Next Article in Journal
Chronic Lymphocytic Inflammation with Pontine Perivascular Enhancement Responsive to Steroids May Extend above and below Pons and Is Associated with Other Autoimmune Diseases
Next Article in Special Issue
PPARα Modulation-Based Therapy in Central Nervous System Diseases
Previous Article in Journal
A Bidirectional Long Short-Term Memory Model Algorithm for Predicting COVID-19 in Gulf Countries
Previous Article in Special Issue
Interactions between Apolipoprotein E Metabolism and Retinal Inflammation in Age-Related Macular Degeneration
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Metabolism in Retinopathy of Prematurity

1
Department of Ophthalmology, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA
2
Department of Ophthalmology, Juntendo University Urayasu Hospital, Chiba 279-0021, Japan
3
Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, 413 19 Gothenburg, Sweden
*
Author to whom correspondence should be addressed.
Life 2021, 11(11), 1119; https://doi.org/10.3390/life11111119
Submission received: 27 September 2021 / Revised: 11 October 2021 / Accepted: 19 October 2021 / Published: 21 October 2021
(This article belongs to the Collection Retinal Disease and Metabolism)

Abstract

:
Retinopathy of prematurity is defined as retinal abnormalities that occur during development as a consequence of disturbed oxygen conditions and nutrient supply after preterm birth. Both neuronal maturation and retinal vascularization are impaired, leading to the compensatory but uncontrolled retinal neovessel growth. Current therapeutic interventions target the hypoxia-induced neovessels but negatively impact retinal neurons and normal vessels. Emerging evidence suggests that metabolic disturbance is a significant and underexplored risk factor in the disease pathogenesis. Hyperglycemia and dyslipidemia correlate with the retinal neurovascular dysfunction in infants born prematurely. Nutritional and hormonal supplementation relieve metabolic stress and improve retinal maturation. Here we focus on the mechanisms through which metabolism is involved in preterm-birth-related retinal disorder from clinical and experimental investigations. We will review and discuss potential therapeutic targets through the restoration of metabolic responses to prevent disease development and progression.

1. Introduction

Retinopathy of prematurity (ROP) is a leading cause of blindness in children worldwide, [1] and about 14,000–16,000 infants develop ROP in the US every year. After preterm birth, ROP begins with suppression in the growth of immature retinal vasculature (phase I ROP) (Figure 1A,B), secondary to oxygen supplementation and loss of growth factors normally provided in utero [2]. As the neural retina slowly matures, the increased metabolic demand for nutrients and oxygen is not met in the avascular retinal region. Hypoxia and nutrient deprivation are driving forces to induce retinal vessel growth [3,4]. However, these newly-formed vessels are uncontrolled and fragile (phase II ROP). Phase II ROP starts at postmenstrual age 30–32 weeks, which coincides with the rapid development of rod photoreceptors [5,6]. In a rat model of ROP, early photoreceptor dysfunction also predicts subsequent neovascularization [7]. Therefore, modulating retinal metabolic needs may preserve neuronal function and prevent pathologic angiogenesis. Emerging investigations of ROP metabolic changes have been reported with a focus on nutritional interventions such as essential omega-3 and omega-6 long-chain polyunsaturated fatty acids (LCPUFA), insulin-like growth factor 1 (IGF-1), and adiponectin [8,9,10]. Recently, novel blood metabolic biomarkers for ROP have been identified with metabolomics and lipidomics to predict ROP incidence and severity. In this review, we will summarize our current knowledge of metabolic changes and modulations in ROP gained from clinical and experimental investigations.

2. Clinical Investigations of Metabolic Changes in ROP

Poor postnatal weight gain predicts severe ROP in preterm infants [11,12]. Thus, improving nutritional support may improve weight gain and subsequently prevent ROP. The LACTACOL trial investigated growth rate in a cohort of preterm infants (gestational age [GA] 30–31 weeks) who were fed their own mother’s breast milk throughout the hospital stay (49–51 days) in relation to the metabolic signature of the maternal milk. The milk from the mothers of faster-growing infants contained more arginine, tyrosine, medium-chain saturated fatty acid, and omega-3 LCPUFA (docosahexaenoic acid (DHA), eicosapentaenoic acid (EPA)), as well as less glycine, taurine, and oleic/cis-vaccenic acid [13,14]. LCPUFA (omega-3, omega-6, and omega-9) in the milk from mothers who deliver before 28 weeks of pregnancy declines rapidly between postnatal day 7 and a postmenstrual age of 40 weeks, suggesting that this already low source of LCPUFA becomes increasingly inadequate to support the development of the preterm infant [15]. LCPUFA shortage is associated with ROP progression [8,9]. Specifically, both omega-3 and omega-6 LCPUFA and their relative distribution are likely necessary factors that promote normal vessel formation and prevent ROP [16]. A low intake of lipids, carbohydrates, and total calories correlates with an increased risk of severe ROP in preterm infants with GA <28 weeks in the ELGAN study [17]. Further elucidation of the nutrients and metabolites associated with ROP would help optimize maternal diet and parenteral nutrition, as well as personalize the nutritional care of preterm infants to prevent ROP. However, there are very limited studies that correlate blood metabolites with ROP in premature infants at the current stage.

2.1. Lipidomics

Postnatal blood levels of essential fatty acids DHA and arachidonic acid (AA) are low in premature infants and are correlated with ROP progression [8,9,18]. Clinical trials supplementing preterm infants with DHA to improve the development of visual function and prevent ROP have yielded inconsistent results. Two strategies have been used to restore infant DHA: (I) administration via intravenous lipid emulsions containing fish oil [19,20,21,22] or (II) enteral supplementation using DHA from single-cell oils or fish oil [23,24,25]. Most studies assessing the effect of DHA-rich intravenous lipid emulsions have been retrospective comparative studies while only a few have prospectively investigated their role in ROP outcome, which likely contributes to the heterogeneity of the reported results. Another limitation of intravenous supplementation is duration, as preterm infants only rely on parenteral nutrition for a limited time, usually in the range of a few days to weeks. A recent Cochrane review found no support that the use of fish oil containing lipid emulsions compared to non-fish oil lipid emulsions in preterm infants reduces severe ROP (stage 3 or greater or requiring surgery), although the evidence was very low quality [26]. Contrary, a meta-analysis comparing pooled results from randomized clinical trials of early administration of fish oil vs. non-fish oil lipid emulsions found a significant reduction in the relative risk of severe ROP favoring fish oil lipids [27]. A potentially negative aspect of supplementing preterm infants with fish oil, which is naturally rich in omega-3 EPA and DHA but relatively low in omega-6 AA, is that it causes a decrease in circulating AA [24,28,29]. Daily enteral DHA and AA (100 mg and 50 mg/kg/day, respectively) given to preterm infants from birth to term equivalent age increases circulating DHA and AA levels and reduces severe ROP [25,30]. Enteral DHA supplementation at 75 mg/kg/day to preterm infants for two weeks significantly lowers the risk for stage 3 ROP [23]. Full-term infants supplemented with DHA:AA in the formula at a 1:2 ratio improves visual acuity [31]. However, doubling or tripling DHA does not confer additional benefits [31]. Meta-analysis of the randomized comparisons of DHA-supplemented formula vs. DHA-free formula to preterm infants shows improved visual resolution acuity at 2 and 4 months of corrected age [32]. In other studies, very preterm infants supplemented with DHA in the first months of life do not have better visual processing [22,33].
In mouse ROP, dietary DHA has been found to decrease retinal neovascularization [9,34,35]. DHA metabolites via lipoxygenase (LOX) inhibit while AA metabolites via LOX induce retinal angiogenesis [36]. However, both DHA and AA metabolites via cytochrome P450 oxidases (CYP) exert pro-angiogenic effects in increasing retinal neovascularization [37,38]. Interestingly, the DHA-derived diol 19,20-dihydroxydocosapentaenoic acid (19,20-DHDP) via the soluble epoxide hydrolase (sEH) pathway reduces retinal neovascularization and prevents astrocytic loss by targeting the mitochondrial membrane [39]. Moreover, both dietary DHA (no AA) and AA (no DHA) in rats profoundly alter cardiac mitochondrial phospholipid fatty acid compositions and suppress Ca2+-induced opening of the mitochondrial permeability transition pore with cell death [40,41]. Dietary DHA (no AA) also depletes cardiac mitochondrial AA content [40]. These findings suggest that the impacts of dietary DHA on ROP might be influenced by the balance between DHA and AA, as well as DHA and AA metabolites via LOX and CYP pathways.
Nilsson et al. showed the correlation of serum sphingolipids with ROP in 47 preterm infants born at GA < 28 weeks [42]. Low postnatal sphingosine-1-phosphate (S1P) levels are strongly associated with severe ROP after adjusting for GA and birth weight (Figure 2) [42]. S1P is a lysophospholipid and serves as a bioactive lipid mediator for intracellular and extracellular signals [43,44]. S1P signaling is needed for retinal vascular specialization, and the loss of S1P receptors causes extremely dense and disorganized retinal vascular plexi during development [45]. However, blockade of S1P with sonepcizumab suppresses hypoxia-induced retinal neovascularization in mouse ROP [46]. Further investigations are needed to explore the role of S1P signaling in ROP severity.

2.2. Proteomics

Lynch et al. reported that low levels of plasma mitochondrial superoxide dismutase (MnSOD) within the first postnatal week are associated with increased risk of severe ROP in 35 preterm infants with GA < 29 weeks [47], suggesting potentially disturbed metabolic status in ROP infants. SOD is a strong antioxidant in scavenging oxygen radicals generated from metabolic processes, and high SOD activity ameliorates pathological retinal angiogenesis in mice modeling phase II ROP [48]. Increased peroxidant antioxidant balance predicts the severity of ROP [49]. Prooxidant parameters including total oxidative status and malondialdehyde are higher in infants with ROP vs no ROP [50]. Furthermore, preterm infants with ROP have lower levels of the antioxidant glutathione (GSH) in their red blood cells during the first two weeks of life [51]. Premature infants are susceptible to oxygen-related damage due to their low levels of antioxidants (vitamin A and E, SOD, and catalase) [52]. Results from a recent pilot study in preterm infants suggests that enteral supplementation with AA and DHA in a 2:1 ratio can improve the antioxidant to oxidant balance [53]. Danielsson et al. further profiled the longitudinal serum protein levels between postnatal day 1 and postmenstrual age 40 weeks in 14 preterm infants with GA 22.9 to 27.6 weeks [54]. Serum proteins, such as AGER, ANGPT1, APP, CD40LG, GDF2, HBEGF, MMP12, and SERPINE1 involved in lipid metabolism are persistently lower in patients who develop severe ROP [54], suggesting a disturbed lipid metabolic status in ROP.

2.3. Metabolomics

Yang et al. reported that blood malonylcarnitine (C3DC) and glycine are higher in ROP (40 infants, 15 males, and 25 females) vs. non-ROP controls (41 infants, 30 males, 11 females) after adjusting for sex [55]. C3DC is produced from malonyl-coenzyme A (CoA), and C3DC levels reflect malonyl-CoA as patients born with malonyl-CoA decarboxylase deficiency have elevated C3DC [56,57]. Therefore, high blood C3DC levels in ROP infants indicate high concentrations of malonyl-CoA and potential disruptions of fatty acid oxidation, as malonyl-CoA inhibits carnitine palmitoyltransferase 1A (CPT1A, transporting lipids into mitochondria) [58]. Pathological angiogenesis is induced in mouse retinas with low lipid uptake and reduced fatty acid oxidation [3]. Therefore, restoration of fatty acid oxidation may prevent ROP progression.
Meanwhile, Zhou et al. also found that 11 out of 29 significantly altered blood metabolites between severe ROP (38 cases) vs. age-matched infants (23 cases) are amino acids and their derivatives [59]. Elevated plasma amino acids such as citrulline, proline, threonine, and tryptophan in ROP patients are also observed in retinas from mice modeling phase II ROP [59,60,61]. Experimental evidence shows a significant contribution of amino acids (such as proline, arginine, and glutamine) to retinal vascular function [62,63,64]. Further exploration of amino acid metabolism in ROP may identify new biomarkers for the disease development and progression, as well as uncover new therapeutic targets.
Together, clinical investigations suggest a prominent role of lipid and amino acid metabolism in ROP. Some but not all have experimental evidence. Further validation with increased clinical cases and experimental examination is needed to confirm these findings.

3. Experimental Investigations of Retinal Metabolism in ROP

3.1. Oxygen-Induced Retinopathy (OIR)

OIR has been developed in various species, such as in dogs [65], cats [66], rats [67] and mice [4] to mimic human ROP. The mouse OIR model (Figure 3A), with the advantage of genetic manipulation, has been widely used to study retinal vascular and neuronal changes in ROP. Mouse neonates with their nursing dam at postnatal day (P) 7 are exposed to 75% oxygen for five days and returned to room air (21% oxygen) at P12 [4]. Hyperoxic exposure induced retinal vessel loss and the relative hypoxia-induced retinal neovascularization reaches maximal levels at P17 [4]. This model has contributed to the developing of anti-vascular endothelial growth factor (anti-VEGFA) therapy to improve retinal neovascular diseases [68,69,70,71]. Hypoxia-inducible factor (HIF), a transcriptional factor responding to hypoxia in the tissue, regulates angiogenic genes such as VEGFA [72,73,74]. Miwa et al. reported that topotecan (HIF inhibitor) administered during the hypoxic phase (P12 to P16) suppresses the HIF pathway and the expression of Vegf, resulting in the prevention of retinal neovascularization in OIR mice [75]. Usui-Ouchi et al. reported that intravitreal injection of peptides derived from intrinsically disordered protein CITED2, a negative feedback regulator for HIF activation, inhibited retinal neovascularization and vaso-obliteration in OIR [76]. Meanwhile, Hoppe et al. suggested that stabilizing HIF-1 during the hyperoxic phase prevents vaso-obliteration and subsequent neovascularization in OIR mice [77]. Elevated aerobic glycolysis in response to HIF stabilization with HIF prolyl hydroxylase inhibitors before or during hyperoxia might contribute to the neurovascular protection of retina in OIR mice [78,79]. In addition, serine metabolism is also required for HIF-1 mediated protection against retinopathy in OIR mice [78]. These studies demonstrate HIF as a crucial factor for retinal angiogenesis and its metabolic modulation of the retina in early ROP. Inducing aerobic glycolysis and modulation of serine metabolism may prevent hyperoxia-induced retinal vessel loss in ROP.
Metabolomics profiling of the retina in OIR mice reveals disrupted glycine/creatine pathway with high retinal glycine and low creatine levels [60]. Supplementation of creatine during the hypoxic phase (P12 to P16) inhibits retinal neovascularization in OIR mice [60]. Recent studies also suggest that low glycine and serine levels are correlated with retinal degeneration [80,81]. Glycine promotes angiogenesis in mouse hind-limb ischemia in vivo and in human umbilical vein endothelial cells in vitro, as well as protects endothelial cell mitochondrial function [82]. Together, the glycine-creatine pathway may have a crucial role in ROP development.
Paris et al. reported that increases in arginine-to-proline pathway and other metabolites in the urea cycle, and decrease in purine metabolism in the whole eye from OIR versus control mice [61]. Lu et al. also showed induced plasma proline, ornithine, and glutamine, which are essential components of the arginine and proline pathway in OIR rats [83]. Systemic administration of the dipeptide arginyl-glutamine during hypoxia (P12 to P17) inhibits retinal neovascularization by ~80% and decreases neovascular tuft leakage in OIR mice [63]. Taken together, these results may suggest arginine and proline pathways as the potential diagnosis and treatments for ROP.

3.2. Hyperglycemia-Associated Retinopathy (HAR)

In addition to oxygen, hyperglycemia, which commonly occurs in preterm infants (~80% with birth weight <750 g and ~45% with birth weight <1000 g) [84], is the other significant risk factor for ROP. Hyperglycemia, particularly in the first postnatal weeks, highly correlates with delayed retinal vascularization [85] and ROP progression in preterm infants [86,87,88,89,90,91,92,93,94,95,96]. However, hyperglycemia is understudied as the current OIR model has limitations in mimicking the hyperglycemic aspect of ROP. In patients with diabetes, about one-third develop some signs of diabetic retinopathy [97], the leading cause of blindness in working-aged people [98]. Therefore, there is an urgent need to better understand the impacts of postnatal hyperglycemia on ROP. The mouse model of HAR (Figure 3B) [99] is established to investigate the impacts of metabolic dysregulation on retinal vessels and neurons at the early stages of development.
In mouse HAR model [99], hyperglycemia is induced with daily intraperitoneal injection of streptozotocin (50 mg/kg) from P1 to P9. High blood glucose is observed around P8. At P10, deep retinal vascular plexus formation between the inner nuclear layer and the photoreceptors is delayed along with the induction of hyperglycemia. Insulin treatment from P7 to P9 partially reverses the delay in retinal vessel growth. At P30, there is remarkable decrease in retinal neuronal function and retinal thickness. These observations suggest that hyperglycemia in early postnatal days induces retinal vascular and neuronal pathology, corresponding to the strong correlation between postnatal hyperglycemia and ROP progression in preterm infants. The mouse HAR model is a feasible tool to explore clinical risk factors for early ROP and potential therapeutic interventions to prevent disease progression. In preterm infants, hyperglycemia positively correlates with low serum adiponectin (APN) levels, and low serum APN levels positively correlates with delayed retinal vascularization (phase 1 ROP) [99]. In mouse HAR, activation of the APN pathway is found to be a compensatory response to improve retinal neurovascular development [99]. More interestingly, photoreceptor metabolism is reported to control the formation of deep retinal vascular plexus; improving photoreceptor metabolism leads to neurovascular protection in mouse HAR [99]. Neural control of retinal vascular stability and growth [3,100,101] is based on metabolic demands of neurons dictating growth (or loss) of vessels to supply oxygen and nutrients. Photoreceptors have the highest density of mitochondria and the highest energy demand of any cell in the body [102]. Photoreceptor energy demands are likely a major driving factor for vessel growth [3,99]. Thus, the HAR model makes it possible to explore the risk factors for ROP progression and expand our current understanding of retinal metabolism in neurovascular function.

4. Regulation of Retinal Metabolism

4.1. Nutrients

4.1.1. Glucose

Glucose metabolism is one of the most important factors controlling endothelial cell (EC) proliferation, migration, and neovascularization [103,104,105]. Blood-derived glucose penetrates the RPE and the blood–retinal barrier and arrives at the retina facilitated by sodium-independent glucose transporter 1 (Glut1) generating ATP by aerobic glycolysis [106]. ECs rely on glycolysis rather than OXPHOS for ATP production and vessel sprouting, and ECs nearly double their glycolytic flux, particularly in tip cells exposed to angiogenic stimuli, such as VEGF [107]. Glycolysis in ECs is modulated by the rate-limiting enzyme, 6-phosphofructo-2-kinase/fructose-2,6-biophosphatase 3 (PFKFB3). Pharmacological inhibition of PFKFB3 or EC-specific genetic deletion of Pfkfb3 inhibits pathological retinal neovascularization in mouse OIR [108,109]. Promotion of glucose uptake during hyperoxia in rat OIR through the inhibition of mitochondrial uncoupling protein 2 (UCP2), a cellular glucose regulator that decreases glucose uptake through Glut1, attenuates the retinal vaso-obliteration and subsequent neovascularization [110]. The adenosine A2a receptor (ADORA2A) promotes HIF-1-dependent endothelial cell glycolysis, and the EC-specific Adora2a deletion decreases retinal neovascularization in mouse OIR [111]. In addition, under physiological conditions, glycolysis converts glucose to energy, with less than 3% of glucose diverted into the polyol pathway, which reduces glucose to sorbitol and increases oxidative stress through the production of highly toxic advanced glycation end products [112]. Aldose reductase is the rate-limiting enzyme in the polyol pathway, and the deletion of the enzyme reduces retinal neovascularization through the attenuation of oxidative stress and protects retinal neurons in mouse OIR [113,114]. These findings suggest that targeting retinal glucose metabolism is an effective way to control pathological retinal angiogenesis.
Recently, single-cell RNA sequencing reveals that glycolysis gene expression is upregulated in proliferating ECs, but less in tip and immature ECs in a mouse model of choroidal neovascularization [115]. Proliferating ECs also upregulated genes involved in one-carbon metabolism, nucleotide synthesis, TCA cycle and OXPHOS [115], suggesting the involvement of other metabolic pathways in modulating pathological ocular angiogenesis. Further exploration of their role in ROP is needed.

4.1.2. Amino Acids

Premature infants frequently lack arginine and glutamine because they are unable to maintain the endogenous synthesis of these conditionally essential amino acids [116,117]. The supplementation of arginine and glutamine (Arg-Gln) suppresses pathological neovascularization in OIR; an in vitro experiment in human RPE cells showed that Arg-Gln decreases VEGF expression [63]. ECs have high glutaminase (GLS) activity, which is the enzyme that converts glutamine and glutaminase in the first and rate-limiting step of glutaminolysis, producing energy for proliferation [118]. Glutamine is indispensable for vessel sprouting, and the inhibition of GLS1 causes sprouting defects in vitro and in mouse models of developmental angiogenesis and pathological neovascularization in OIR in vivo [119].
Serine metabolism via phosphoglycerate dehydrogenase (PHGDH), a key enzyme in the serine synthesis pathway, is important for retinal cell survival, including in EC [80,120]. Loss of Phgdh in ECs cause defects in retinal angiogenesis and promotes EC apoptosis via heme deficiency, which induces mitochondrial respiration defects and oxidative stress [121]. Activation of serine and one carbon metabolism is required for HIF-1 stabilization to protect against hyperoxia-induced retinal vaso-obliteration in mouse OIR [78]. Meanwhile, disruption of serine synthesis in the Müller glia also induces mitochondrial dysfunction [122] and the Müller glia relies on serine biosynthesis to combat oxidative stress [123]. Müller glia is the primary source of VEGF in neovascular retina [124,125]. Therefore, targeting retinal serine metabolism may protect against retinal neovascularization in ROP.

4.1.3. Fatty Acids

Fatty acids are the other major substrate for energy production in ECs. In vitro, glucose deprivation causes ECs to increase fatty acid oxidation (FAO) flux in an AMP-activated protein kinase (AMPK)-dependent manner [103]. Endothelial FAO plays an important role in regulating vessel sprouting [126]. As the rate-limiting enzyme of FAO, carnitine palmitoyltransferase 1a (CPT1a) imports FAs into the mitochondria. The endothelial loss of CPT1a causes retinal vascular sprouting defects due to impaired proliferation (not migration) through the inhibition of de novo nucleotide synthesis for DNA replication [126]. ECs express fatty acid synthase (FAS), and FAS-mediated de novo lipogenesis is required for vascular sprouting and permeability [127]. VEGF enhances the expression of fatty acid uptake and trafficking protein FABP4, which is required for normal EC proliferation [128]. Moreover, decreases in both FAO and glycolysis in photoreceptors also induces HIF stabilization and VEGF production, resulting in retinal neovascularization in mice [3,129]. These findings suggest modulating retinal FAO may also prevent neovascular ROP.

4.2. Hormones

4.2.1. Adiponectin (APN)

APN is an abundant circulating adipokine involved in metabolic modulation [2]. In premature infants, low circulating APN levels correlate with delayed retinal vascularization and ROP progression [9]. In mouse OIR, loss of APN exacerbates and APN administration decreases retinal neovascularization [130]. Loss of APN receptor 1 in mice leads to abolished DHA uptake, retention, conservation, elongation in photoreceptors, and eventual photoreceptor degeneration [131,132]. In mouse HAR, pharmacologic activation of the APN pathway by recombinant APN or APN receptor agonist exerts protective effects on retinal vessel growth and neuronal development [99]. These studies suggest that increasing circulating APN levels might benefit the preterm infants and decrease the risk for ROP incidence and progression.
Omega-3 LCPUFA increases circulating APN, which mediates omega-3 LCPUFA’s inhibitory effects on neovascularization in OIR mice [9], as well as in other mouse models with proliferative retinopathy [133]. In premature infants, circulating APN is positively correlated with DHA [9]. The increase in circulating APN by dietary omega-3 LCPUFA has also been demonstrated in various studies [134,135,136,137]. These reports suggest that omega-3 LCPUFA supplementation is essential in maintaining circulating APN levels to prevent ROP.
In addition, APN levels could be modulated by fibroblast growth factor 21 (FGF21) [138], which is expressed in many tissues but mainly in the liver under physiologic conditions [139]. FGF21 plays an essential role in modulating lipid and glucose use [140,141,142]. FGF21 is also a key regulator of browning of white adipose tissue and increases energy expenditure [143]. FGF21 via APN inhibits choroidal and retinal neovascularization in mice [144]. FGF21 also increases APN secretion in obese mice [138] and protects diabetes-induced retinal neuronal dysfunction [145]. Furthermore, FGF21 preserves retinal neuronal responses in mice with inherited retinal degeneration [146]. In preterm infants, circulating FGF21 levels are very low, and the postnatal increase in FGF21 observed in full-term infants seems absent in preterm infants [147,148,149]. Taken together, these reports suggest that circulating FGF21 levels may be correlated with increase in APN levels and ROP progression in preterm infants. Further clinic investigations are needed to validate this hypothesis.

4.2.2. Insulin-Growth Factor 1 (IGF-1)

IGF-1 is an important liver-derived growth factor and a key regulator of body growth and development [150,151]. In premature infants, persistent low circulating IGF-1 levels strongly correlate with ROP development [94,152,153,154,155,156]. IGF-1 is critical for normal retinal vascularization as a lack of IGF-1 in mice prevents retinal vessel growth [152]. IGF-1 also supports VEGF activation of endothelial cell proliferation [152,157]. Therefore, early restoration of IGF-1 may prevent ROP. Mice with early supplementation of IGF-1 before exposure to hyperoxia have less vessel loss and neovascularization in the OIR model [158]. In premature infants with postnatal hyperglycemia in the first month, there are also lower plasma IGF-1 levels [94]. In mouse OIR model combined with the HAR model, decreased liver IGF-1 expression is observed before the induction of hyperglycemia; IGF-1 treatment reduces retinal neovascularization and improves retinal revascularization [94]. These findings suggest that early supplementation of IGF-1 may improve retinal vascularization and decrease ROP risk. The phase 2 randomized controlled trial (ClinicalTrials.gov Identifier: NCT01096784) shows that rhIGF-1/rhIGFBP-3 decreases the occurrence of severe bronchopulmonary dysplasia, but the dose needs to be further optimized for ROP prevention [159]. Increasing the number of patients in the study would also help evaluate the effects of IGF-1 on ROP with completion of the current phase 2b clinical trial using SHP607 (recombinant protein complex of IGF-1/IGFBP3) in preterm infants (ClinicalTrials.gov Identifier: NCT03253263). Moreover, recent investigations have also demonstrated that low circulating IGF-1 levels are correlated with low weekly platelet counts [156], which is associated with ROP progression in premature infants [160,161]. Platelet transfusions inhibit retinal neovascularization in OIR mice [160], suggesting that normalizing platelet levels and platelet-derived growth factors (IGF-1, VEGFA, PDGFBB [156]) might prevent ROP in premature infants.

4.3. Other Related to Metabolism

4.3.1. Peroxisome Proliferator-Activated Receptor α (PPARα) Agonist

Fenofibrate, a PPARα agonist, is an antihyperlipidemic drug. The FIELD and ACCORD studies have shown that fenofibrate suppresses the progression of diabetic retinopathy [162,163]. Fenofibrate modulates lipid metabolism, reduces triglyceride (TG), and increases high-density lipoprotein (HDL) cholesterol [164]. Fenofibrate inhibits neovascularization in OIR mice through the suppression of HIF-1α and VEGF [165]. However, fenofibrate is not recommended for patients with renal dysfunction because it metabolizes in the kidney. However, kidneys are often underdeveloped in premature infants [166]. Recently, pemafibrate, which is a selective PPARα modulator, has been approved for use in Japan. Pemafibrate is as effective as fenofibrate in modulating hyperlipidemia and reduces the associated risks in the liver and kidney [167,168], possibly due to the structural differences between fenofibrate and pemafibrate [169]. Pemafibrate decreases retinal neovascularization in OIR mice and protects retinal function in diabetic mice model by inducing FGF21 [170,171]. Pemafibrate also suppresses HIF1α and Vegf in OIR retinas [170]. Currently, phase 3 clinical trials of the use of pemafibrate to reduce cardiovascular outcomes by reducing triglycerides in patients with type 2 diabetes (PROMINENT) is ongoing (ClinicalTrials.gov Identifier: NCT03071692). With the potential application of pamafibrate in treating diabetes and diabetic retinopathy, pamafibrate may also be a therapeutic potential for other retinal metabolic disorders such as ROP.

4.3.2. Rapamycin

Rapamycin (Sirolimus) is an inhibitor of mammalian target of rapamycin (mTOR), with anti-proliferative, antiangiogenic, and immunosuppressive properties [172]. Rapamycin is used to prevent organ transplant rejection and treat lymphangioleiomyomatosis, a rare lung disease [173,174]. mTOR is a serine-threonine protein kinase and functions as two distinct signaling complexes: mTOR complex 1 (mTORC1) and mTORC2 [175]. mTORC1 is involved in immune responses and lipid metabolism in the human body [176]. In the context of eye disease, several studies showed that systemic rapamycin treatment reduces retinal neovascularization in OIR mice [177,178]. Rapamycin also reduces vascular apoptosis and promotes proliferation and tip cell function in OIR mice [179]. Together, these data suggest that rapamycin may be a promising strategy for early intervention of ROP.

4.3.3. Rho-Associated Kinase (ROCK) Inhibitor

ROCK is involved in inflammation, angiogenesis, apoptosis, and cytoskeletal rearrangement [180,181,182]. ROCK is identified as a downstream effector of the small GTP-binding protein Rho and has two isoforms, ROCK1 and ROCK2 [183]. Noda et al. showed that inhibition of Rho-kinase increases energy expenditure via AMPK activation in brown adipose tissue and improves metabolic disorders [184]. Several ROCK inhibitors exhibit suppression of pathological neovascularization in OIR rodent models up to date, such as Fasudil, Ripasudil, Y27632, and AMA 0428 [185,186,187]. Ripasudil, in particular, induces pericyte coverage and improves retinal vascular perfusion in mouse OIR [186]. Several clinical trials are currently active to evaluate whether Fasudil or Ripasudil eye drop affects ROP prevention (ClinicalTrials.gov Identifier: NCT04191954, NCT04621136). Topically applied ROCK inhibitors would be potentially beneficial for ROP treatment.

4.3.4. Autophagy

Autophagy is a cellular process induced by many stresses, including hypoxia, starvation, and infection, to maintain homeostasis [188]. Autophagy increases during the first postnatal days and decreases as the retina reaches full vascularization in rats [189]. Elevated retinal reactive oxygen species and attenuated autophagy is shown in OIR mice [190]. Knockdown of β5i, an immunosubunit of the immunoproteasome, increases autophagy-related protein 5 (ATG5) and inhibits retinal neovascularization in mouse OIR [191]. On the other hand, a significant increase in autophagy flux is reported in mouse OIR retinas, particularly in proliferating endothelial cells [192]. Endothelial-specific deletion of ATG5 attenuates retinal neovascularization in OIR mice [193]. Further studies are needed to elucidate the role of autophagic imbalance in ocular angiogenesis. Overall, autophagy could be a novel target for pharmacological intervention in ROP patients.

5. Future Perspectives

We here summarized the current understanding of metabolic impacts on ROP (Figure 4). Overall, metabolic disturbances in glucose, amino acid, and lipid use may contribute to ROP development and progression. Hormonal modulation plays an essential role in maintaining metabolic homeostasis. Further understanding of the relationship and interaction among risk factors at early and late stages of ROP is essential for clinical intervention. Our current knowledge gained from DHA and AA supplementation in premature infants suggests that maintaining adequate AA levels is required for DHA to exert protective effects against ROP [25,30,194]. In addition, loss of APN largely abolishes DHA’s role of inhibition on retinal neovascularization in mouse OIR [9]. Restoration of APN levels in premature infants may further exaggerate DHA protection against ROP. Moreover, there is a potential concern that continuous peroxidation of VLCFA may cause chronic inflammation and neuronal damage [195,196]. Thus, the timing in the administration of DHA and AA intervention also needs to be evaluated. The same concept also applies to IGF-1 supplementation (and potentially other therapeutic targets as well) as IGF-1 improves retinal vascularization in phase I ROP and may exacerbate VEGF-induced neovascularization at phase II ROP [152,157].
To expand our current knowledge of nutritional and hormonal regulation in retinal metabolism and ROP, we need to further understand the types of metabolic substrates in retinal neuronal and endothelial cells, as well as the interaction among the different types of retinal cells. Endothelial cell metabolism (glycolysis, fatty acid oxidation, and serine synthesis) controls physiological and pathological retinal angiogenesis [103,121], which may be controlled by Müller glia [125,197]. Müller glial cells also produce and transfer nutrients (such as lactate) to photoreceptors [198] in addition to the uptake and conversion of glutamate to glutamine [199]. Moreover, retinal pigmented epithelium maintains photoreceptor metabolism by transferring glucose and passing and recycling lipids to photoreceptors [200,201,202,203,204,205,206]. Photoreceptor metabolism also controls physiological and pathological retinal vessel growth [3,99]. Taken together, there are complex interactions among retinal cells. Specific cellular responses and cell-cell interaction need to be considered and long-term impacts need to be examined.

Author Contributions

Writing—original draft preparation, Y.T., A.U.-O., and Z.F.; writing—review and editing, A.K.N., A.H., Z.F., J.Y., and M.K.; funding acquisition, Z.F. All authors have read and agreed to the published version of the manuscript.

Funding

Z.F. was funded by NEI 1R01EY032492, Boston Children’s Hospital (OFD/BTREC/CTREC Faculty Career Development Grant 97906, Pilot Grant 92214, and Ophthalmology Foundation 85010), and Mass Lions Eye Foundation 87820; Y.T. was funded by Manpei Suzuki Diabetes Foundation, Alcon Research Institute 77486 and Bert M. Glaser, MD Award 80131; A.U.O. was funded by Manpei Suzuki Diabetes Foundation and JSPS KAKENHI Grant Number 17K16984 and 21K09727.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Hellstrom, A.; Smith, L.E.; Dammann, O. Retinopathy of prematurity. Lancet 2013, 382, 1445–1457. [Google Scholar] [CrossRef] [Green Version]
  2. Fu, Z.; Gong, Y.; Lofqvist, C.; Hellstrom, A.; Smith, L.E. Review: Adiponectin in retinopathy. Biochim. Biophys. Acta 2016, 1862, 1392–1400. [Google Scholar] [CrossRef] [PubMed]
  3. Joyal, J.S.; Sun, Y.; Gantner, M.L.; Shao, Z.; Evans, L.P.; Saba, N.; Fredrick, T.; Burnim, S.; Kim, J.S.; Patel, G.; et al. Retinal lipid and glucose metabolism dictates angiogenesis through the lipid sensor Ffar1. Nat. Med. 2016, 22, 439–445. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Smith, L.E.; Wesolowski, E.; McLellan, A.; Kostyk, S.K.; D’Amato, R.; Sullivan, R.; D’Amore, P.A. Oxygen-induced retinopathy in the mouse. Investig. Ophthalmol. Vis. Sci. 1994, 35, 101–111. [Google Scholar]
  5. Hansen, R.M.; Moskowitz, A.; Akula, J.D.; Fulton, A.B. The neural retina in retinopathy of prematurity. Prog. Retin. Eye Res. 2017, 56, 32–57. [Google Scholar] [CrossRef] [Green Version]
  6. Fulton, A.B.; Dodge, J.; Hansen, R.M.; Williams, T.P. The rhodopsin content of human eyes. Investig. Ophthalmol. Vis. Sci. 1999, 40, 1878–1883. [Google Scholar]
  7. Akula, J.D.; Hansen, R.M.; Tzekov, R.; Favazza, T.L.; Vyhovsky, T.C.; Benador, I.Y.; Mocko, J.A.; McGee, D.; Kubota, R.; Fulton, A.B. Visual cycle modulation in neurovascular retinopathy. Exp. Eye Res. 2010, 91, 153–161. [Google Scholar] [CrossRef]
  8. Lofqvist, C.A.; Najm, S.; Hellgren, G.; Engstrom, E.; Savman, K.; Nilsson, A.K.; Andersson, M.X.; Hard, A.L.; Smith, L.E.H.; Hellstrom, A. Association of Retinopathy of Prematurity With Low Levels of Arachidonic Acid: A Secondary Analysis of a Randomized Clinical Trial. JAMA Ophthalmol. 2018, 136, 271–277. [Google Scholar] [CrossRef] [Green Version]
  9. Fu, Z.; Lofqvist, C.A.; Shao, Z.; Sun, Y.; Joyal, J.S.; Hurst, C.G.; Cui, R.Z.; Evans, L.P.; Tian, K.; SanGiovanni, J.P.; et al. Dietary omega-3 polyunsaturated fatty acids decrease retinal neovascularization by adipose-endoplasmic reticulum stress reduction to increase adiponectin. Am. J. Clin. Nutr. 2015, 101, 879–888. [Google Scholar] [CrossRef] [Green Version]
  10. Smith, L.E. IGF-1 and retinopathy of prematurity in the preterm infant. Biol. Neonate 2005, 88, 237–244. [Google Scholar] [CrossRef] [PubMed]
  11. Lundgren, P.; Stoltz Sjostrom, E.; Domellof, M.; Kallen, K.; Holmstrom, G.; Hard, A.L.; Smith, L.E.; Lofqvist, C.; Hellstrom, A. WINROP identifies severe retinopathy of prematurity at an early stage in a nation-based cohort of extremely preterm infants. PLoS ONE 2013, 8, e73256. [Google Scholar] [CrossRef]
  12. Wallace, D.K.; Kylstra, J.A.; Phillips, S.J.; Hall, J.G. Poor postnatal weight gain: A risk factor for severe retinopathy of prematurity. JAAPOS 2000, 4, 343–347. [Google Scholar] [CrossRef]
  13. Alexandre-Gouabau, M.C.; Moyon, T.; David-Sochard, A.; Fenaille, F.; Cholet, S.; Royer, A.L.; Guitton, Y.; Billard, H.; Darmaun, D.; Roze, J.C.; et al. Comprehensive Preterm Breast Milk Metabotype Associated with Optimal Infant Early Growth Pattern. Nutrients 2019, 11, 528. [Google Scholar] [CrossRef] [Green Version]
  14. Alexandre-Gouabau, M.C.; Moyon, T.; Cariou, V.; Antignac, J.P.; Qannari, E.M.; Croyal, M.; Soumah, M.; Guitton, Y.; David-Sochard, A.; Billard, H.; et al. Breast Milk Lipidome Is Associated with Early Growth Trajectory in Preterm Infants. Nutrients 2018, 10, 164. [Google Scholar] [CrossRef] [Green Version]
  15. Nilsson, A.K.; Lofqvist, C.; Najm, S.; Hellgren, G.; Savman, K.; Andersson, M.X.; Smith, L.E.H.; Hellstrom, A. Long-chain polyunsaturated fatty acids decline rapidly in milk from mothers delivering extremely preterm indicating the need for supplementation. Acta Paediatr. 2018, 107, 1020–1027. [Google Scholar] [CrossRef] [Green Version]
  16. Crawford, M.A.; Costeloe, K.; Ghebremeskel, K.; Phylactos, A.; Skirvin, L.; Stacey, F. Are deficits of arachidonic and docosahexaenoic acids responsible for the neural and vascular complications of preterm babies? Am. J. Clin. Nutr. 1997, 66, 1032S–1041S. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  17. VanderVeen, D.K.; Martin, C.R.; Mehendale, R.; Allred, E.N.; Dammann, O.; Leviton, A.; Investigators, E.S. Early nutrition and weight gain in preterm newborns and the risk of retinopathy of prematurity. PLoS ONE 2013, 8, e64325. [Google Scholar] [CrossRef] [PubMed]
  18. Lapillonne, A.; Eleni dit Trolli, S.; Kermorvant-Duchemin, E. Postnatal docosahexaenoic acid deficiency is an inevitable consequence of current recommendations and practice in preterm infants. Neonatology 2010, 98, 397–403. [Google Scholar] [CrossRef]
  19. Pawlik, D.; Lauterbach, R.; Walczak, M.; Hurkala, J.; Sherman, M.P. Fish-oil fat emulsion supplementation reduces the risk of retinopathy in very low birth weight infants: A prospective, randomized study. JPEN J. Parenter. Enter. Nutr. 2014, 38, 711–716. [Google Scholar] [CrossRef] [PubMed]
  20. Pawlik, D.; Lauterbach, R.; Turyk, E. Fish-oil fat emulsion supplementation may reduce the risk of severe retinopathy in VLBW infants. Pediatrics 2011, 127, 223–228. [Google Scholar] [CrossRef] [PubMed]
  21. Beken, S.; Dilli, D.; Fettah, N.D.; Kabatas, E.U.; Zenciroglu, A.; Okumus, N. The influence of fish-oil lipid emulsions on retinopathy of prematurity in very low birth weight infants: A randomized controlled trial. Early Hum. Dev. 2014, 90, 27–31. [Google Scholar] [CrossRef]
  22. Najm, S.; Lofqvist, C.; Hellgren, G.; Engstrom, E.; Lundgren, P.; Hard, A.L.; Lapillonne, A.; Savman, K.; Nilsson, A.K.; Andersson, M.X.; et al. Effects of a lipid emulsion containing fish oil on polyunsaturated fatty acid profiles, growth and morbidities in extremely premature infants: A randomized controlled trial. Clin. Nutr. ESPEN 2017, 20, 17–23. [Google Scholar] [CrossRef] [Green Version]
  23. Bernabe-Garcia, M.; Villegas-Silva, R.; Villavicencio-Torres, A.; Calder, P.C.; Rodriguez-Cruz, M.; Maldonado-Hernandez, J.; Macias-Loaiza, D.; Lopez-Alarcon, M.; Inda-Icaza, P.; Cruz-Reynoso, L. Enteral Docosahexaenoic Acid and Retinopathy of Prematurity: A Randomized Clinical Trial. JPEN J. Parenter. Enter. Nutr. 2019, 43, 874–882. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Collins, C.T.; Makrides, M.; McPhee, A.J.; Sullivan, T.R.; Davis, P.G.; Thio, M.; Simmer, K.; Rajadurai, V.S.; Travadi, J.; Berry, M.J.; et al. Docosahexaenoic Acid and Bronchopulmonary Dysplasia in Preterm Infants. N. Engl. J. Med. 2017, 376, 1245–1255. [Google Scholar] [CrossRef] [PubMed]
  25. Hellstrom, A.; Nilsson, A.K.; Wackernagel, D.; Pivodic, A.; Vanpee, M.; Sjobom, U.; Hellgren, G.; Hallberg, B.; Domellof, M.; Klevebro, S.; et al. Effect of Enteral Lipid Supplement on Severe Retinopathy of Prematurity: A Randomized Clinical Trial. JAMA Pediatr. 2021, 175, 359–367. [Google Scholar] [CrossRef]
  26. Kapoor, V.; Malviya, M.N.; Soll, R. Lipid emulsions for parenterally fed preterm infants. Cochrane Database Syst. Rev. 2019, 6, CD013163. [Google Scholar] [CrossRef] [PubMed]
  27. Vayalthrikkovil, S.; Bashir, R.A.; Rabi, Y.; Amin, H.; Spence, J.M.; Robertson, H.L.; Lodha, A. Parenteral Fish-Oil Lipid Emulsions in the Prevention of Severe Retinopathy of Prematurity: A Systematic Review and Meta-Analysis. Am. J. Perinatol. 2017, 34, 705–715. [Google Scholar] [CrossRef]
  28. Zhao, Y.; Wu, Y.; Pei, J.; Chen, Z.; Wang, Q.; Xiang, B. Safety and efficacy of parenteral fish oil-containing lipid emulsions in premature neonates. J. Pediatr. Gastroenterol. Nutr. 2015, 60, 708–716. [Google Scholar] [CrossRef]
  29. D’Ascenzo, R.; Savini, S.; Biagetti, C.; Bellagamba, M.P.; Marchionni, P.; Pompilio, A.; Cogo, P.E.; Carnielli, V.P. Higher docosahexaenoic acid, lower arachidonic acid and reduced lipid tolerance with high doses of a lipid emulsion containing 15% fish oil: A randomized clinical trial. Clin. Nutr. 2014, 33, 1002–1009. [Google Scholar] [CrossRef]
  30. Hellstrom, A.; Pivodic, A.; Granse, L.; Lundgren, P.; Sjobom, U.; Nilsson, A.K.; Soderling, H.; Hard, A.L.; Smith, L.E.H.; Lofqvist, C.A. Association of Docosahexaenoic Acid and Arachidonic Acid Serum Levels With Retinopathy of Prematurity in Preterm Infants. JAMA Netw. Open 2021, 4, e2128771. [Google Scholar] [CrossRef]
  31. Birch, E.E.; Carlson, S.E.; Hoffman, D.R.; Fitzgerald-Gustafson, K.M.; Fu, V.L.; Drover, J.R.; Castaneda, Y.S.; Minns, L.; Wheaton, D.K.; Mundy, D.; et al. The DIAMOND (DHA Intake And Measurement Of Neural Development) Study: A double-masked, randomized controlled clinical trial of the maturation of infant visual acuity as a function of the dietary level of docosahexaenoic acid. Am. J. Clin. Nutr. 2010, 91, 848–859. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. SanGiovanni, J.P.; Parra-Cabrera, S.; Colditz, G.A.; Berkey, C.S.; Dwyer, J.T. Meta-analysis of dietary essential fatty acids and long-chain polyunsaturated fatty acids as they relate to visual resolution acuity in healthy preterm infants. Pediatrics 2000, 105, 1292–1298. [Google Scholar] [CrossRef] [PubMed]
  33. Molloy, C.S.; Stokes, S.; Makrides, M.; Collins, C.T.; Anderson, P.J.; Doyle, L.W. Long-term effect of high-dose supplementation with DHA on visual function at school age in children born at <33 wk gestational age: Results from a follow-up of a randomized controlled trial. Am. J. Clin. Nutr. 2016, 103, 268–275. [Google Scholar] [CrossRef] [PubMed]
  34. Connor, K.M.; SanGiovanni, J.P.; Lofqvist, C.; Aderman, C.M.; Chen, J.; Higuchi, A.; Hong, S.; Pravda, E.A.; Majchrzak, S.; Carper, D.; et al. Increased dietary intake of omega-3-polyunsaturated fatty acids reduces pathological retinal angiogenesis. Nat. Med. 2007, 13, 868–873. [Google Scholar] [CrossRef] [Green Version]
  35. Stahl, A.; Sapieha, P.; Connor, K.M.; Sangiovanni, J.P.; Chen, J.; Aderman, C.M.; Willett, K.L.; Krah, N.M.; Dennison, R.J.; Seaward, M.R.; et al. Short communication: PPAR gamma mediates a direct antiangiogenic effect of omega 3-PUFAs in proliferative retinopathy. Circ. Res. 2010, 107, 495–500. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Sapieha, P.; Stahl, A.; Chen, J.; Seaward, M.R.; Willett, K.L.; Krah, N.M.; Dennison, R.J.; Connor, K.M.; Aderman, C.M.; Liclican, E.; et al. 5-Lipoxygenase metabolite 4-HDHA is a mediator of the antiangiogenic effect of omega-3 polyunsaturated fatty acids. Sci. Transl. Med. 2011, 3, 69ra12. [Google Scholar] [CrossRef] [Green Version]
  37. Gong, Y.; Fu, Z.; Edin, M.L.; Liu, C.H.; Wang, Z.; Shao, Z.; Fredrick, T.W.; Saba, N.J.; Morss, P.C.; Burnim, S.B.; et al. Cytochrome P450 Oxidase 2C Inhibition Adds to omega-3 Long-Chain Polyunsaturated Fatty Acids Protection Against Retinal and Choroidal Neovascularization. Arter. Thromb. Vasc. Biol. 2016, 36, 1919–1927. [Google Scholar] [CrossRef] [Green Version]
  38. Gong, Y.; Shao, Z.; Fu, Z.; Edin, M.L.; Sun, Y.; Liegl, R.G.; Wang, Z.; Liu, C.H.; Burnim, S.B.; Meng, S.S.; et al. Fenofibrate Inhibits Cytochrome P450 Epoxygenase 2C Activity to Suppress Pathological Ocular Angiogenesis. EBioMedicine 2016, 13, 201–211. [Google Scholar] [CrossRef] [Green Version]
  39. Hu, J.; Bibli, S.I.; Wittig, J.; Zukunft, S.; Lin, J.; Hammes, H.P.; Popp, R.; Fleming, I. Soluble epoxide hydrolase promotes astrocyte survival in retinopathy of prematurity. J. Clin. Investig. 2019, 129, 5204–5218. [Google Scholar] [CrossRef]
  40. Khairallah, R.J.; Kim, J.; O’Shea, K.M.; O’Connell, K.A.; Brown, B.H.; Galvao, T.; Daneault, C.; Des Rosiers, C.; Polster, B.M.; Hoppel, C.L.; et al. Improved mitochondrial function with diet-induced increase in either docosahexaenoic acid or arachidonic acid in membrane phospholipids. PLoS ONE 2012, 7, e34402. [Google Scholar] [CrossRef] [Green Version]
  41. Khairallah, R.J.; Sparagna, G.C.; Khanna, N.; O’Shea, K.M.; Hecker, P.A.; Kristian, T.; Fiskum, G.; Des Rosiers, C.; Polster, B.M.; Stanley, W.C. Dietary supplementation with docosahexaenoic acid, but not eicosapentaenoic acid, dramatically alters cardiac mitochondrial phospholipid fatty acid composition and prevents permeability transition. Biochim. Biophys. Acta 2010, 1797, 1555–1562. [Google Scholar] [CrossRef] [Green Version]
  42. Nilsson, A.K.; Andersson, M.X.; Sjobom, U.; Hellgren, G.; Lundgren, P.; Pivodic, A.; Smith, L.E.H.; Hellstrom, A. Sphingolipidomics of serum in extremely preterm infants: Association between low sphingosine-1-phosphate levels and severe retinopathy of prematurity. Biochim. Biophys. Acta Mol. Cell Biol. Lipids 2021, 1866, 158939. [Google Scholar] [CrossRef]
  43. Victoria, S.M.; Basu, S.K.; Bano, Q.; Richard, G.; Rotstein, N.P.; Nawajes, M. Sphingolipids as critical players in retinal physiology and pathology. J. Lipid Res. 2021, 62, 100037. [Google Scholar] [CrossRef]
  44. Miranda, G.E.; Abrahan, C.E.; Politi, L.E.; Rotstein, N.P. Sphingosine-1-phosphate is a key regulator of proliferation and differentiation in retina photoreceptors. Investig. Ophthalmol. Vis. Sci. 2009, 50, 4416–4428. [Google Scholar] [CrossRef]
  45. Yanagida, K.; Engelbrecht, E.; Niaudet, C.; Jung, B.; Gaengel, K.; Holton, K.; Swendeman, S.; Liu, C.H.; Levesque, M.V.; Kuo, A.; et al. Sphingosine 1-Phosphate Receptor Signaling Establishes AP-1 Gradients to Allow for Retinal Endothelial Cell Specialization. Dev. Cell 2020, 52, 779–793.e7. [Google Scholar] [CrossRef] [PubMed]
  46. Xie, B.; Shen, J.; Dong, A.; Rashid, A.; Stoller, G.; Campochiaro, P.A. Blockade of sphingosine-1-phosphate reduces macrophage influx and retinal and choroidal neovascularization. J. Cell. Physiol. 2009, 218, 192–198. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Lynch, A.M.; Wagner, B.D.; Mandava, N.; Palestine, A.G.; Mourani, P.M.; McCourt, E.A.; Oliver, S.C.; Abman, S.H. The Relationship of Novel Plasma Proteins in the Early Neonatal Period With Retinopathy of Prematurity. Investig. Ophthalmol. Vis. Sci. 2016, 57, 5076–5082. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  48. Spierer, A.; Rabinowitz, R.; Pri-Chen, S.; Rosner, M. An increase in superoxide dismutase ameliorates oxygen-induced retinopathy in transgenic mice. Eye 2005, 19, 86–91. [Google Scholar] [CrossRef]
  49. Boskabadi, H.; Marefat, M.; Maamouri, G.; Abrishami, M.; Abrishami, M.; Shoeibi, N.; Sanjari, M.S.; Mobarhan, M.G.; Shojaei, S.R.H.; Tavallaei, S.; et al. Evaluation of pro-oxidant antioxidant balance in retinopathy of prematurity. Eye 2021. [Google Scholar] [CrossRef]
  50. Banjac, L.; Banjac, G.; Kotur-Stevuljevic, J.; Spasojevic-Kalimanovska, V.; Gojkovic, T.; Bogavac-Stanojevic, N.; Jelic-Ivanovic, Z.; Banjac, G. Pro-Oxidants and Antioxidants in Retinopathy of Prematurity. Acta Clin. Croat. 2018, 57, 458–463. [Google Scholar] [CrossRef] [Green Version]
  51. Ozieblo-Kupczyk, M.; Bakunowicz-Lazarczyk, A.; Dzienis, K.; Skrzydlewska, E.; Szczepanski, M.; Waszkiewiczz, E. The estimation of selected parameters in antioxidant system in red blood cells in ROP screening of premature infants. Klin. Ocz. 2006, 108, 413–415. [Google Scholar]
  52. Kumar, A.; Ranjan, R.; Basu, S.; Khanna, H.D.; Bhargava, V. Antioxidant levels in cord blood of low birth weight newborns. Indian Pediatr. 2008, 45, 583–585. [Google Scholar] [PubMed]
  53. Ramiro-Cortijo, D.; Lopez de Pablo, A.L.; Lopez-Gimenez, M.R.; Martin, C.R.; Brown, J.; de Pipaon, M.S.; Arribas, S.M. Plasma Oxidative Status in Preterm Infants Receiving LCPUFA Supplementation: A Pilot Study. Nutrients 2020, 12, 122. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Danielsson, H.; Tebani, A.; Zhong, W.; Fagerberg, L.; Brusselaers, N.; Hard, A.L.; Uhlen, M.; Hellstrom, A. Blood protein profiles related to preterm birth and retinopathy of prematurity. Pediatr. Res. 2021. [Google Scholar] [CrossRef]
  55. Yang, Y.; Wu, Z.; Li, S.; Yang, M.; Xiao, X.; Lian, C.; Wen, W.; He, H.; Zeng, J.; Wang, J.; et al. Targeted Blood Metabolomic Study on Retinopathy of Prematurity. Investig. Ophthalmol. Vis. Sci. 2020, 61, 12. [Google Scholar] [CrossRef] [Green Version]
  56. Hozyasz, K.K.; Oltarzewski, M.; Dudkiewicz, Z. Malonylcarnitine in newborns with non-syndromic cleft lip with or without cleft palate. Int. J. Oral Sci. 2010, 2, 136–141. [Google Scholar] [CrossRef] [Green Version]
  57. Lee, S.H.; Ko, J.M.; Song, M.K.; Song, J.; Park, K.S. A Korean child diagnosed with malonic aciduria harboring a novel start codon mutation following presentation with dilated cardiomyopathy. Mol. Genet. Genom. Med. 2020, 8, e1379. [Google Scholar] [CrossRef]
  58. Foster, D.W. Malonyl-CoA: The regulator of fatty acid synthesis and oxidation. J. Clin. Investig. 2012, 122, 1958–1959. [Google Scholar] [CrossRef] [Green Version]
  59. Zhou, Y.; Xu, Y.; Zhang, X.; Zhao, P.; Gong, X.; He, M.; Cao, J.; Jiang, B.; Yoshida, S.; Li, Y. Plasma metabolites in treatment-requiring retinopathy of prematurity: Potential biomarkers identified by metabolomics. Exp. Eye Res. 2020, 199, 108198. [Google Scholar] [CrossRef] [PubMed]
  60. Tomita, Y.; Cagnone, G.; Fu, Z.; Cakir, B.; Kotoda, Y.; Asakage, M.; Wakabayashi, Y.; Hellstrom, A.; Joyal, J.S.; Talukdar, S.; et al. Vitreous metabolomics profiling of proliferative diabetic retinopathy. Diabetologia 2021, 64, 70–82. [Google Scholar] [CrossRef] [PubMed]
  61. Paris, L.P.; Johnson, C.H.; Aguilar, E.; Usui, Y.; Cho, K.; Hoang, L.T.; Feitelberg, D.; Benton, H.P.; Westenskow, P.D.; Kurihara, T.; et al. Global metabolomics reveals metabolic dysregulation in ischemic retinopathy. Metabolomics 2016, 12, 15. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Fouda, A.Y.; Eldahshan, W.; Narayanan, S.P.; Caldwell, R.W.; Caldwell, R.B. Arginase Pathway in Acute Retina and Brain Injury: Therapeutic Opportunities and Unexplored Avenues. Front. Pharmacol. 2020, 11, 277. [Google Scholar] [CrossRef] [PubMed]
  63. Neu, J.; Afzal, A.; Pan, H.; Gallego, E.; Li, N.; Li Calzi, S.; Caballero, S.; Spoerri, P.E.; Shaw, L.C.; Grant, M.B. The dipeptide Arg-Gln inhibits retinal neovascularization in the mouse model of oxygen-induced retinopathy. Investig. Ophthalmol. Vis. Sci. 2006, 47, 3151–3155. [Google Scholar] [CrossRef] [Green Version]
  64. Kim, B.; Li, J.; Jang, C.; Arany, Z. Glutamine fuels proliferation but not migration of endothelial cells. EMBO J. 2017, 36, 2321–2333. [Google Scholar] [CrossRef]
  65. McLeod, D.S.; D’Anna, S.A.; Lutty, G.A. Clinical and histopathologic features of canine oxygen-induced proliferative retinopathy. Investig. Ophthalmol. Vis. Sci. 1998, 39, 1918–1932. [Google Scholar]
  66. Kremer, I.; Kissun, R.; Nissenkorn, I.; Ben-Sira, I.; Garner, A. Oxygen-induced retinopathy in newborn kittens. A model for ischemic vasoproliferative retinopathy. Investig. Ophthalmol. Vis. Sci. 1987, 28, 126–130. [Google Scholar]
  67. Ricci, B. Oxygen-induced retinopathy in the rat model. Doc. Ophthalmol. Proc. Ser. 1990, 74, 171–177. [Google Scholar] [CrossRef] [PubMed]
  68. Pierce, E.A.; Foley, E.D.; Smith, L.E. Regulation of vascular endothelial growth factor by oxygen in a model of retinopathy of prematurity. Arch. Ophthalmol. 1996, 114, 1219–1228. [Google Scholar] [CrossRef]
  69. Rabinowitz, R.; Priel, A.; Rosner, M.; Pri-Chen, S.; Spierer, A. Avastin treatment reduces retinal neovascularization in a mouse model of retinopathy of prematurity. Curr. Eye Res 2012, 37, 624–629. [Google Scholar] [CrossRef]
  70. Jiang, C.; Ruan, L.; Zhang, J.; Huang, X. Inhibitory Effects On Retinal Neovascularization by Ranibizumab and sTie2-Fc in An Oxygen-Induced Retinopathy Mouse Model. Curr. Eye Res 2018, 43, 1190–1198. [Google Scholar] [CrossRef]
  71. Sone, H.; Kawakami, Y.; Segawa, T.; Okuda, Y.; Sekine, Y.; Honmura, S.; Segawa, T.; Suzuki, H.; Yamashita, K.; Yamada, N. Effects of intraocular or systemic administration of neutralizing antibody against vascular endothelial growth factor on the murine experimental model of retinopathy. Life Sci. 1999, 65, 2573–2580. [Google Scholar] [CrossRef]
  72. Semenza, G.L.; Nejfelt, M.K.; Chi, S.M.; Antonarakis, S.E. Hypoxia-inducible nuclear factors bind to an enhancer element located 3’ to the human erythropoietin gene. Proc. Natl. Acad. Sci. USA 1991, 88, 5680–5684. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Semenza, G.L.; Wang, G.L. A nuclear factor induced by hypoxia via de novo protein synthesis binds to the human erythropoietin gene enhancer at a site required for transcriptional activation. Mol. Cell. Biol. 1992, 12, 5447–5454. [Google Scholar] [CrossRef] [Green Version]
  74. Semenza, G.L. Hypoxia-inducible factor 1: Master regulator of O2 homeostasis. Curr. Opin. Genet. Dev. 1998, 8, 588–594. [Google Scholar] [CrossRef]
  75. Miwa, Y.; Hoshino, Y.; Shoda, C.; Jiang, X.; Tsubota, K.; Kurihara, T. Pharmacological HIF inhibition prevents retinal neovascularization with improved visual function in a murine oxygen-induced retinopathy model. Neurochem. Int. 2019, 128, 21–31. [Google Scholar] [CrossRef] [PubMed]
  76. Usui-Ouchi, A.; Aguilar, E.; Murinello, S.; Prins, M.; Gantner, M.L.; Wright, P.E.; Berlow, R.B.; Friedlander, M. An allosteric peptide inhibitor of HIF-1alpha regulates hypoxia-induced retinal neovascularization. Proc. Natl. Acad. Sci. USA 2020, 117, 28297–28306. [Google Scholar] [CrossRef]
  77. Hoppe, G.; Bolok, Y.; McCollum, L.; Zhang, J.; Sears, J.E. Rank Order of Small Molecule Induced Hypoxiamimesis to Prevent Retinopathy of Prematurity. Front. Cell Dev. Biol. 2020, 8, 488. [Google Scholar] [CrossRef]
  78. Singh, C.; Hoppe, G.; Tran, V.; McCollum, L.; Bolok, Y.; Song, W.; Sharma, A.; Brunengraber, H.; Sears, J.E. Serine and 1-carbon metabolism are required for HIF-mediated protection against retinopathy of prematurity. JCI Insight 2019, 4, e129398. [Google Scholar] [CrossRef]
  79. Hoppe, G.; Yoon, S.; Gopalan, B.; Savage, A.R.; Brown, R.; Case, K.; Vasanji, A.; Chan, E.R.; Silver, R.B.; Sears, J.E. Comparative systems pharmacology of HIF stabilization in the prevention of retinopathy of prematurity. Proc. Natl. Acad. Sci. USA 2016, 113, E2516–E2525. [Google Scholar] [CrossRef] [Green Version]
  80. Gantner, M.L.; Eade, K.; Wallace, M.; Handzlik, M.K.; Fallon, R.; Trombley, J.; Bonelli, R.; Giles, S.; Harkins-Perry, S.; Heeren, T.F.C.; et al. Serine and Lipid Metabolism in Macular Disease and Peripheral Neuropathy. N. Engl. J. Med. 2019, 381, 1422–1433. [Google Scholar] [CrossRef]
  81. Shen, W.; Lee, S.R.; Mathai, A.E.; Zhang, R.; Du, J.; Yam, M.X.; Pye, V.; Barnett, N.L.; Rayner, C.L.; Zhu, L.; et al. Effect of selectively knocking down key metabolic genes in Muller glia on photoreceptor health. Glia 2021, 69, 1966–1986. [Google Scholar] [CrossRef]
  82. Guo, D.; Murdoch, C.E.; Xu, H.; Shi, H.; Duan, D.D.; Ahmed, A.; Gu, Y. Vascular endothelial growth factor signaling requires glycine to promote angiogenesis. Sci. Rep. 2017, 7, 14749. [Google Scholar] [CrossRef]
  83. Lu, F.; Liu, Y.; Guo, Y.; Gao, Y.; Piao, Y.; Tan, S.; Tang, Y. Metabolomic changes of blood plasma associated with two phases of rat OIR. Exp. Eye Res. 2020, 190, 107855. [Google Scholar] [CrossRef] [PubMed]
  84. Dungan, K.M.; Braithwaite, S.S.; Preiser, J.C. Stress hyperglycaemia. Lancet 2009, 373, 1798–1807. [Google Scholar] [CrossRef]
  85. Pelikanova, T. Diabetic retinopathy: Pathogenesis and therapeutic implications. Vnitr. Lek. 2016, 62, 620–628. [Google Scholar]
  86. Au, S.C.; Tang, S.M.; Rong, S.S.; Chen, L.J.; Yam, J.C. Association between hyperglycemia and retinopathy of prematurity: A systemic review and meta-analysis. Sci. Rep. 2015, 5, 9091. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  87. Ahmadpour-Kacho, M.; Motlagh, A.J.; Rasoulinejad, S.A.; Jahangir, T.; Bijani, A.; Pasha, Y.Z. Correlation between hyperglycemia and retinopathy of prematurity. Pediatr. Int. 2014, 56, 726–730. [Google Scholar] [CrossRef] [PubMed]
  88. Garg, R.; Agthe, A.G.; Donohue, P.K.; Lehmann, C.U. Hyperglycemia and retinopathy of prematurity in very low birth weight infants. J. Perinatol. Off. J. Calif. Perinat. Assoc. 2003, 23, 186–194. [Google Scholar] [CrossRef] [Green Version]
  89. Mohamed, S.; Murray, J.C.; Dagle, J.M.; Colaizy, T. Hyperglycemia as a risk factor for the development of retinopathy of prematurity. BMC Pediatr. 2013, 13, 78. [Google Scholar] [CrossRef] [Green Version]
  90. Kaempf, J.W.; Kaempf, A.J.; Wu, Y.; Stawarz, M.; Niemeyer, J.; Grunkemeier, G. Hyperglycemia, insulin and slower growth velocity may increase the risk of retinopathy of prematurity. J. Perinatol. Off. J. Calif. Perinat. Assoc. 2011, 31, 251–257. [Google Scholar] [CrossRef] [Green Version]
  91. Mohsen, L.; Abou-Alam, M.; El-Dib, M.; Labib, M.; Elsada, M.; Aly, H. A prospective study on hyperglycemia and retinopathy of prematurity. J. Perinatol. Off. J. Calif. Perinat. Assoc. 2014, 34, 453–457. [Google Scholar] [CrossRef]
  92. Ertl, T.; Gyarmati, J.; Gaal, V.; Szabo, I. Relationship between hyperglycemia and retinopathy of prematurity in very low birth weight infants. Biol. Neonate 2006, 89, 56–59. [Google Scholar] [CrossRef]
  93. Chavez-Valdez, R.; McGowan, J.; Cannon, E.; Lehmann, C.U. Contribution of early glycemic status in the development of severe retinopathy of prematurity in a cohort of ELBW infants. J. Perinatol. Off. J. Calif. Perinat. Assoc. 2011, 31, 749–756. [Google Scholar] [CrossRef] [Green Version]
  94. Cakir, B.; Hellstrom, W.; Tomita, Y.; Fu, Z.; Liegl, R.; Winberg, A.; Hansen-Pupp, I.; Ley, D.; Hellstrom, A.; Lofqvist, C.; et al. IGF1, serum glucose, and retinopathy of prematurity in extremely preterm infants. JCI Insight 2020, 5, e140363. [Google Scholar] [CrossRef]
  95. Lei, C.; Duan, J.; Ge, G.; Zhang, M. Association between neonatal hyperglycemia and retinopathy of prematurity: A meta-analysis. Eur. J. Pediatr. 2021. [Google Scholar] [CrossRef] [PubMed]
  96. Vannadil, H.; Moulick, P.S.; Khan, M.A.; Shankar, S.; Kaushik, J.; Sati, A. Hyperglycaemia as a risk factor for the development of retinopathy of prematurity: A cohort study. Med. J. Armed Forces India 2020, 76, 95–102. [Google Scholar] [CrossRef] [PubMed]
  97. Lee, R.; Wong, T.Y.; Sabanayagam, C. Epidemiology of diabetic retinopathy, diabetic macular edema and related vision loss. Eye Vis. 2015, 2, 17. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  98. Cheung, N.; Mitchell, P.; Wong, T.Y. Diabetic retinopathy. Lancet 2010, 376, 124–136. [Google Scholar] [CrossRef]
  99. Fu, Z.; Lofqvist, C.A.; Liegl, R.; Wang, Z.; Sun, Y.; Gong, Y.; Liu, C.H.; Meng, S.S.; Burnim, S.B.; Arellano, I.; et al. Photoreceptor glucose metabolism determines normal retinal vascular growth. EMBO Mol. Med. 2018, 10, 76–90. [Google Scholar] [CrossRef]
  100. Fu, Z.; Sun, Y.; Cakir, B.; Tomita, Y.; Huang, S.; Wang, Z.; Liu, C.H.; Cho, S.C.; Britton, W.; Kern, T.S.; et al. Targeting Neurovascular Interaction in Retinal Disorders. Int. J. Mol. Sci. 2020, 21, 1503. [Google Scholar] [CrossRef] [Green Version]
  101. Wilson, A.; Sapieha, P. Neurons and guidance cues in retinal vascular diseases. Oncotarget 2016, 7, 9618–9619. [Google Scholar] [CrossRef] [Green Version]
  102. Hoang, Q.V.; Linsenmeier, R.A.; Chung, C.K.; Curcio, C.A. Photoreceptor inner segments in monkey and human retina: Mitochondrial density, optics, and regional variation. Vis. Neurosci. 2002, 19, 395–407. [Google Scholar]
  103. Eelen, G.; de Zeeuw, P.; Treps, L.; Harjes, U.; Wong, B.W.; Carmeliet, P. Endothelial Cell Metabolism. Physiol. Rev. 2018, 98, 3–58. [Google Scholar] [CrossRef]
  104. Eelen, G.; de Zeeuw, P.; Simons, M.; Carmeliet, P. Endothelial cell metabolism in normal and diseased vasculature. Circ. Res. 2015, 116, 1231–1244. [Google Scholar] [CrossRef] [PubMed]
  105. Wong, B.W.; Marsch, E.; Treps, L.; Baes, M.; Carmeliet, P. Endothelial cell metabolism in health and disease: Impact of hypoxia. EMBO J. 2017, 36, 2187–2203. [Google Scholar] [CrossRef]
  106. Narayan, D.S.; Chidlow, G.; Wood, J.P.; Casson, R.J. Glucose metabolism in mammalian photoreceptor inner and outer segments. Clin. Exp. Ophthalmol. 2017, 45, 730–741. [Google Scholar] [CrossRef] [Green Version]
  107. De Bock, K.; Georgiadou, M.; Schoors, S.; Kuchnio, A.; Wong, B.W.; Cantelmo, A.R.; Quaegebeur, A.; Ghesquiere, B.; Cauwenberghs, S.; Eelen, G.; et al. Role of PFKFB3-driven glycolysis in vessel sprouting. Cell 2013, 154, 651–663. [Google Scholar] [CrossRef] [Green Version]
  108. Xu, Y.; An, X.; Guo, X.; Habtetsion, T.G.; Wang, Y.; Xu, X.; Kandala, S.; Li, Q.; Li, H.; Zhang, C.; et al. Endothelial PFKFB3 plays a critical role in angiogenesis. Arterioscler. Thromb. Vasc. Biol. 2014, 34, 1231–1239. [Google Scholar] [CrossRef] [Green Version]
  109. Schoors, S.; De Bock, K.; Cantelmo, A.R.; Georgiadou, M.; Ghesquiere, B.; Cauwenberghs, S.; Kuchnio, A.; Wong, B.W.; Quaegebeur, A.; Goveia, J.; et al. Partial and transient reduction of glycolysis by PFKFB3 blockade reduces pathological angiogenesis. Cell. Metab. 2014, 19, 37–48. [Google Scholar] [CrossRef] [Green Version]
  110. Han, X.; Kong, J.; Hartnett, M.E.; Wang, H. Enhancing Retinal Endothelial Glycolysis by Inhibiting UCP2 Promotes Physiologic Retinal Vascular Development in a Model of Retinopathy of Prematurity. Investig. Ophthalmol. Vis. Sci. 2019, 60, 1604–1613. [Google Scholar] [CrossRef]
  111. Liu, Z.; Yan, S.; Wang, J.; Xu, Y.; Wang, Y.; Zhang, S.; Xu, X.; Yang, Q.; Zeng, X.; Zhou, Y.; et al. Endothelial adenosine A2a receptor-mediated glycolysis is essential for pathological retinal angiogenesis. Nat. Commun. 2017, 8, 584. [Google Scholar] [CrossRef] [Green Version]
  112. Lorenzi, M. The polyol pathway as a mechanism for diabetic retinopathy: Attractive, elusive, and resilient. Exp. Diabetes Res. 2007, 2007, 61038. [Google Scholar] [CrossRef]
  113. Fu, Z.J.; Li, S.Y.; Kociok, N.; Wong, D.; Chung, S.K.; Lo, A.C. Aldose reductase deficiency reduced vascular changes in neonatal mouse retina in oxygen-induced retinopathy. Investig. Ophthalmol. Vis. Sci. 2012, 53, 5698–5712. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Fu, Z.; Nian, S.; Li, S.Y.; Wong, D.; Chung, S.K.; Lo, A.C. Deficiency of aldose reductase attenuates inner retinal neuronal changes in a mouse model of retinopathy of prematurity. Graefes Arch. Clin. Exp. Ophthalmol. 2015, 253, 1503–1513. [Google Scholar] [CrossRef] [PubMed]
  115. Rohlenova, K.; Goveia, J.; Garcia-Caballero, M.; Subramanian, A.; Kalucka, J.; Treps, L.; Falkenberg, K.D.; de Rooij, L.; Zheng, Y.; Lin, L.; et al. Single-Cell RNA Sequencing Maps Endothelial Metabolic Plasticity in Pathological Angiogenesis. Cell Metab. 2020, 31, 862–877.e14. [Google Scholar] [CrossRef] [PubMed]
  116. Neu, J. Glutamine supplements in premature infants: Why and how. J. Pediatr. Gastroenterol. Nutr. 2003, 37, 533–535. [Google Scholar] [CrossRef] [PubMed]
  117. Wu, G.; Jaeger, L.A.; Bazer, F.W.; Rhoads, J.M. Arginine deficiency in preterm infants: Biochemical mechanisms and nutritional implications. J. Nutr. Biochem. 2004, 15, 442–451. [Google Scholar] [CrossRef]
  118. Wu, G.; Haynes, T.E.; Li, H.; Meininger, C.J. Glutamine metabolism in endothelial cells: Ornithine synthesis from glutamine via pyrroline-5-carboxylate synthase. Comp. Biochem. Physiol. A Mol. Integr. Physiol. 2000, 126, 115–123. [Google Scholar] [CrossRef]
  119. Huang, H.; Vandekeere, S.; Kalucka, J.; Bierhansl, L.; Zecchin, A.; Bruning, U.; Visnagri, A.; Yuldasheva, N.; Goveia, J.; Cruys, B.; et al. Role of glutamine and interlinked asparagine metabolism in vessel formation. EMBO J. 2017, 36, 2334–2352. [Google Scholar] [CrossRef]
  120. Eade, K.; Gantner, M.L.; Hostyk, J.A.; Nagasaki, T.; Giles, S.; Fallon, R.; Harkins-Perry, S.; Baldini, M.; Lim, E.W.; Scheppke, L.; et al. Serine biosynthesis defect due to haploinsufficiency of PHGDH causes retinal disease. Nat. Metab. 2021, 3, 366–377. [Google Scholar] [CrossRef]
  121. Vandekeere, S.; Dubois, C.; Kalucka, J.; Sullivan, M.R.; Garcia-Caballero, M.; Goveia, J.; Chen, R.; Diehl, F.F.; Bar-Lev, L.; Souffreau, J.; et al. Serine Synthesis via PHGDH Is Essential for Heme Production in Endothelial Cells. Cell Metab. 2018, 28, 573–587.e13. [Google Scholar] [CrossRef] [Green Version]
  122. Zhang, T.; Gillies, M.C.; Madigan, M.C.; Shen, W.; Du, J.; Grunert, U.; Zhou, F.; Yam, M.; Zhu, L. Disruption of De Novo Serine Synthesis in Muller Cells Induced Mitochondrial Dysfunction and Aggravated Oxidative Damage. Mol. Neurobiol. 2018, 55, 7025–7037. [Google Scholar] [CrossRef]
  123. Zhang, T.; Zhu, L.; Madigan, M.C.; Liu, W.; Shen, W.; Cherepanoff, S.; Zhou, F.; Zeng, S.; Du, J.; Gillies, M.C. Human macular Muller cells rely more on serine biosynthesis to combat oxidative stress than those from the periphery. eLife 2019, 8, e43598. [Google Scholar] [CrossRef]
  124. Becker, S.; Wang, H.; Simmons, A.B.; Suwanmanee, T.; Stoddard, G.J.; Kafri, T.; Hartnett, M.E. Targeted Knockdown of Overexpressed VEGFA or VEGF164 in Muller cells maintains retinal function by triggering different signaling mechanisms. Sci. Rep. 2018, 8, 2003. [Google Scholar] [CrossRef] [Green Version]
  125. Le, Y.Z. VEGF production and signaling in Muller glia are critical to modulating vascular function and neuronal integrity in diabetic retinopathy and hypoxic retinal vascular diseases. Vis. Res. 2017, 139, 108–114. [Google Scholar] [CrossRef]
  126. Schoors, S.; Bruning, U.; Missiaen, R.; Queiroz, K.C.; Borgers, G.; Elia, I.; Zecchin, A.; Cantelmo, A.R.; Christen, S.; Goveia, J.; et al. Fatty acid carbon is essential for dNTP synthesis in endothelial cells. Nature 2015, 520, 192–197. [Google Scholar] [CrossRef] [Green Version]
  127. Wei, X.; Schneider, J.G.; Shenouda, S.M.; Lee, A.; Towler, D.A.; Chakravarthy, M.V.; Vita, J.A.; Semenkovich, C.F. De novo lipogenesis maintains vascular homeostasis through endothelial nitric-oxide synthase (eNOS) palmitoylation. J. Biol. Chem. 2011, 286, 2933–2945. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  128. Elmasri, H.; Karaaslan, C.; Teper, Y.; Ghelfi, E.; Weng, M.; Ince, T.A.; Kozakewich, H.; Bischoff, J.; Cataltepe, S. Fatty acid binding protein 4 is a target of VEGF and a regulator of cell proliferation in endothelial cells. FASEB J. 2009, 23, 3865–3873. [Google Scholar] [CrossRef] [Green Version]
  129. Joyal, J.S.; Gantner, M.L.; Smith, L.E.H. Retinal energy demands control vascular supply of the retina in development and disease: The role of neuronal lipid and glucose metabolism. Prog. Retin. Eye Res. 2018, 64, 131–156. [Google Scholar] [CrossRef] [PubMed]
  130. Higuchi, A.; Ohashi, K.; Kihara, S.; Walsh, K.; Ouchi, N. Adiponectin suppresses pathological microvessel formation in retina through modulation of tumor necrosis factor-alpha expression. Circ. Res. 2009, 104, 1058–1065. [Google Scholar] [CrossRef] [PubMed]
  131. Rice, D.S.; Calandria, J.M.; Gordon, W.C.; Jun, B.; Zhou, Y.; Gelfman, C.M.; Li, S.; Jin, M.; Knott, E.J.; Chang, B.; et al. Adiponectin receptor 1 conserves docosahexaenoic acid and promotes photoreceptor cell survival. Nat Commun. 2015, 6, 6228. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  132. Sluch, V.M.; Banks, A.; Li, H.; Crowley, M.A.; Davis, V.; Xiang, C.; Yang, J.; Demirs, J.T.; Vrouvlianis, J.; Leehy, B.; et al. ADIPOR1 is essential for vision and its RPE expression is lost in the Mfrp(rd6) mouse. Sci. Rep. 2018, 8, 14339. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  133. Fu, Z.; Liegl, R.; Wang, Z.; Gong, Y.; Liu, C.H.; Sun, Y.; Cakir, B.; Burnim, S.B.; Meng, S.S.; Lofqvist, C.; et al. Adiponectin Mediates Dietary Omega-3 Long-Chain Polyunsaturated Fatty Acid Protection Against Choroidal Neovascularization in Mice. Investig. Ophthalmol. Vis. Sci. 2017, 58, 3862–3870. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  134. Zhang, J.; Wang, C.; Li, L.; Man, Q.; Meng, L.; Song, P.; Froyland, L.; Du, Z.Y. Dietary inclusion of salmon, herring and pompano as oily fish reduces CVD risk markers in dyslipidaemic middle-aged and elderly Chinese women. Br. J. Nutr. 2012, 108, 1455–1465. [Google Scholar] [CrossRef] [Green Version]
  135. Olza, J.; Mesa, M.D.; Aguilera, C.M.; Moreno-Torres, R.; Jimenez, A.; Perez de la Cruz, A.; Gil, A. Influence of an eicosapentaenoic and docosahexaenoic acid-enriched enteral nutrition formula on plasma fatty acid composition and biomarkers of insulin resistance in the elderly. Clin. Nutr. 2010, 29, 31–37. [Google Scholar] [CrossRef]
  136. Kuda, O.; Jelenik, T.; Jilkova, Z.; Flachs, P.; Rossmeisl, M.; Hensler, M.; Kazdova, L.; Ogston, N.; Baranowski, M.; Gorski, J.; et al. n-3 fatty acids and rosiglitazone improve insulin sensitivity through additive stimulatory effects on muscle glycogen synthesis in mice fed a high-fat diet. Diabetologia 2009, 52, 941–951. [Google Scholar] [CrossRef] [Green Version]
  137. Prostek, A.; Gajewska, M.; Kamola, D.; Balasinska, B. The influence of EPA and DHA on markers of inflammation in 3T3-L1 cells at different stages of cellular maturation. Lipids Health Dis. 2014, 13, 3. [Google Scholar] [CrossRef] [Green Version]
  138. Holland, W.L.; Adams, A.C.; Brozinick, J.T.; Bui, H.H.; Miyauchi, Y.; Kusminski, C.M.; Bauer, S.M.; Wade, M.; Singhal, E.; Cheng, C.C.; et al. An FGF21-adiponectin-ceramide axis controls energy expenditure and insulin action in mice. Cell Metab. 2013, 17, 790–797. [Google Scholar] [CrossRef] [Green Version]
  139. Owen, B.M.; Mangelsdorf, D.J.; Kliewer, S.A. Tissue-specific actions of the metabolic hormones FGF15/19 and FGF21. Trends Endocrinol. Metab. 2015, 26, 22–29. [Google Scholar] [CrossRef] [Green Version]
  140. Kharitonenkov, A.; Larsen, P. FGF21 reloaded: Challenges of a rapidly growing field. Trends Endocrinol. Metab. 2011, 22, 81–86. [Google Scholar] [CrossRef]
  141. Lin, Z.; Gong, Q.; Wu, C.; Yu, J.; Lu, T.; Pan, X.; Lin, S.; Li, X. Dynamic change of serum FGF21 levels in response to glucose challenge in human. J. Clin. Endocrinol. Metab. 2012, 97, E1224–E1228. [Google Scholar] [CrossRef] [Green Version]
  142. Markan, K.R.; Naber, M.C.; Ameka, M.K.; Anderegg, M.D.; Mangelsdorf, D.J.; Kliewer, S.A.; Mohammadi, M.; Potthoff, M.J. Circulating FGF21 is liver derived and enhances glucose uptake during refeeding and overfeeding. Diabetes 2014, 63, 4057–4063. [Google Scholar] [CrossRef] [Green Version]
  143. Cuevas-Ramos, D.; Mehta, R.; Aguilar-Salinas, C.A. Fibroblast Growth Factor 21 and Browning of White Adipose Tissue. Front. Physiol. 2019, 10, 37. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  144. Fu, Z.; Gong, Y.; Liegl, R.; Wang, Z.; Liu, C.H.; Meng, S.S.; Burnim, S.B.; Saba, N.J.; Fredrick, T.W.; Morss, P.C.; et al. FGF21 Administration Suppresses Retinal and Choroidal Neovascularization in Mice. Cell Rep. 2017, 18, 1606–1613. [Google Scholar] [CrossRef] [PubMed]
  145. Fu, Z.; Wang, Z.; Liu, C.H.; Gong, Y.; Cakir, B.; Liegl, R.; Sun, Y.; Meng, S.S.; Burnim, S.B.; Arellano, I.; et al. Fibroblast Growth Factor 21 Protects Photoreceptor Function in Type 1 Diabetic Mice. Diabetes 2018, 67, 974–985. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  146. Fu, Z.; Qiu, C.; Cagnone, G.; Tomita, Y.; Huang, S.; Cakir, B.; Kotoda, Y.; Allen, W.; Bull, E.; Akula, J.D.; et al. Retinal glial remodeling by FGF21 preserves retinal function during photoreceptor degeneration. iScience 2021, 24, 102376. [Google Scholar] [CrossRef]
  147. Sanchez-Infantes, D.; Gallego-Escuredo, J.M.; Diaz, M.; Aragones, G.; Sebastiani, G.; Lopez-Bermejo, A.; de Zegher, F.; Domingo, P.; Villarroya, F.; Ibanez, L. Circulating FGF19 and FGF21 surge in early infancy from infra- to supra-adult concentrations. Int. J. Obes. 2015, 39, 742–746. [Google Scholar] [CrossRef]
  148. Guasti, L.; Silvennoinen, S.; Bulstrode, N.W.; Ferretti, P.; Sankilampi, U.; Dunkel, L. Elevated FGF21 leads to attenuated postnatal linear growth in preterm infants through GH resistance in chondrocytes. J. Clin. Endocrinol. Metab. 2014, 99, E2198–E2206. [Google Scholar] [CrossRef] [Green Version]
  149. Mericq, V.; De Luca, F.; Hernandez, M.I.; Pena, V.; Rossel, K.; Garcia, M.; Avila, A.; Cavada, G.; Iniguez, G. Serum fibroblast growth factor 21 levels are inversely associated with growth rates in infancy. Horm. Res. Paediatr. 2014, 82, 324–331. [Google Scholar] [CrossRef]
  150. Daughaday, W.H.; Rotwein, P. Insulin-like growth factors I and II. Peptide, messenger ribonucleic acid and gene structures, serum, and tissue concentrations. Endocr. Rev. 1989, 10, 68–91. [Google Scholar] [CrossRef]
  151. Liegl, R.; Lofqvist, C.; Hellstrom, A.; Smith, L.E. IGF-1 in retinopathy of prematurity, a CNS neurovascular disease. Early Hum. Dev. 2016, 102, 13–19. [Google Scholar] [CrossRef] [Green Version]
  152. Hellstrom, A.; Perruzzi, C.; Ju, M.; Engstrom, E.; Hard, A.L.; Liu, J.L.; Albertsson-Wikland, K.; Carlsson, B.; Niklasson, A.; Sjodell, L.; et al. Low IGF-I suppresses VEGF-survival signaling in retinal endothelial cells: Direct correlation with clinical retinopathy of prematurity. Proc. Natl. Acad. Sci. USA 2001, 98, 5804–5808. [Google Scholar] [CrossRef] [Green Version]
  153. Hard, A.L.; Smith, L.E.; Hellstrom, A. Nutrition, insulin-like growth factor-1 and retinopathy of prematurity. Semin. Fetal Neonatal. Med. 2013, 18, 136–142. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  154. Hellstrom, A.; Engstrom, E.; Hard, A.L.; Albertsson-Wikland, K.; Carlsson, B.; Niklasson, A.; Lofqvist, C.; Svensson, E.; Holm, S.; Ewald, U.; et al. Postnatal serum insulin-like growth factor I deficiency is associated with retinopathy of prematurity and other complications of premature birth. Pediatrics 2003, 112, 1016–1020. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  155. Jensen, A.K.; Ying, G.S.; Huang, J.; Quinn, G.E.; Binenbaum, G. Postnatal Serum Insulin-Like Growth Factor I and Retinopathy of Prematurity. Retina 2017, 37, 867–872. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  156. Hellgren, G.; Lundgren, P.; Pivodic, A.; Lofqvist, C.; Nilsson, A.K.; Ley, D.; Savman, K.; Smith, L.E.; Hellstrom, A. Decreased Platelet Counts and Serum Levels of VEGF-A, PDGF-BB, and BDNF in Extremely Preterm Infants Developing Severe ROP. Neonatology 2021, 118, 18–27. [Google Scholar] [CrossRef] [PubMed]
  157. Smith, L.E.; Shen, W.; Perruzzi, C.; Soker, S.; Kinose, F.; Xu, X.; Robinson, G.; Driver, S.; Bischoff, J.; Zhang, B.; et al. Regulation of vascular endothelial growth factor-dependent retinal neovascularization by insulin-like growth factor-1 receptor. Nat. Med. 1999, 5, 1390–1395. [Google Scholar] [CrossRef] [PubMed]
  158. Vanhaesebrouck, S.; Daniels, H.; Moons, L.; Vanhole, C.; Carmeliet, P.; De Zegher, F. Oxygen-induced retinopathy in mice: Amplification by neonatal IGF-I deficit and attenuation by IGF-I administration. Pediatr. Res. 2009, 65, 307–310. [Google Scholar] [CrossRef] [Green Version]
  159. Ley, D.; Hallberg, B.; Hansen-Pupp, I.; Dani, C.; Ramenghi, L.A.; Marlow, N.; Beardsall, K.; Bhatti, F.; Dunger, D.; Higginson, J.D.; et al. rhIGF-1/rhIGFBP-3 in Preterm Infants: A Phase 2 Randomized Controlled Trial. J. Pediatr. 2019, 206, 56–65.e8. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  160. Cakir, B.; Liegl, R.; Hellgren, G.; Lundgren, P.; Sun, Y.; Klevebro, S.; Lofqvist, C.; Mannheimer, C.; Cho, S.; Poblete, A.; et al. Thrombocytopenia is associated with severe retinopathy of prematurity. JCI Insight 2018, 3, e99448. [Google Scholar] [CrossRef]
  161. Jensen, A.K.; Ying, G.S.; Huang, J.; Quinn, G.E.; Binenbaum, G. Longitudinal study of the association between thrombocytopenia and retinopathy of prematurity. J. AAPOS Off. Publ. Am. Assoc. Pediatr. Ophthalmol. Strabismus 2018, 22, 119–123. [Google Scholar] [CrossRef]
  162. Keech, A.C.; Mitchell, P.; Summanen, P.A.; O’Day, J.; Davis, T.M.; Moffitt, M.S.; Taskinen, M.R.; Simes, R.J.; Tse, D.; Williamson, E.; et al. Effect of fenofibrate on the need for laser treatment for diabetic retinopathy (FIELD study): A randomised controlled trial. Lancet 2007, 370, 1687–1697. [Google Scholar] [CrossRef]
  163. Group, A.S.; Group, A.E.S.; Chew, E.Y.; Ambrosius, W.T.; Davis, M.D.; Danis, R.P.; Gangaputra, S.; Greven, C.M.; Hubbard, L.; Esser, B.A.; et al. Effects of medical therapies on retinopathy progression in type 2 diabetes. N. Engl. J. Med. 2010, 363, 233–244. [Google Scholar] [CrossRef] [Green Version]
  164. Najib, J. Fenofibrate in the treatment of dyslipidemia: A review of the data as they relate to the new suprabioavailable tablet formulation. Clin. Ther. 2002, 24, 2022–2050. [Google Scholar] [CrossRef]
  165. Chen, Y.; Hu, Y.; Lin, M.; Jenkins, A.J.; Keech, A.C.; Mott, R.; Lyons, T.J.; Ma, J.X. Therapeutic effects of PPARalpha agonists on diabetic retinopathy in type 1 diabetes models. Diabetes 2013, 62, 261–272. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  166. Csaicsich, D.; Russo-Schlaff, N.; Messerschmidt, A.; Weninger, M.; Pollak, A.; Aufricht, C. Renal failure, comorbidity and mortality in preterm infants. Wien Klin. Wochenschr. 2008, 120, 153–157. [Google Scholar] [CrossRef] [PubMed]
  167. Arai, H.; Yamashita, S.; Yokote, K.; Araki, E.; Suganami, H.; Ishibashi, S.; Group, K.S. Efficacy and Safety of Pemafibrate Versus Fenofibrate in Patients with High Triglyceride and Low HDL Cholesterol Levels: A Multicenter, Placebo-Controlled, Double-Blind, Randomized Trial. J. Atheroscler. Thromb. 2018, 25, 521–538. [Google Scholar] [CrossRef] [Green Version]
  168. Yamazaki, Y.; Abe, K.; Toma, T.; Nishikawa, M.; Ozawa, H.; Okuda, A.; Araki, T.; Oda, S.; Inoue, K.; Shibuya, K.; et al. Design and synthesis of highly potent and selective human peroxisome proliferator-activated receptor alpha agonists. Bioorgan. Med. Chem. Lett. 2007, 17, 4689–4693. [Google Scholar] [CrossRef] [PubMed]
  169. Tomita, Y.; Lee, D.; Tsubota, K.; Kurihara, T. PPARalpha Agonist Oral Therapy in Diabetic Retinopathy. Biomedicines 2020, 8, 433. [Google Scholar] [CrossRef] [PubMed]
  170. Tomita, Y.; Ozawa, N.; Miwa, Y.; Ishida, A.; Ohta, M.; Tsubota, K.; Kurihara, T. Pemafibrate Prevents Retinal Pathological Neovascularization by Increasing FGF21 Level in a Murine Oxygen-Induced Retinopathy Model. Int. J. Mol. Sci. 2019, 20, 5878. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  171. Tomita, Y.; Lee, D.; Miwa, Y.; Jiang, X.; Ohta, M.; Tsubota, K.; Kurihara, T. Pemafibrate Protects Against Retinal Dysfunction in a Murine Model of Diabetic Retinopathy. Int. J. Mol. Sci. 2020, 21, 6243. [Google Scholar] [CrossRef]
  172. Swarbrick, A.W.; Frederiks, A.J.; Foster, R.S. Systematic review of sirolimus in dermatological conditions. Australas. J. Derm. 2021. [Google Scholar] [CrossRef]
  173. Yu, J.; Parkhitko, A.A.; Henske, E.P. Mammalian target of rapamycin signaling and autophagy: Roles in lymphangioleiomyomatosis therapy. Proc. Am. Thorac. Soc. 2010, 7, 48–53. [Google Scholar] [CrossRef] [PubMed]
  174. Saunders, R.N.; Metcalfe, M.S.; Nicholson, M.L. Rapamycin in transplantation: A review of the evidence. Kidney Int. 2001, 59, 3–16. [Google Scholar] [CrossRef] [Green Version]
  175. Saxton, R.A.; Sabatini, D.M. mTOR Signaling in Growth, Metabolism, and Disease. Cell 2017, 169, 361–371. [Google Scholar] [CrossRef]
  176. Cheon, S.Y.; Cho, K. Lipid metabolism, inflammation, and foam cell formation in health and metabolic disorders: Targeting mTORC1. J. Mol. Med. 2021. [Google Scholar] [CrossRef] [PubMed]
  177. Dejneka, N.S.; Kuroki, A.M.; Fosnot, J.; Tang, W.; Tolentino, M.J.; Bennett, J. Systemic rapamycin inhibits retinal and choroidal neovascularization in mice. Mol. Vis. 2004, 10, 964–972. [Google Scholar]
  178. Yagasaki, R.; Nakahara, T.; Ushikubo, H.; Mori, A.; Sakamoto, K.; Ishii, K. Anti-angiogenic effects of mammalian target of rapamycin inhibitors in a mouse model of oxygen-induced retinopathy. Biol. Pharm. Bull. 2014, 37, 1838–1842. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  179. Zhang, J.; Zhu, M.; Ruan, L.; Jiang, C.; Yang, Q.; Chang, Q.; Huang, X. Protective effects of rapamycin on the retinal vascular bed during the vaso-obliteration phase in mouse oxygen-induced retinopathy model. FASEB J. 2020, 34, 15822–15836. [Google Scholar] [CrossRef] [PubMed]
  180. Riento, K.; Ridley, A.J. Rocks: Multifunctional kinases in cell behaviour. Nat. Rev. Mol. Cell Biol. 2003, 4, 446–456. [Google Scholar] [CrossRef] [PubMed]
  181. Arita, R.; Hata, Y.; Nakao, S.; Kita, T.; Miura, M.; Kawahara, S.; Zandi, S.; Almulki, L.; Tayyari, F.; Shimokawa, H.; et al. Rho kinase inhibition by fasudil ameliorates diabetes-induced microvascular damage. Diabetes 2009, 58, 215–226. [Google Scholar] [CrossRef] [Green Version]
  182. Kan, L.; Smith, A.; Chen, M.; Ledford, B.T.; Fan, H.; Liu, Z.; He, J.Q. Rho-Associated Kinase Inhibitor (Y-27632) Attenuates Doxorubicin-Induced Apoptosis of Human Cardiac Stem Cells. PLoS ONE 2015, 10, e0144513. [Google Scholar] [CrossRef] [PubMed]
  183. Nakagawa, O.; Fujisawa, K.; Ishizaki, T.; Saito, Y.; Nakao, K.; Narumiya, S. ROCK-I and ROCK-II, two isoforms of Rho-associated coiled-coil forming protein serine/threonine kinase in mice. FEBS Lett. 1996, 392, 189–193. [Google Scholar] [CrossRef] [Green Version]
  184. Noda, K.; Nakajima, S.; Godo, S.; Saito, H.; Ikeda, S.; Shimizu, T.; Enkhjargal, B.; Fukumoto, Y.; Tsukita, S.; Yamada, T.; et al. Rho-kinase inhibition ameliorates metabolic disorders through activation of AMPK pathway in mice. PLoS ONE 2014, 9, e110446. [Google Scholar] [CrossRef]
  185. Fang, X.; Ueno, M.; Yamashita, T.; Ikuno, Y. RhoA activation and effect of Rho-kinase inhibitor in the development of retinal neovascularization in a mouse model of oxygen-induced retinopathy. Curr. Eye Res 2011, 36, 1028–1036. [Google Scholar] [CrossRef]
  186. Yamaguchi, M.; Nakao, S.; Arita, R.; Kaizu, Y.; Arima, M.; Zhou, Y.; Kita, T.; Yoshida, S.; Kimura, K.; Isobe, T.; et al. Vascular Normalization by ROCK Inhibitor: Therapeutic Potential of Ripasudil (K-115) Eye Drop in Retinal Angiogenesis and Hypoxia. Investig. Ophthalmol. Vis. Sci. 2016, 57, 2264–2276. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  187. Hollanders, K.; Hove, I.V.; Sergeys, J.; Bergen, T.V.; Lefevere, E.; Kindt, N.; Castermans, K.; Vandewalle, E.; van Pelt, J.; Moons, L.; et al. AMA0428, A Potent Rock Inhibitor, Attenuates Early and Late Experimental Diabetic Retinopathy. Curr. Eye Res 2017, 42, 260–272. [Google Scholar] [CrossRef] [PubMed]
  188. Blasiak, J.; Petrovski, G.; Vereb, Z.; Facsko, A.; Kaarniranta, K. Oxidative stress, hypoxia, and autophagy in the neovascular processes of age-related macular degeneration. Biomed. Res. Int. 2014, 2014, 768026. [Google Scholar] [CrossRef] [PubMed]
  189. Pesce, N.A.; Canovai, A.; Lardner, E.; Cammalleri, M.; Kvanta, A.; Andre, H.; Dal Monte, M. Autophagy Involvement in the Postnatal Development of the Rat Retina. Cells 2021, 10, 177. [Google Scholar] [CrossRef]
  190. Wang, S.; Ji, L.Y.; Li, L.; Li, J.M. Oxidative stress, autophagy and pyroptosis in the neovascularization of oxygeninduced retinopathy in mice. Mol. Med. Rep. 2019, 19, 927–934. [Google Scholar] [CrossRef] [Green Version]
  191. Ji, L.; Li, L.; Zhao, Y.; Liu, S.; Li, J.; Zhang, J.; Zhao, Q.; Wang, S. Immunosubunit beta5i Knockout Suppresses Neovascularization and Restores Autophagy in Retinal Neovascularization by Targeting ATG5 for Degradation. Investig. Ophthalmol. Vis. Sci. 2020, 61, 30. [Google Scholar] [CrossRef]
  192. Subirada, P.V.; Paz, M.C.; Ridano, M.E.; Lorenc, V.E.; Fader, C.M.; Chiabrando, G.A.; Sanchez, M.C. Effect of Autophagy Modulators on Vascular, Glial, and Neuronal Alterations in the Oxygen-Induced Retinopathy Mouse Model. Front. Cell. Neurosci. 2019, 13, 279. [Google Scholar] [CrossRef] [Green Version]
  193. Sprott, D.; Poitz, D.M.; Korovina, I.; Ziogas, A.; Phieler, J.; Chatzigeorgiou, A.; Mitroulis, I.; Deussen, A.; Chavakis, T.; Klotzsche-von Ameln, A. Endothelial-Specific Deficiency of ATG5 (Autophagy Protein 5) Attenuates Ischemia-Related Angiogenesis. Arter. Thromb. Vasc. Biol. 2019, 39, 1137–1148. [Google Scholar] [CrossRef] [PubMed]
  194. Smithers, L.G.; Gibson, R.A.; McPhee, A.; Makrides, M. Higher dose of docosahexaenoic acid in the neonatal period improves visual acuity of preterm infants: Results of a randomized controlled trial. Am. J. Clin. Nutr. 2008, 88, 1049–1056. [Google Scholar] [CrossRef] [Green Version]
  195. Dyall, S.C. Interplay Between n-3 and n-6 Long-Chain Polyunsaturated Fatty Acids and the Endocannabinoid System in Brain Protection and Repair. Lipids 2017, 52, 885–900. [Google Scholar] [CrossRef] [PubMed]
  196. Dierge, E.; Debock, E.; Guilbaud, C.; Corbet, C.; Mignolet, E.; Mignard, L.; Bastien, E.; Dessy, C.; Larondelle, Y.; Feron, O. Peroxidation of n-3 and n-6 polyunsaturated fatty acids in the acidic tumor environment leads to ferroptosis-mediated anticancer effects. Cell Metab. 2021. [Google Scholar] [CrossRef] [PubMed]
  197. Shen, W.; Fruttiger, M.; Zhu, L.; Chung, S.H.; Barnett, N.L.; Kirk, J.K.; Lee, S.; Coorey, N.J.; Killingsworth, M.; Sherman, L.S.; et al. Conditional Mullercell ablation causes independent neuronal and vascular pathologies in a novel transgenic model. J. Neurosci. 2012, 32, 15715–15727. [Google Scholar] [CrossRef] [PubMed]
  198. Poitry-Yamate, C.L.; Poitry, S.; Tsacopoulos, M. Lactate released by Muller glial cells is metabolized by photoreceptors from mammalian retina. J. Neurosci. 1995, 15, 5179–5191. [Google Scholar] [CrossRef]
  199. Toft-Kehler, A.K.; Skytt, D.M.; Poulsen, K.A.; Braendstrup, C.T.; Gegelashvili, G.; Waagepetersen, H.; Kolko, M. Limited energy supply in Muller cells alters glutamate uptake. Neurochem. Res. 2014, 39, 941–949. [Google Scholar] [CrossRef]
  200. Kanow, M.A.; Giarmarco, M.M.; Jankowski, C.S.; Tsantilas, K.; Engel, A.L.; Du, J.; Linton, J.D.; Farnsworth, C.C.; Sloat, S.R.; Rountree, A.; et al. Biochemical adaptations of the retina and retinal pigment epithelium support a metabolic ecosystem in the vertebrate eye. eLife 2017, 6. [Google Scholar] [CrossRef]
  201. Bibb, C.; Young, R.W. Renewal of fatty acids in the membranes of visual cell outer segments. J. Cell Biol. 1974, 61, 327–343. [Google Scholar] [CrossRef] [PubMed]
  202. Palczewski, K. Retinoids for treatment of retinal diseases. Trends Pharm. Sci. 2010, 31, 284–295. [Google Scholar] [CrossRef] [Green Version]
  203. Orban, T.; Palczewska, G.; Palczewski, K. Retinyl ester storage particles (retinosomes) from the retinal pigmented epithelium resemble lipid droplets in other tissues. J. Biol. Chem. 2011, 286, 17248–17258. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  204. Puchalska, P.; Crawford, P.A. Multi-dimensional Roles of Ketone Bodies in Fuel Metabolism, Signaling, and Therapeutics. Cell Metab. 2017, 25, 262–284. [Google Scholar] [CrossRef] [Green Version]
  205. Bazan, N.G.; Gordon, W.C.; Rodriguez de Turco, E.B. Docosahexaenoic acid uptake and metabolism in photoreceptors: Retinal conservation by an efficient retinal pigment epithelial cell-mediated recycling process. Adv. Exp. Med. Biol. 1992, 318, 295–306. [Google Scholar] [CrossRef]
  206. Gordon, W.C.; Rodriguez de Turco, E.B.; Bazan, N.G. Retinal pigment epithelial cells play a central role in the conservation of docosahexaenoic acid by photoreceptor cells after shedding and phagocytosis. Curr. Eye Res 1992, 11, 73–83. [Google Scholar] [CrossRef]
Figure 1. ROP progression in premature infants. (A) Schematics of the progression of human retinopathy of prematurity (ROP). Phases 1 and 2 of ROP are associated with different oxygen levels. Loss of essential nutrients and pro-angiogenic growth factors after birth in combination with provision of high supplemental oxygen, leads to hyperoxia that suppresses retinal vascularization (Phase 1). In the second phase of ROP (Phase 2), relative hypoxia and increased nutrient demands of the avascular retina drives fibrovascular proliferation. ROP Phase 2 is defined by anatomic changes, such as the demarcation line (stage 1), ridge (stage 2), extraretinal fibrovascular proliferation (stage 3), partial retinal detachment (stage 4), and total retinal detachment (stage 5). Any stage can develop into aggressive posterior ROP (APROP), which rapidly progresses to tractional retinal detachment (stage 4 or 5). Image made with graphics from ©BioRender (https://biorender.com/ (accessed on 18 October 2021) Agreement number: IA22XF3W0H) (B) Illustration of retinopathy of prematurity (ROP) development, from normal retinal neuro-vascular development, via stage 2 with ridge (arrow), stage 3 with neovascularization and hemorrhage (arrows), stage 3 with plus disease (arrow), APROP with central changes (arrow) and laser treatment (arrow) of stage 3 ROP.
Figure 1. ROP progression in premature infants. (A) Schematics of the progression of human retinopathy of prematurity (ROP). Phases 1 and 2 of ROP are associated with different oxygen levels. Loss of essential nutrients and pro-angiogenic growth factors after birth in combination with provision of high supplemental oxygen, leads to hyperoxia that suppresses retinal vascularization (Phase 1). In the second phase of ROP (Phase 2), relative hypoxia and increased nutrient demands of the avascular retina drives fibrovascular proliferation. ROP Phase 2 is defined by anatomic changes, such as the demarcation line (stage 1), ridge (stage 2), extraretinal fibrovascular proliferation (stage 3), partial retinal detachment (stage 4), and total retinal detachment (stage 5). Any stage can develop into aggressive posterior ROP (APROP), which rapidly progresses to tractional retinal detachment (stage 4 or 5). Image made with graphics from ©BioRender (https://biorender.com/ (accessed on 18 October 2021) Agreement number: IA22XF3W0H) (B) Illustration of retinopathy of prematurity (ROP) development, from normal retinal neuro-vascular development, via stage 2 with ridge (arrow), stage 3 with neovascularization and hemorrhage (arrows), stage 3 with plus disease (arrow), APROP with central changes (arrow) and laser treatment (arrow) of stage 3 ROP.
Life 11 01119 g001
Figure 2. Serum S1P levels and ROP in premature infants. Dots show measured serum S1P levels and lines (with 95% CI) represent estimates from mixed model for repeated measures adjusted for GA at birth and weight standard deviation score. Graph area highlighted in yellow represents time points where curves differ significantly after adjustment for multiplicity. n = 28 for no/moderate ROP, n = 19 for severe ROP. Graph was adapted from [42].
Figure 2. Serum S1P levels and ROP in premature infants. Dots show measured serum S1P levels and lines (with 95% CI) represent estimates from mixed model for repeated measures adjusted for GA at birth and weight standard deviation score. Graph area highlighted in yellow represents time points where curves differ significantly after adjustment for multiplicity. n = 28 for no/moderate ROP, n = 19 for severe ROP. Graph was adapted from [42].
Life 11 01119 g002
Figure 3. Mouse models of ROP. (A) Mouse model of oxygen-induced retinopathy (OIR). Mouse neonates at postnatal day (P) 7 with their nursing dam are exposed to 75% oxygen for five days. At P12, mice are returned to room air. At P17, neovascularization (NV) reaches the maximum. Retinal vasculature is visualized with isolectin (red) staining and NV area is highlighted in white. Scale bar, 1 mm. (B) Mouse model of hyperglycemia-associated retinopathy (HAR). Hyperglycemia is induced in mouse neonates with streptozotocin (STZ, 50 mg/kg) intraperitoneally (i.p.) from P1 to P9. At P10, retinal vessels in the deep vascular plexus are visualized with isolectin (red) staining. Reduced retinal vascular density is observed in HAR. Scale bar, 50 µm.
Figure 3. Mouse models of ROP. (A) Mouse model of oxygen-induced retinopathy (OIR). Mouse neonates at postnatal day (P) 7 with their nursing dam are exposed to 75% oxygen for five days. At P12, mice are returned to room air. At P17, neovascularization (NV) reaches the maximum. Retinal vasculature is visualized with isolectin (red) staining and NV area is highlighted in white. Scale bar, 1 mm. (B) Mouse model of hyperglycemia-associated retinopathy (HAR). Hyperglycemia is induced in mouse neonates with streptozotocin (STZ, 50 mg/kg) intraperitoneally (i.p.) from P1 to P9. At P10, retinal vessels in the deep vascular plexus are visualized with isolectin (red) staining. Reduced retinal vascular density is observed in HAR. Scale bar, 50 µm.
Life 11 01119 g003
Figure 4. Summarized current findings of metabolism in ROP. Lipid and amino acid metabolic disturbance, hyperglycemia as well as unbalanced antioxidant system were found. Hormones including adiponectin and IGF-1 are essential in modulating metabolic responses. The combination of nutrients and the timing of nutritional and hormonal intervention, as well as the corresponding specific cell responses need to be carefully evaluated.
Figure 4. Summarized current findings of metabolism in ROP. Lipid and amino acid metabolic disturbance, hyperglycemia as well as unbalanced antioxidant system were found. Hormones including adiponectin and IGF-1 are essential in modulating metabolic responses. The combination of nutrients and the timing of nutritional and hormonal intervention, as well as the corresponding specific cell responses need to be carefully evaluated.
Life 11 01119 g004
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Tomita, Y.; Usui-Ouchi, A.; Nilsson, A.K.; Yang, J.; Ko, M.; Hellström, A.; Fu, Z. Metabolism in Retinopathy of Prematurity. Life 2021, 11, 1119. https://doi.org/10.3390/life11111119

AMA Style

Tomita Y, Usui-Ouchi A, Nilsson AK, Yang J, Ko M, Hellström A, Fu Z. Metabolism in Retinopathy of Prematurity. Life. 2021; 11(11):1119. https://doi.org/10.3390/life11111119

Chicago/Turabian Style

Tomita, Yohei, Ayumi Usui-Ouchi, Anders K. Nilsson, Jay Yang, Minji Ko, Ann Hellström, and Zhongjie Fu. 2021. "Metabolism in Retinopathy of Prematurity" Life 11, no. 11: 1119. https://doi.org/10.3390/life11111119

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop