD, a combined therapy of anti‐VEGF and ω‐3 PUFAs supplementation was associated with a greater reduction of VEGF levels in the vitreous, than with anti‐VEGF alone (Rezende et al., 2014). Furthermore, eyes derived from AMD donors had a lower level of very long chain PUFAs than those from normal donors, supporting the notion of the protective effect of PUFAs in AMD (Gorusupudi et al., 2016). On the other hand, oxidized phospholipids were reported to be increased in the eyes of AMD patients and to induce CNV in mice (Suzuki et al., 2007). Secondly, the sphingolipids, including ceramide (Cer), sphingosine (Sph) or sphingosine‐1‐phosphate (S1P) have been implicated in AMD, as well. Although S1P and Cer‐1‐phosphate appear to have a protective effect on photoreceptors and are involved in their differentiation and development (Miranda et al., 2009; Miranda et al., 2011), others showed that S1P can induce angiogenesis in either the choroid or the retina (Caballero et al., 2009; Xie et al., 2009). Lastly, accumulated cholesterol in retinal endothelial cells, macrophages and RPE cells is involved in the pathogenesis of AMD (Omarova et al., 2012; Saadane et al., 2014).
In DR, diabetes induces specific changes in retinal lipid metabolism. Diets rich in beneficial lipids, such as DHA or EPA, have been suggested as an alternative therapeutic strategy to prevent DR (Connor et al., 2007; Tikhonenko et al., 2010). DHA expresses anti‐inflammatory effects and it protected against pathological angiogenesis of the retina (Chen et al., 2005b; Connor et al., 2007). On the contrary, 5‐LO (ALOX5) and 12/15‐LO (ALOX15) both may have a role in the pathogenesis of DR. However, the findings suggested a differential role of the two enzymes. Retinas of diabetic 5‐Lo −/− mice and not 12/15‐Lo −/− showed less degeneration of retinal capillaries after 9 months of diabetes while retinas of both diabetic 5‐Lo −/− and 12/15‐Lo −/− showed less leucocyte adhesion at 3 months (Gubitosi‐Klug et al., 2008). Moreover, 12/15‐LO‐derived bioactive lipids were significantly increased in the vitreous of patients with proliferative DR (Al‐Shabrawey et al., 2011) and in epiretinal membranes as well (Augustin et al., 1997).
ALOX15 has been associated with regulation of endothelial permeability which contributes to the pathogenesis of vascular diseases including DR. 15S‐HETE, the major ALOX15 metabolite of AA in humans, increased endothelial barrier permeability via phosphorylation of zonula occludens‐1 and ‐2, which leads to disruption of the tight junctions causing a decrease in the integrity of the endothelial barrier and increasing the permeability (Kundumani‐Sridharan et al., 2013; Chattopadhyay et al., 2014; Ibrahim et al., 2015a). Moreover, ALOX15 is involved in disruption of endothelial tight junctions (Kundumani‐Sridharan et al., 2013; Chattopadhyay et al., 2015). We have reported that ALOX15 bioactive lipids induce retinal endothelial cell barrier dysfunction in retinal endothelial cells via NADPH‐oxidase dependent mechanisms, which include inhibition of protein tyrosine phosphatase and activation of the VEGF‐receptor 2 signalling pathway (Othman et al., 2013). The involvement of NADPH oxidase in ALOX15‐induced retinal endothelial barrier dysfunction highlighted the importance of the cellular redox state for the integrity of the endothelial barrier. Other reports suggested for the first time a role of ALOX15 in the process of autophagy, as cells lacking ALOX15 showed dysfunctional autophagy (Morgan et al., 2015).
Several studies by us and others have elaborated a possible role of ALOX15 in the pathogenesis of inflammation in DR and other diseases (Othman et al., 2013; Radmark et al., 2015; Ibrahim et al., 2015a ; Elmasry et al., 2018). Of note, activation of inflammatory signalling has been linked to the development of pathological RNV. The effect of ALOX15 during inflammation depends to a great extent on the substrate, as its products can exhibit both proinflammatory or anti‐inflammatory properties. For instance, with AA/LA as substrates, the main oxygenation products of ALOX15 (15‐HETE, 12‐HETE, 13‐HODE) exhibit pro‐inflammatory activities in various inflammation models (Kuhn, 1996). Reports suggested that the pro‐inflammatory properties of ALOX15 may be in part through affecting the cellular redox state and induction of oxidative stress (Kundumani‐Sridharan et al., 2013; Othman et al., 2013; Chattopadhyay et al., 2015; Ibrahim et al., 2015a; Elmasry et al., 2018). On the other hand, ALOX15 showed anti‐inflammatory properties when the substrates are either DHA or EPA via synthesis of pro‐resolving bioactive lipids that act to resolve inflammation (Sala et al., 2010; Serhan et al., 2015a). During the active process of resolution of inflammation, the pro‐inflammatory mediators are down‐regulated whereas the anti‐inflammatory mediators are up‐regulated. These anti‐inflammatory and anti‐angiogenic products of ALOX15 include lipoxins (Ryan and Godson, 2010), resolvins (Spite et al., 2014), protectins and maresins (Serhan et al., 2015b). These mediators induce some anti‐inflammatory processes including inhibition of leucocyte migration (Freire and Van Dyke, 2013), stabilization of vascular permeability (Ereso et al., 2009), apoptosis of proinflammatory neutrophils (El Kebir and Filep, 2013) and polarization of macrophages (Ohira et al., 2010). In addition to their resolving effects on innate immunity, lipoxins and recolvins elicit anti‐angiogenic effects in the eye through regulating VEGF and VEGFR2 expression and signalling (Wolfe, 1991; Jin et al., 2009).
To study the biological effects of 12/15‐LO metabolites, several pharmacological inhibitors have been developed. However, many of the 12/15‐LO inhibitors used (NDGA, CDC, baicalein) did not show a clear isoform specificity (Rai et al., 2010; Kenyon et al., 2011). Moreover, these inhibitors display a species‐specificity which means some inhibitors that effectively inhibit human ALOX15 may not inhibit orthologous enzymes of other species. In addition, those inhibitors had off‐target effects. 12/15‐LO inhibitors have anti‐oxidative properties and they affect the cellular redox homeostasis. It is difficult to distinguish which of the two functions (LO inhibition or redox homeostasis) is the main cause for the detected biological result (Goswami, 2013; Kim et al., 2013). Therefore, results acquired with these inhibitors must be interpreted with caution. To avoid these problems, the inhibitor studies should always be confirmed by another method, for example, the use of the 12/15‐Lo siRNA or 12/15‐Lo −/− mice.
We have been studying the potential role of 12/15‐LO metabolites in the pathogenesis of retinal vascular dysfunction in ischaemic retinopathies for several years. Our human studies showed significant increases in the vitreous levels of 12‐ and 15‐HETEs in patients with proliferative DR. Furthermore, we reported up‐regulation of retinal 12/15‐LO expression and activity in experimental models of DR and OIR (Al‐Shabrawey et al., 2011). Pharmacological inhibition and genetic deletion of 12/15‐LO significantly reduced RNV in OIR (Al‐Shabrawey et al., 2011) and preserved barrier function in DR (Ibrahim et al., 2015a). Recently, we elaborated the underlying cellular and molecular mechanisms of the pro‐inflammatory and pro‐angiogenic effects of 12/15‐LO in ischaemic retinopathies. We found that increased 12/15‐LO in OIR and DR was localized mainly in retinal vessels (Al‐Shabrawey et al., 2011). This was supported by in vitro studies in which the endothelial, rather than the leucocytic, 12/15‐LO was involved in hyperglycaemia‐induced dysfunction of retinal endothelial cell (RECs) (Ibrahim et al., 2017). Treatment of Müller cells with HETEs, metabolites of 12/15‐LO, up‐regulated the proangiogenic and proinflammatory factors VEGF, IL6 and TNFα but suppressed the expression of the angiostatic and neurotrophic factor PEDF (Al‐Shabrawey et al., 2011; Ibrahim et al., 2015b). On the other hand, they induced ER stress, NADPH‐oxidase‐derived ROS and adhesion molecules in RECs (Ibrahim et al., 2015a; Elmasry et al., 2018). Interestingly, while HETEs did not change the levels of VEGF expression in RECs, they increased the levels of phosphorylated VEGFR2 and inhibited the protein tyrosine phosphatase (PTP) (Othman et al., 2013; Elmasry et al., 2018). Taken together, our data suggest that activation of endothelial 12/15‐LO during ischaemic retinopathies induces two parallel paracrine and autocrine proangiogenic pathways through activation of Müller and RECs respectively. Endothelial‐derived HETEs up‐regulate VEGF and suppress PEDF in Müller cells. On the other hand, they activate VEGFR2 in REC via ER stress/NADPH oxidase‐dependent pathways that involve the dysregulation of PTP to release its inhibitory effect on VEGFR2. These effects are summarized in Figure 4.
Figure 4.

Role of 12/15‐lipoxygenase in retinal neovascularization. Our studies demonstrated increased expression and activity of 12/15‐lipoxygenase in retinal endothelial cells of experimental models of diabetes and oxygen‐induced retinopathy. Endothelial‐derived metabolites of 12/15‐lipoxygenase (12‐ and 15‐HETEs) induce retinal neovascularization via paracrine and autocrine loops that activates Müller and endothelial cells respectively. Activation of Müller cells by HETEs (products of endothelial 12/15‐LO) up‐regulates VEGF and inflammatory cytokines and suppresses the angiostatic factor PEDF. However, activation of retinal endothelial cells by HETEs causes ER stress and activation of NADPH oxidase that lead to activation of VEGFR2 without affecting the endothelial levels of VEGF.
COX‐derived eicosanoids play a regulatory role in VEGF signalling and angiogenesis. While AA metabolites of COX elicit pro‐angiogenic effects, the underlying mechanism by which ω‐3 PUFAs regulate angiogenesis is via decreased synthesis of pro‐angiogenic ω‐6 prostanoids coupled with increased levels of less active ω‐3 prostanoids such as PGE3, PGI3, TXA3 and LTB5 (Figure 3B). Selective inhibition of COX2 has been reported to reduce inflammation and pathological RNV in experimental models of DR, CNV and OIR (Wilkinson‐Berka et al., 2003; Suzuki et al., 2012; Zhang et al., 2016).
Conclusion
Although VEGF plays a central role in the development of DR, AMD and ROP, anti‐VEGF therapies do not totally suppress the pathological RNV. PUFA‐derived metabolites regulate the production of VEGF, oxidative stress and inflammatory cytokines, as well as being involved in the pathophysiology of these diseases. The development of LC–MS assays allows detailed retinal lipidomic analysis in retinal diseases to be a reality. Furthermore, the availability of COX, LO, CYP enzyme inhibitors and transgenic mice permits the study of the functions and dysfunctions of PUFA signalling in proliferative retinopathies. By taking the advantage of these invaluable tools, we need to deepen our understanding of the role of PUFA‐derived metabolites in the pathophysiology of these vision threatening diseases and to clinically evaluate the possible therapeutic use of COX, LO and CYP enzyme inhibitors in these conditions. As shown in Figure 5, the metabolites of these three enzymatic pathways may induce retinal angiogenic and angiostatic effects, depending on the PUFA substrate. If the substrate is AA, most of the metabolites will be pro‐angiogenic, whereas if the substrate is DHA or EPA, the anti‐angiogenic effects will be more dominant. This information is the foundation of dietary supplementation of ω‐3 PUFA as a potential treatment of ischaemic retinopathies to halt the progression of RNV.
Nomenclature of targets and ligands
Key protein targets and ligands in this article are hyperlinked to corresponding entries in http://www.guidetopharmacology.org, the common portal for data from the IUPHAR/BPS Guide to PHARMACOLOGY (Harding et al., 2018), and are permanently archived in the Concise Guide to PHARMACOLOGY 2017/18 (Alexander et al., 2017a, 2017b).
Conflict of interest
The authors declare no conflicts of interest.
Acknowledgements
This work has been supported by the National Eye Institute grant R01EY023315 (MA) and the American Heart Association grant 18CDA34080403 (ASI).
Elmasry, K. , Ibrahim, A. S. , Abdulmoneim, S. , and Al‐Shabrawey, M. (2019) Bioactive lipids and pathological retinal angiogenesis. British Journal of Pharmacology, 176: 93–109. 10.1111/bph.14507.
References
- Age‐Related Eye Disease Study Research G (2001a). A randomized, placebo‐controlled, clinical trial of high‐dose supplementation with vitamins C and E and β‐carotene for age‐related cataract and vision loss: AREDS report no. 9. Arch Ophthalmol 119: 1439–1452. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Age‐Related Eye Disease Study Research G (2001b). A randomized, placebo‐controlled, clinical trial of high‐dose supplementation with vitamins C and E, β‐carotene, and zinc for age‐related macular degeneration and vision loss: AREDS report no. 8. Arch Ophthalmol 119: 1417–1436. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Alexander SPH, Fabbro D, Kelly E, Marrion NV, Peters JA, Faccenda E et al (2017a). The Concise Guide to PHARMACOLOGY 2017/18: Enzymes. Br J Pharmacol 174: S272–S359. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Alexander SPH, Kelly E, Marrion NV, Peters JA, Faccenda E, Harding SD et al (2017b). The Concise Guide to PHARMACOLOGY 2017/18: Other proteins. Br J Pharmacol 174: S1–S16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Al‐Shabrawey M, Bartoli M, El‐Remessy AB, Platt DH, Matragoon S, Behzadian MA et al (2005). Inhibition of NAD(P) H oxidase activity blocks vascular endothelial growth factor overexpression and neovascularization during ischemic retinopathy. Am J Pathol 167: 599–607. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Al‐Shabrawey M, Mussell R, Kahook K, Tawfik A, Eladl M, Sarthy V et al (2011). Increased expression and activity of 12‐lipoxygenase in oxygen‐induced ischemic retinopathy and proliferative diabetic retinopathy: implications in retinal neovascularization. Diabetes 60: 614–624. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Altmann C, Schmidt MHH (2018). The role of microglia in diabetic retinopathy: inflammation, microvasculature defects and neurodegeneration. Int J Mol Sci 19. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Anderson CG, Benitz WE, Madan A (2004). Retinopathy of prematurity and pulse oximetry: a national survey of recent practices. J Perinatol 24: 164–168. [DOI] [PubMed] [Google Scholar]
- Anderson DH, Fisher SK (1976). The photoreceptors of diurnal squirrels: outer segment structure, disc shedding, and protein renewal. J Ultrastruct Res 55: 119–141. [DOI] [PubMed] [Google Scholar]
- Arroba AI, Valverde AM (2017). Modulation of microglia in the retina: new insights into diabetic retinopathy. Acta Diabetol 54: 527–533. [DOI] [PubMed] [Google Scholar]
- Ashton N, Ward B, Serpell G (1954). Effect of oxygen on developing retinal vessels with particular reference to the problem of retrolental fibroplasia. Br J Ophthalmol 38: 397–432. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Attachoo A, Horatanaruang D, Chongarunngamsang W, Lauhsattana S (2014). The incidence and risk factors of apnea in premature infants underwent general anesthesia for cryotherapy or laser photocoagulation for treatment of retinopathy of prematurity at Queen Sirikit National Institute of Child Health. J Med Assoc Thai 97 (Suppl. 6): S26–S32. [PubMed] [Google Scholar]
- Augustin AJ, Grus FH, Koch F, Spitznas M (1997). Detection of eicosanoids in epiretinal membranes of patients suffering from proliferative vitreoretinal diseases. Br J Ophthalmol 81: 58–60. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Avitabile T, Longo A, Reibaldi A (2005). Intravitreal triamcinolone compared with macular laser grid photocoagulation for the treatment of cystoid macular edema. Am J Ophthalmol 140: 695–702. [DOI] [PubMed] [Google Scholar]
- Bandello F, Sacconi R, Querques L, Corbelli E, Cicinelli MV, Querques G (2017). Recent advances in the management of dry age‐related macular degeneration: a review. F1000Res 6: 245. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Belkner J, Wiesner R, Kuhn H, Lankin VZ (1991). The oxygenation of cholesterol esters by the reticulocyte lipoxygenase. FEBS Lett 279: 110–114. [DOI] [PubMed] [Google Scholar]
- Belkner J, Wiesner R, Rathman J, Barnett J, Sigal E, Kuhn H (1993). Oxygenation of lipoproteins by mammalian lipoxygenases. Eur J Biochem 213: 251–261. [DOI] [PubMed] [Google Scholar]
- Bleich D, Chen S, Gu JL, Nadler JL (1998). The role of 12‐lipoxygenase in pancreatic‐cells (review). Int J Mol Med 1: 265–272. [PubMed] [Google Scholar]
- Bleich D, Chen S, Zipser B, Sun D, Funk CD, Nadler JL (1999). Resistance to type 1 diabetes induction in 12‐lipoxygenase knockout mice. J Clin Invest 103: 1431–1436. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bonilha VL (2014). Retinal pigment epithelium (RPE) cytoskeleton in vivo and in vitro . Exp Eye Res 126: 38–45. [DOI] [PubMed] [Google Scholar]
- Bourne RRA, Jonas JB, Bron AM, Cicinelli MV, Das A, Flaxman SR et al (2018). Prevalence and causes of vision loss in high‐income countries and in Eastern and Central Europe in 2015: magnitude, temporal trends and projections. Br J Ophthalmol 102: 575–585. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bressler NM, Bressler SB, Congdon NG, Ferris FL 3rd, Friedman DS, Klein R et al (2003). Potential public health impact of Age‐Related Eye Disease Study results: AREDS report no. 11. Arch Ophthalmol 121: 1621–1624. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bressler SB, Qin H, Melia M, Bressler NM, Beck RW, Chan CK et al (2013). Exploratory analysis of the effect of intravitreal ranibizumab or triamcinolone on worsening of diabetic retinopathy in a randomized clinical trial. JAMA Ophthalmol 131: 1033–1040. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Brzezinski JA, Reh TA (2015). Photoreceptor cell fate specification in vertebrates. Development 142: 3263–3273. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Caballero S, Swaney J, Moreno K, Afzal A, Kielczewski J, Stoller G et al (2009). Anti‐sphingosine‐1‐phosphate monoclonal antibodies inhibit angiogenesis and sub‐retinal fibrosis in a murine model of laser‐induced choroidal neovascularization. Exp Eye Res 88: 367–377. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cabral T, Mello LGM, Lima LH, Polido J, Regatieri CV, Belfort R Jr et al (2017). Retinal and choroidal angiogenesis: a review of new targets. Int J Retina Vitreous 3: 31. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Callanan DG, Gupta S, Boyer DS, Ciulla TA, Singer MA, Kuppermann BD et al (2013). Dexamethasone intravitreal implant in combination with laser photocoagulation for the treatment of diffuse diabetic macular edema. Ophthalmology 120: 1843–1851. [DOI] [PubMed] [Google Scholar]
- Campbell M, Humphries P (2012). The blood‐retina barrier: tight junctions and barrier modulation. Adv Exp Med Biol 763: 70–84. [PubMed] [Google Scholar]
- Campbell WB, Gebremedhin D, Pratt PF, Harder DR (1996). Identification of epoxyeicosatrienoic acids as endothelium‐derived hyperpolarizing factors. Circ Res 78: 415–423. [DOI] [PubMed] [Google Scholar]
- Campochiaro PA, Aiello LP, Rosenfeld PJ (2016). Anti‐vascular endothelial growth factor agents in the treatment of retinal disease: from bench to bedside. Ophthalmology 123: S78–S88. [DOI] [PubMed] [Google Scholar]
- Capozzi ME, McCollum GW, Penn JS (2014). The role of cytochrome P450 epoxygenases in retinal angiogenesis. Invest Ophthalmol Vis Sci 55: 4253–4260. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Capra V, Back M, Barbieri SS, Camera M, Tremoli E, Rovati GE (2013). Eicosanoids and their drugs in cardiovascular diseases: focus on atherosclerosis and stroke. Med Res Rev 33: 364–438. [DOI] [PubMed] [Google Scholar]
- Catella‐Lawson F, McAdam B, Morrison BW, Kapoor S, Kujubu D, Antes L et al (1999). Effects of specific inhibition of cyclooxygenase‐2 on sodium balance, hemodynamics, and vasoactive eicosanoids. J Pharmacol Exp Ther 289: 735–741. [PubMed] [Google Scholar]
- Caughey GE, Cleland LG, Gamble JR, James MJ (2001a). Up‐regulation of endothelial cyclooxygenase‐2 and prostanoid synthesis by platelets. Role of thromboxane A2. J Biol Chem 276: 37839–37845. [DOI] [PubMed] [Google Scholar]
- Caughey GE, Cleland LG, Penglis PS, Gamble JR, James MJ (2001b). Roles of cyclooxygenase (COX)‐1 and COX‐2 in prostanoid production by human endothelial cells: selective up‐regulation of prostacyclin synthesis by COX‐2. J Immunol 167: 2831–2838. [DOI] [PubMed] [Google Scholar]
- Chattopadhyay R, Dyukova E, Singh NK, Ohba M, Mobley JA, Rao GN (2014). Vascular endothelial tight junctions and barrier function are disrupted by 15(S)‐hydroxyeicosatetraenoic acid partly via protein kinase C epsilon‐mediated zona occludens‐1 phosphorylation at threonine 770/772. J Biol Chem 289: 3148–3163. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chattopadhyay R, Tinnikov A, Dyukova E, Singh NK, Kotla S, Mobley JA et al (2015). 12/15‐Lipoxygenase‐dependent ROS production is required for diet‐induced endothelial barrier dysfunction. J Lipid Res 56: 562–577. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- Chen M, Yang ZD, Smith KM, Carter JD, Nadler JL (2005a). Activation of 12‐lipoxygenase in proinflammatory cytokine‐mediated beta cell toxicity. Diabetologia 48: 486–495. [DOI] [PubMed] [Google Scholar]
- Chen M, Zhao J, Luo C, Pandi SP, Penalva RG, Fitzgerald DC et al (2012). Para‐inflammation‐mediated retinal recruitment of bone marrow‐derived myeloid cells following whole‐body irradiation is CCL2 dependent. Glia 60: 833–842. [DOI] [PubMed] [Google Scholar]
- Chen W, Esselman WJ, Jump DB, Busik JV (2005b). Anti‐inflammatory effect of docosahexaenoic acid on cytokine‐induced adhesion molecule expression in human retinal vascular endothelial cells. Invest Ophthalmol Vis Sci 46: 4342–4347. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chen XS, Kurre U, Jenkins NA, Copeland NG, Funk CD (1994). cDNA cloning, expression, mutagenesis of C‐terminal isoleucine, genomic structure, and chromosomal localizations of murine 12‐lipoxygenases. J Biol Chem 269: 13979–13987. [PubMed] [Google Scholar]
- Cheung N, Mitchell P, Wong TY (2010). Diabetic retinopathy. Lancet 376: 124–136. [DOI] [PubMed] [Google Scholar]
- Chow LC, Wright KW, Sola A, Group COAS (2003). Can changes in clinical practice decrease the incidence of severe retinopathy of prematurity in very low birth weight infants? Pediatrics 111: 339–345. [DOI] [PubMed] [Google Scholar]
- Claar D, Hartert TV, Peebles RS Jr (2015). The role of prostaglandins in allergic lung inflammation and asthma. Expert Rev Respir Med 9: 55–72. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Clark SJ, Keenan TD, Fielder HL, Collinson LJ, Holley RJ, Merry CL et al (2011). Mapping the differential distribution of glycosaminoglycans in the adult human retina, choroid, and sclera. Invest Ophthalmol Vis Sci 52: 6511–6521. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Connor KM, SanGiovanni JP, Lofqvist C, Aderman CM, Chen J, Higuchi A et al (2007). Increased dietary intake of ω‐3‐polyunsaturated fatty acids reduces pathological retinal angiogenesis. Nat Med 13: 868–873. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Copland DA, Hussain K, Baalasubramanian S, Hughes TR, Morgan BP, Xu H et al (2010). Systemic and local anti‐C5 therapy reduces the disease severity in experimental autoimmune uveoretinitis. Clin Exp Immunol 159: 303–314. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cunha‐Vaz J, Bernardes R, Lobo C (2011). Blood‐retinal barrier. Eur J Ophthalmol 21 (Suppl 6): S3–S9. [DOI] [PubMed] [Google Scholar]
- Dai M, Xia XB, Xiong SQ (2012). BDNF regulates GLAST and glutamine synthetase in mouse retinal Muller cells. J Cell Physiol 227: 596–603. [DOI] [PubMed] [Google Scholar]
- Delaey C, Van De Voorde J (2000). Regulatory mechanisms in the retinal and choroidal circulation. Ophthalmic Res 32: 249–256. [DOI] [PubMed] [Google Scholar]
- Diabetes C, Complications Trial/Epidemiology of Diabetes I, Complications Research G , Lachin JM, White NH, Hainsworth DP et al (2015). Effect of intensive diabetes therapy on the progression of diabetic retinopathy in patients with type 1 diabetes: 18 years of follow‐up in the DCCT/EDIC. Diabetes 64: 631–642. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Diabetes C, Complications Trial Research G , Nathan DM, Genuth S, Lachin J, Cleary P et al (1993). The effect of intensive treatment of diabetes on the development and progression of long‐term complications in insulin‐dependent diabetes mellitus. N Engl J Med 329: 977–986. [DOI] [PubMed] [Google Scholar]
- Diabetic Retinopathy Clinical Research Network Writing C , Haller JA, Qin H, Apte RS, Beck RR, Bressler NM et al (2010). Vitrectomy outcomes in eyes with diabetic macular edema and vitreomacular traction. Ophthalmology 117: 1087–1093 e1083. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dobrian AD, Lieb DC, Cole BK, Taylor‐Fishwick DA, Chakrabarti SK, Nadler JL (2011). Functional and pathological roles of the 12‐ and 15‐lipoxygenases. Prog Lipid Res 50: 115–131. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dreyfuss JL, Giordano RJ, Regatieri CV (2015). Ocular angiogenesis. J Ophthalmol 2015: 892043. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Duh EJ, Sun JK, Stitt AW (2017). Diabetic retinopathy: current understanding, mechanisms, and treatment strategies. JCI Insight 2: 1–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- El Kebir D, Filep JG (2013). Modulation of neutrophil apoptosis and the resolution of inflammation through β2 integrins. Front Immunol 4: 60. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Elman MJ, Bressler NM, Qin H, Beck RW, Ferris FL 3rd, Friedman SM et al (2011). Expanded 2‐year follow‐up of ranibizumab plus prompt or deferred laser or triamcinolone plus prompt laser for diabetic macular edema. Ophthalmology 118: 609–614. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Elmasry K, Ibrahim AS, Saleh H, Elsherbiny N, Elshafey S, Hussein KA et al (2018). Role of endoplasmic reticulum stress in 12/15‐lipoxygenase‐induced retinal microvascular dysfunction in a mouse model of diabetic retinopathy. Diabetologia 61: 1220–1232. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Elmore MR, Najafi AR, Koike MA, Dagher NN, Spangenberg EE, Rice RA et al (2014). Colony‐stimulating factor 1 receptor signaling is necessary for microglia viability, unmasking a microglia progenitor cell in the adult brain. Neuron 82: 380–397. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ereso AQ, Cureton EL, Cripps MW, Sadjadi J, Dua MM, Curran B et al (2009). Lipoxin a(4) attenuates microvascular fluid leak during inflammation. J Surg Res 156: 183–188. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Escartin C, Pierre K, Colin A, Brouillet E, Delzescaux T, Guillermier M et al (2007). Activation of astrocytes by CNTF induces metabolic plasticity and increases resistance to metabolic insults. J Neurosci 27: 7094–7104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Evans JR, Michelessi M, Virgili G (2014). Laser photocoagulation for proliferative diabetic retinopathy. Cochrane Database Syst Rev : CD011234. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Faulkner J, Pye C, Al‐Shabrawey M, Elmarakby AA (2015). Inhibition of 12/15‐lipoxygenase reduces renal inflammation and injury in streptozotocin‐induced diabetic mice. J Diabetes Metab 6: 1–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ferris FL 3rd, Fine SL, Hyman L (1984). Age‐related macular degeneration and blindness due to neovascular maculopathy. Arch Ophthalmol 102: 1640–1642. [DOI] [PubMed] [Google Scholar]
- Filippi L, Cavallaro G, Bagnoli P, Dal Monte M, Fiorini P, Donzelli G et al (2013). Oral propranolol for retinopathy of prematurity: risks, safety concerns, and perspectives. J Pediatr 163 : 1570–1577 e1576. [DOI] [PubMed] [Google Scholar]
- Freire MO, Van Dyke TE (2013). Natural resolution of inflammation. Periodontol 2000 63: 149–164. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fu Z, Lofqvist CA, Shao Z, Sun Y, Joyal JS, Hurst CG et al (2015). Dietary ω‐3 polyunsaturated fatty acids decrease retinal neovascularization by adipose‐endoplasmic reticulum stress reduction to increase adiponectin. Am J Clin Nutr 101: 879–888. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Funk CD, Chen XS, Johnson EN, Zhao L (2002). Lipoxygenase genes and their targeted disruption. Prostaglandins Other Lipid Mediat 68‐69: 303–312. [DOI] [PubMed] [Google Scholar]
- Garcia‐Layana A, Cabrera‐Lopez F, Garcia‐Arumi J, Arias‐Barquet L, Ruiz‐Moreno JM (2017). Early and intermediate age‐related macular degeneration: update and clinical review. Clin Interv Aging 12: 1579–1587. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gillies MC, McAllister IL, Zhu M, Wong W, Louis D, Arnold JJ et al (2011). Intravitreal triamcinolone prior to laser treatment of diabetic macular edema: 24‐month results of a randomized controlled trial. Ophthalmology 118: 866–872. [DOI] [PubMed] [Google Scholar]
- Gordon WC, Bazan NG (1990). Docosahexaenoic acid utilization during rod photoreceptor cell renewal. J Neurosci 10: 2190–2202. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gorusupudi A, Liu A, Hageman GS, Bernstein PS (2016). Associations of human retinal very long‐chain polyunsaturated fatty acids with dietary lipid biomarkers. J Lipid Res 57: 499–508. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Goswami SK (2013). Cellular redox, epigenetics and diseases. Subcell Biochem 61: 527–542. [DOI] [PubMed] [Google Scholar]
- Gubitosi‐Klug RA, Talahalli R, Du Y, Nadler JL, Kern TS (2008). 5‐Lipoxygenase, but not 12/15‐lipoxygenase, contributes to degeneration of retinal capillaries in a mouse model of diabetic retinopathy. Diabetes 57: 1387–1393. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Harada C, Harada T, Quah HM, Maekawa F, Yoshida K, Ohno S et al (2003). Potential role of glial cell line‐derived neurotrophic factor receptors in Muller glial cells during light‐induced retinal degeneration. Neuroscience 122: 229–235. [DOI] [PubMed] [Google Scholar]
- Harding SD, Sharman JL, Faccenda E, Southan C, Pawson AJ, Ireland S et al (2018). The IUPHAR/BPS guide to PHARMACOLOGY in 2018: updates and expansion to encompass the new guide to IMMUNOPHARMACOLOGY. Nucl Acids Res 46: D1091–D1106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hauck SM, Kinkl N, Deeg CA, Swiatek‐de Lange M, Schoffmann S, Ueffing M (2006). GDNF family ligands trigger indirect neuroprotective signaling in retinal glial cells. Mol Cell Biol 26: 2746–2757. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Heckmann M (2008). Pathogenesis of retinopathy of prematurity. Ophthalmologe 105: 1101–1107. [DOI] [PubMed] [Google Scholar]
- Hernandez‐Zimbron LF, Zamora‐Alvarado R, Ochoa‐De la Paz L, Velez‐Montoya R, Zenteno E, Gulias‐Canizo R et al (2018). Age‐related macular degeneration: new paradigms for treatment and management of AMD. Oxid Med Cell Longev 2018: 8374647. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Holz FG, Strauss EC, Schmitz‐Valckenberg S, van Lookeren Campagne M (2014). Geographic atrophy: clinical features and potential therapeutic approaches. Ophthalmology 121: 1079–1091. [DOI] [PubMed] [Google Scholar]
- Hosoya K, Tachikawa M (2012). The inner blood‐retinal barrier: molecular structure and transport biology. Adv Exp Med Biol 763: 85–104. [PubMed] [Google Scholar]
- Hu J, Popp R, Fromel T, Ehling M, Awwad K, Adams RH et al (2014). Muller glia cells regulate Notch signaling and retinal angiogenesis via the generation of 19,20‐dihydroxydocosapentaenoic acid. J Exp Med 211: 281–295. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Huang H, Al‐Shabrawey M, Wang MH (2016). Cyclooxygenase‐ and cytochrome P450‐derived eicosanoids in stroke. Prostaglandins Other Lipid Mediat 122: 45–53. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ibrahim AS, El‐Remessy AB, Matragoon S, Zhang W, Patel Y, Khan S et al (2011). Retinal microglial activation and inflammation induced by amadori‐glycated albumin in a rat model of diabetes. Diabetes 60: 1122–1133. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ibrahim AS, Elshafey S, Sellak H, Hussein KA, El‐Sherbiny M, Abdelsaid M et al (2015a). A lipidomic screen of hyperglycemia‐treated HRECs links 12/15‐lipoxygenase to microvascular dysfunction during diabetic retinopathy via NADPH oxidase. J Lipid Res 56: 599–611. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ibrahim AS, Saleh H, El‐Shafey M, Hussein KA, El‐Masry K, Baban B et al (2017). Targeting of 12/15‐lipoxygenase in retinal endothelial cells, but not in monocytes/macrophages, attenuates high glucose‐induced retinal leukostasis. Biochim Biophys Acta 1862: 636–645. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ibrahim AS, Tawfik AM, Hussein KA, Elshafey S, Markand S, Rizk N et al (2015b). Pigment epithelium‐derived factor inhibits retinal microvascular dysfunction induced by 12/15‐lipoxygenase‐derived eicosanoids. Biochim Biophys Acta 1851: 290–298. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Issan Y, Hochhauser E, Guo A, Gotlinger KH, Kornowski R, Leshem‐Lev D et al (2013). Elevated level of pro‐inflammatory eicosanoids and EPC dysfunction in diabetic patients with cardiac ischemia. Prostaglandins Other Lipid Mediat 100‐101: 15–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Iwase T, Kobayashi M, Yamamoto K, Ra E, Terasaki H (2017). Effects of photocoagulation on ocular blood flow in patients with severe non‐proliferative diabetic retinopathy. PLoS One 12: e0174427. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jiang B, Liou GI, Behzadian MA, Caldwell RB (1994). Astrocytes modulate retinal vasculogenesis: effects on fibronectin expression. J Cell Sci 107 (Pt 9): 2499–2508. [DOI] [PubMed] [Google Scholar]
- Jin Y, Arita M, Zhang Q, Saban DR, Chauhan SK, Chiang N et al (2009). Anti‐angiogenesis effect of the novel anti‐inflammatory and pro‐resolving lipid mediators. Invest Ophthalmol Vis Sci 50: 4743–4752. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kang SW, Sa HS, Cho HY, Kim JI (2006). Macular grid photocoagulation after intravitreal triamcinolone acetonide for diffuse diabetic macular edema. Arch Ophthalmol 124: 653–658. [DOI] [PubMed] [Google Scholar]
- Keenan TD, Clark SJ, Unwin RD, Ridge LA, Day AJ, Bishop PN (2012). Mapping the differential distribution of proteoglycan core proteins in the adult human retina, choroid, and sclera. Invest Ophthalmol Vis Sci 53: 7528–7538. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kenyon V, Rai G, Jadhav A, Schultz L, Armstrong M, Jameson JB 2nd et al (2011). Discovery of potent and selective inhibitors of human platelet‐type 12‐lipoxygenase. J Med Chem 54: 5485–5497. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kierdorf K, Erny D, Goldmann T, Sander V, Schulz C, Perdiguero EG et al (2013). Microglia emerge from erythromyeloid precursors via Pu.1‐ and Irf8‐dependent pathways. Nat Neurosci 16: 273–280. [DOI] [PubMed] [Google Scholar]
- Kim GH, Ryan JJ, Archer SL (2013). The role of redox signaling in epigenetics and cardiovascular disease. Antioxid Redox Signal 18: 1920–1936. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kinzig A, Heidt M, Furstenberger G, Marks F, Krieg P (1999). cDNA cloning, genomic structure, and chromosomal localization of a novel murine epidermis‐type lipoxygenase. Genomics 58: 158–164. [DOI] [PubMed] [Google Scholar]
- Kolb H (1995a). Inner plexiform layer In: Kolb H, Fernandez E, Nelson R. (eds). Webvision: The Organization of the Retina and Visual System. University of Utah Health Sciences Center: Salt Lake City (UT). [PubMed] [Google Scholar]
- Kolb H (1995b). Simple anatomy of the retina In: Kolb H, Fernandez E, Nelson R. (eds). Webvision: The Organization of the Retina and Visual System. University of Utah Health Sciences Center: Salt Lake City (UT). [PubMed] [Google Scholar]
- Konkel A, Schunck WH (2011). Role of cytochrome P450 enzymes in the bioactivation of polyunsaturated fatty acids. Biochim Biophys Acta 1814: 210–222. [DOI] [PubMed] [Google Scholar]
- Koto T, Nagai N, Mochimaru H, Kurihara T, Izumi‐Nagai K, Satofuka S et al (2007). Eicosapentaenoic acid is anti‐inflammatory in preventing choroidal neovascularization in mice. Invest Ophthalmol Vis Sci 48: 4328–4334. [DOI] [PubMed] [Google Scholar]
- Kuhn H (1996). Biosynthesis, metabolization and biological importance of the primary 15‐lipoxygenase metabolites 15‐hydro (pero)XY‐5Z,8Z,11Z,13E‐eicosatetraenoic acid and 13‐hydro (pero)XY‐9Z,11E‐octadecadienoic acid. Prog Lipid Res 35: 203–226. [DOI] [PubMed] [Google Scholar]
- Kuhn H, Banthiya S, van Leyen K (2015). Mammalian lipoxygenases and their biological relevance. Biochim Biophys Acta 1851: 308–330. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kuhn H, Belkner J, Wiesner R (1990). Subcellular distribution of lipoxygenase products in rabbit reticulocyte membranes. Eur J Biochem 191: 221–227. [DOI] [PubMed] [Google Scholar]
- Kuhn H, Schewe T, Rapoport SM (1986). The stereochemistry of the reactions of lipoxygenases and their metabolites. Proposed nomenclature of lipoxygenases and related enzymes. Adv Enzymol Relat Areas Mol Biol 58: 273–311. [DOI] [PubMed] [Google Scholar]
- Kundumani‐Sridharan V, Dyukova E, Hansen DE 3rd, Rao GN (2013). 12/15‐Lipoxygenase mediates high‐fat diet‐induced endothelial tight junction disruption and monocyte transmigration: a new role for 15(S)‐hydroxyeicosatetraenoic acid in endothelial cell dysfunction. J Biol Chem 288: 15830–15842. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lam DS, Chan CK, Mohamed S, Lai TY, Lee VY, Liu DT et al (2007). Intravitreal triamcinolone plus sequential grid laser versus triamcinolone or laser alone for treating diabetic macular edema: six‐month outcomes. Ophthalmology 114: 2162–2167. [DOI] [PubMed] [Google Scholar]
- Lan YL, Chen JJ, Hu G, Xu J, Xiao M, Li S (2017). Aquaporin 4 in astrocytes is a target for therapy in Alzheimer's disease. Curr Pharm Des 23: 4948–4957. [DOI] [PubMed] [Google Scholar]
- Laybutt DR, Sharma A, Sgroi DC, Gaudet J, Bonner‐Weir S, Weir GC (2002). Genetic regulation of metabolic pathways in beta‐cells disrupted by hyperglycemia. J Biol Chem 277: 10912–10921. [DOI] [PubMed] [Google Scholar]
- Lee KS, Kim SR, Park HS, Jin GY, Lee YC (2004). Cysteinyl leukotriene receptor antagonist regulates vascular permeability by reducing vascular endothelial growth factor expression. J Allergy Clin Immunol 114: 1093–1099. [DOI] [PubMed] [Google Scholar]
- Lee R, Wong TY, Sabanayagam C (2015). Epidemiology of diabetic retinopathy, diabetic macular edema and related vision loss. Eye Vis (Lond) 2: 17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lee SJ, Kim ET, Moon YS (2011). Intravitreal bevacizumab alone versus combined with macular photocoagulation in diabetic macular edema. Korean J Ophthalmol 25: 299–304. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li Z, Wang J, Gao F, Zhang J, Tian H, Shi X et al (2016). Human adipose‐derived stem cells delay retinal degeneration in royal college of surgeons rats through anti‐apoptotic and VEGF‐mediated neuroprotective effects. Curr Mol Med 16: 553–566. [DOI] [PubMed] [Google Scholar]
- Ma Y, Qu Y, Fei Z (2011). Vascular endothelial growth factor in cerebral ischemia. J Neurosci Res 89: 969–978. [DOI] [PubMed] [Google Scholar]
- Malamas A, Chranioti A, Tsakalidis C, Dimitrakos SA, Mataftsi A (2017). The ω‐3 and retinopathy of prematurity relationship. Int J Ophthalmol 10: 300–305. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Meruvu S, Walther M, Ivanov I, Hammarstrom S, Furstenberger G, Krieg P et al (2005). Sequence determinants for the reaction specificity of murine (12R)‐lipoxygenase: targeted substrate modification and site‐directed mutagenesis. J Biol Chem 280: 36633–36641. [DOI] [PubMed] [Google Scholar]
- Meyer‐Kirchrath J, Debey S, Glandorff C, Kirchrath L, Schror K (2004). Gene expression profile of the Gs‐coupled prostacyclin receptor in human vascular smooth muscle cells. Biochem Pharmacol 67: 757–765. [DOI] [PubMed] [Google Scholar]
- Michaelis UR, Xia N, Barbosa‐Sicard E, Falck JR, Fleming I (2008). Role of cytochrome P450 2C epoxygenases in hypoxia‐induced cell migration and angiogenesis in retinal endothelial cells. Invest Ophthalmol Vis Sci 49: 1242–1247. [DOI] [PubMed] [Google Scholar]
- Mintz‐Hittner HA, Kennedy KA, Chuang AZ, Group B‐RC (2011). Efficacy of intravitreal bevacizumab for stage 3+ retinopathy of prematurity. N Engl J Med 364: 603–615. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Miranda GE, Abrahan CE, Agnolazza DL, Politi LE, Rotstein NP (2011). Ceramide‐1‐phosphate, a new mediator of development and survival in retina photoreceptors. Invest Ophthalmol Vis Sci 52: 6580–6588. [DOI] [PubMed] [Google Scholar]
- Miranda GE, Abrahan CE, Politi LE, Rotstein NP (2009). Sphingosine‐1‐phosphate is a key regulator of proliferation and differentiation in retina photoreceptors. Invest Ophthalmol Vis Sci 50: 4416–4428. [DOI] [PubMed] [Google Scholar]
- Mitchell P, Bandello F, Schmidt‐Erfurth U, Lang GE, Massin P, Schlingemann RO et al (2011). The RESTORE study: ranibizumab monotherapy or combined with laser versus laser monotherapy for diabetic macular edema. Ophthalmology 118: 615–625. [DOI] [PubMed] [Google Scholar]
- Morgan AH, Hammond VJ, Sakoh‐Nakatogawa M, Ohsumi Y, Thomas CP, Blanchet F et al (2015). A novel role for 12/15‐lipoxygenase in regulating autophagy. Redox Biol 4: 40–47. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nguyen QD, Shah SM, Heier JS, Do DV, Lim J, Boyer D et al (2009). Primary end point (six months) results of the Ranibizumab for Edema of the mAcula in diabetes (READ‐2) study. Ophthalmology 116 e2171: 2175–2181. [DOI] [PubMed] [Google Scholar]
- Nunemaker CS, Chen M, Pei H, Kimble SD, Keller SR, Carter JD et al (2008). 12‐Lipoxygenase‐knockout mice are resistant to inflammatory effects of obesity induced by Western diet. Am J Physiol Endocrinol Metab 295: E1065–E1075. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ohira T, Arita M, Omori K, Recchiuti A, Van Dyke TE, Serhan CN (2010). Resolvin E1 receptor activation signals phosphorylation and phagocytosis. J Biol Chem 285: 3451–3461. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Omarova S, Charvet CD, Reem RE, Mast N, Zheng W, Huang S et al (2012). Abnormal vascularization in mouse retina with dysregulated retinal cholesterol homeostasis. J Clin Invest 122: 3012–3023. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Omri S, Omri B, Savoldelli M, Jonet L, Thillaye‐Goldenberg B, Thuret G et al (2010). The outer limiting membrane (OLM) revisited: clinical implications. Clin Ophthalmol 4: 183–195. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Oosthuyse B, Moons L, Storkebaum E, Beck H, Nuyens D, Brusselmans K et al (2001). Deletion of the hypoxia‐response element in the vascular endothelial growth factor promoter causes motor neuron degeneration. Nat Genet 28: 131–138. [DOI] [PubMed] [Google Scholar]
- Osaadon P, Fagan XJ, Lifshitz T, Levy J (2014). A review of anti‐VEGF agents for proliferative diabetic retinopathy. Eye (Lond) 28: 510–520. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Othman A, Ahmad S, Megyerdi S, Mussell R, Choksi K, Maddipati KR et al (2013). 12/15‐Lipoxygenase‐derived lipid metabolites induce retinal endothelial cell barrier dysfunction: contribution of NADPH oxidase. PLoS One 8: e57254. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Park HY, Kim JH, Park CK (2014). Neuronal cell death in the inner retina and the influence of vascular endothelial growth factor inhibition in a diabetic rat model. Am J Pathol 184: 1752–1762. [DOI] [PubMed] [Google Scholar]
- Patz A, Eastham A, Higginbotham DH, Kleh T (1953). Oxygen studies in retrolental fibroplasia. II. The production of the microscopic changes of retrolental fibroplasia in experimental animals. Am J Ophthalmol 36: 1511–1522. [PubMed] [Google Scholar]
- Pearson PA, Comstock TL, Ip M, Callanan D, Morse LS, Ashton P et al (2011). Fluocinolone acetonide intravitreal implant for diabetic macular edema: a 3‐year multicenter, randomized, controlled clinical trial. Ophthalmology 118: 1580–1587. [DOI] [PubMed] [Google Scholar]
- Politi LE, Rotstein NP, Carri NG (2001). Effect of GDNF on neuroblast proliferation and photoreceptor survival: additive protection with docosahexaenoic acid. Invest Ophthalmol Vis Sci 42: 3008–3015. [PubMed] [Google Scholar]
- Radmark O, Werz O, Steinhilber D, Samuelsson B (2015). 5‐Lipoxygenase, a key enzyme for leukotriene biosynthesis in health and disease. Biochim Biophys Acta 1851: 331–339. [DOI] [PubMed] [Google Scholar]
- Rai G, Kenyon V, Jadhav A, Schultz L, Armstrong M, Jameson JB et al (2010). Discovery of potent and selective inhibitors of human reticulocyte 15‐lipoxygenase‐1. J Med Chem 53: 7392–7404. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Reichenbach A, Bringmann A (2013). New functions of Muller cells. Glia 61: 651–678. [DOI] [PubMed] [Google Scholar]
- Rezende FA, Lapalme E, Qian CX, Smith LE, SanGiovanni JP, Sapieha P (2014). Omega‐3 supplementation combined with anti‐vascular endothelial growth factor lowers vitreal levels of vascular endothelial growth factor in wet age‐related macular degeneration. Am J Ophthalmol 158: 1071–1078. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ricci B (1990). Oxygen‐induced retinopathy in the rat model. Doc Ophthalmol 74: 171–177. [DOI] [PubMed] [Google Scholar]
- Rickert KW, Klinman JP (1999). Nature of hydrogen transfer in soybean lipoxygenase 1: separation of primary and secondary isotope effects. Biochemistry 38: 12218–12228. [DOI] [PubMed] [Google Scholar]
- Rizzolo LJ (2014). Barrier properties of cultured retinal pigment epithelium. Exp Eye Res 126: 16–26. [DOI] [PubMed] [Google Scholar]
- Ryan A, Godson C (2010). Lipoxins: regulators of resolution. Curr Opin Pharmacol 10: 166–172. [DOI] [PubMed] [Google Scholar]
- Saadane A, Mast N, Charvet CD, Omarova S, Zheng W, Huang SS et al (2014). Retinal and nonocular abnormalities in Cyp27a1(−/−)Cyp46a1(−/−) mice with dysfunctional metabolism of cholesterol. Am J Pathol 184: 2403–2419. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sala A, Folco G, Murphy RC (2010). Transcellular biosynthesis of eicosanoids. Pharmacol Rep 62: 503–510. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sales KJ, Grant V, Jabbour HN (2008). Prostaglandin E2 and F2α activate the FP receptor and up‐regulate cyclooxygenase‐2 expression via the cyclic AMP response element. Mol Cell Endocrinol 285: 51–61. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sampat KM, Garg SJ (2010). Complications of intravitreal injections. Curr Opin Ophthalmol 21: 178–183. [DOI] [PubMed] [Google Scholar]
- Sapieha P, Stahl A, Chen J, Seaward MR, Willett KL, Krah NM et al (2011). 5‐Lipoxygenase metabolite 4‐HDHA is a mediator of the antiangiogenic effect of ω‐3 polyunsaturated fatty acids. Sci Transl Med 3: 69ra12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Schafer DP, Lehrman EK, Stevens B (2013). The “quad‐partite” synapse: microglia‐synapse interactions in the developing and mature CNS. Glia 61: 24–36. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Schewe T, Halangk W, Hiebsch C, Rapoport SM (1975). A lipoxygenase in rabbit reticulocytes which attacks phospholipids and intact mitochondria. FEBS Lett 60: 149–152. [DOI] [PubMed] [Google Scholar]
- Sears DD, Miles PD, Chapman J, Ofrecio JM, Almazan F, Thapar D et al (2009). 12/15‐lipoxygenase is required for the early onset of high fat diet‐induced adipose tissue inflammation and insulin resistance in mice. PLoS One 4: e7250. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Serhan CN, Chiang N, Dalli J (2015a). The resolution code of acute inflammation: novel pro‐resolving lipid mediators in resolution. Semin Immunol 27: 200–215. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Serhan CN, Dalli J, Colas RA, Winkler JW, Chiang N (2015b). Protectins and maresins: new pro‐resolving families of mediators in acute inflammation and resolution bioactive metabolome. Biochim Biophys Acta 1851: 397–413. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Shen J, He J, Wang F (2016). Association of lipids with age‐related macular degeneration. Discov Med 22: 129–145. [PubMed] [Google Scholar]
- Shin ES, Sorenson CM, Sheibani N (2014). Diabetes and retinal vascular dysfunction. J Ophthalmic Vis Res 9: 362–373. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Smith LE (2004). Pathogenesis of retinopathy of prematurity. Growth Horm IGF Res 14 (Suppl A): S140–S144. [DOI] [PubMed] [Google Scholar]
- Smith LE, Wesolowski E, McLellan A, Kostyk SK, D'Amato R, Sullivan R et al (1994). Oxygen‐induced retinopathy in the mouse. Invest Ophthalmol Vis Sci 35: 101–111. [PubMed] [Google Scholar]
- Spite M, Claria J, Serhan CN (2014). Resolvins, specialized proresolving lipid mediators, and their potential roles in metabolic diseases. Cell Metab 19: 21–36. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Stahl A (2018). Review of clinical trials in retinopathy of prematurity: current state and future perspectives. Ophthalmologe 115: 456–463. [DOI] [PubMed] [Google Scholar]
- Su Y, Wu J, Gu Y (2018). Photodynamic therapy in combination with ranibizumab versus ranibizumab monotherapy for wet age‐related macular degeneration: a systematic review and meta‐analysis. Photodiagnosis Photodyn Ther 22: 263–273. [DOI] [PubMed] [Google Scholar]
- Sun D, Funk CD (1996). Disruption of 12/15‐lipoxygenase expression in peritoneal macrophages. Enhanced utilization of the 5‐lipoxygenase pathway and diminished oxidation of low density lipoprotein. J Biol Chem 271: 24055–24062. [PubMed] [Google Scholar]
- Suzuki M, Kamei M, Itabe H, Yoneda K, Bando H, Kume N et al (2007). Oxidized phospholipids in the macula increase with age and in eyes with age‐related macular degeneration. Mol Vis 13: 772–778. [PMC free article] [PubMed] [Google Scholar]
- Suzuki M, Suzuki T, Nagano A, Hirasawa M, Sakuyama K, Mizuki N (2012). Cyclooxygenase inhibitor improved an exudative lesion of choroidal neovascularization in age‐related macular degeneration. Eur J Ophthalmol 22: 495–498. [DOI] [PubMed] [Google Scholar]
- Terry TL (1946). Retrolental fibroplasia. J Pediatr 29: 770–773. [DOI] [PubMed] [Google Scholar]
- Tikhonenko M, Lydic TA, Wang Y, Chen W, Opreanu M, Sochacki A et al (2010). Remodeling of retinal fatty acids in an animal model of diabetes: a decrease in long‐chain polyunsaturated fatty acids is associated with a decrease in fatty acid elongases Elovl2 and Elovl4. Diabetes 59: 219–227. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tin W, Milligan DW, Pennefather P, Hey E (2001). Pulse oximetry, severe retinopathy, and outcome at one year in babies of less than 28 weeks gestation. Arch Dis Child Fetal Neonatal Ed 84: F106–F110. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tin W, Wariyar U (2002). Giving small babies oxygen: 50 years of uncertainty. Semin Neonatol 7: 361–367. [DOI] [PubMed] [Google Scholar]
- Tserentsoodol N, Sztein J, Campos M, Gordiyenko NV, Fariss RN, Lee JW et al (2006). Uptake of cholesterol by the retina occurs primarily via a low density lipoprotein receptor‐mediated process. Mol Vis 12: 1306–1318. [PubMed] [Google Scholar]
- UK Prospective Diabetes Study (UKPDS) Group (1998). Intensive blood‐glucose control with sulphonylureas or insulin compared with conventional treatment and risk of complications in patients with type 2 diabetes (UKPDS 33). Lancet 352: 837–853. [PubMed] [Google Scholar]
- Unterlauft JD, Eichler W, Kuhne K, Yang XM, Yafai Y, Wiedemann P et al (2012). Pigment epithelium‐derived factor released by Muller glial cells exerts neuroprotective effects on retinal ganglion cells. Neurochem Res 37: 1524–1533. [DOI] [PMC free article] [PubMed] [Google Scholar]
- van Leyen K, Duvoisin RM, Engelhardt H, Wiedmann M (1998). A function for lipoxygenase in programmed organelle degradation. Nature 395: 392–395. [DOI] [PubMed] [Google Scholar]
- Vaziri K, Schwartz SG, Relhan N, Kishor KS, Flynn HW Jr (2015). New therapeutic approaches in diabetic retinopathy. Rev Diabet Stud 12: 196–210. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Vecino E, Rodriguez FD, Ruzafa N, Pereiro X, Sharma SC (2016). Glia‐neuron interactions in the mammalian retina. Prog Retin Eye Res 51: 1–40. [DOI] [PubMed] [Google Scholar]
- Ventresca MR, Gonder JR, Tanswell AK (1990). Oxygen‐induced proliferative retinopathy in the newborn rat. Can J Ophthalmol 25: 186–189. [PubMed] [Google Scholar]
- Villegas‐Becerril E, Gonzalez‐Fernandez R, Perula‐Torres L, Gallardo‐Galera JM (2006). IGF‐I, VEGF and bFGF as predictive factors for the onset of retinopathy of prematurity (ROP). Arch Soc Esp Oftalmol 81: 641–646. [DOI] [PubMed] [Google Scholar]
- Wallace JL (2018). Eicosanoids in the gastrointestinal tract. Br J Pharmacol; 10.1111/bph.14178. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wilkinson‐Berka JL, Alousis NS, Kelly DJ, Gilbert RE (2003). COX‐2 inhibition and retinal angiogenesis in a mouse model of retinopathy of prematurity. Invest Ophthalmol Vis Sci 44: 974–979. [DOI] [PubMed] [Google Scholar]
- Wolfe SA (1991). Frontal sinus hypertrophy. Plast Reconstr Surg 88: 174. [DOI] [PubMed] [Google Scholar]
- Wong WL, Su X, Li X, Cheung CM, Klein R, Cheng CY et al (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. [DOI] [PubMed] [Google Scholar]
- Woodward DF, Jones RL, Narumiya S (2011). International Union of Basic and Clinical Pharmacology. LXXXIII: classification of prostanoid receptors, updating 15 years of progress. Pharmacol Rev 63: 471–538. [DOI] [PubMed] [Google Scholar]
- Xie B, Shen J, Dong A, Rashid A, Stoller G, Campochiaro PA (2009). Blockade of sphingosine‐1‐phosphate reduces macrophage influx and retinal and choroidal neovascularization. J Cell Physiol 218: 192–198. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Xu H, Chen M (2017). Diabetic retinopathy and dysregulated innate immunity. Vision Res 139: 39–46. [DOI] [PubMed] [Google Scholar]
- Yanai R, Mulki L, Hasegawa E, Takeuchi K, Sweigard H, Suzuki J et al (2014). Cytochrome P450‐generated metabolites derived from ω‐3 fatty acids attenuate neovascularization. Proc Natl Acad Sci U S A 111: 9603–9608. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yao H, Wang T, Deng J, Liu D, Li X, Deng J (2014). The development of blood‐retinal barrier during the interaction of astrocytes with vascular wall cells. Neural Regen Res 9: 1047–1054. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yu Z, Schneider C, Boeglin WE, Brash AR (2006). Human and mouse eLOX3 have distinct substrate specificities: implications for their linkage with lipoxygenases in skin. Arch Biochem Biophys 455: 188–196. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhang HJ, Sun CH, Kuang HY, Jiang XY, Liu HL, Hua WF et al (2013). 12S‐hydroxyeicosatetraenoic acid levels link to coronary artery disease in type 2 diabetic patients. J Endocrinol Invest 36: 385–389. [DOI] [PubMed] [Google Scholar]
- Zhang R, Liu Z, Zhang H, Zhang Y, Lin D (2016). The COX‐2‐selective antagonist (NS‐398) inhibits choroidal neovascularization and subretinal fibrosis. PLoS One 11: e0146808. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhang S, Leske DA, Holmes JM (2000). Neovascularization grading methods in a rat model of retinopathy of prematurity. Invest Ophthalmol Vis Sci 41: 887–891. [PubMed] [Google Scholar]
- Zheng XR, Zhang SS, Yin F, Tang JL, Yang YJ, Wang X et al (2012). Neuroprotection of VEGF‐expression neural stem cells in neonatal cerebral palsy rats. Behav Brain Res 230: 108–115. [DOI] [PubMed] [Google Scholar]
- Zhou X, Li F, Kong L, Chodosh J, Cao W (2009). Anti‐inflammatory effect of pigment epithelium‐derived factor in DBA/2J mice. Mol Vis 15: 438–450. [PMC free article] [PubMed] [Google Scholar]
