Take a look at the Recent articles

Potential molecular biomarkers of oxidative stress in age-related macular degeneration

Rogil Jose de Almeida Torres

Sao Paulo State University (UNESP), Medical School, Botucatu, SP, Brazil

E-mail : bhuvaneswari.bibleraaj@uhsm.nhs.uk

Andrea Luchini

Centro Oftalmologico de Curitiba, Curitiba, PR, Brazil

Rogerio Joao de Almeida Torres

Hospital Angelina Caron, Campina Grande do Sul, PR, Brazil

Ana Lucia dos Anjos Ferreira

Sao Paulo State University (UNESP), Medical School, Botucatu, SP, Brazil

DOI: 10.15761/NFO.1000259

Article
Article Info
Author Info
Figures & Data

Abstract

Molecular biomarkers are of utmost importance for the diagnosis, prognosis, and treatment monitoring of several diseases. In age-related macular degeneration (AMD), the major cause of irreversible blindness in the elderly, the diagnosis, prognosis, and therapeutic assessment are performed by means of imaging exams such as color fundus photography, fundus autofluorescence, optical coherence tomography, fluorescein angiography and indocyanine green. The use of molecular biomarkers to warn physicians and patients about a favorable or unfavorable progression of AMD is not usual. The current review aims to describe the participation of oxidative molecules in triggering AMD and correlate them with the respective serum, plasma, and urine measurements, non-invasive and low-cost methods, which provide a personalized pre-clinical, and consequently a pre-symptomatic, approach for AMD screening, control, and prevention. The data sources used in this review study include Pubmed, MedlinePlus Health Information, and Elsevier Science. Articles cited in the reference list obtained through this search were also reviewed, whenever relevant.

Key words

age-related macular degeneration, biomarker, free radicals, aging, antioxidant/pro-oxidant balance, oxidative stress, macromolecular damage

Introduction

Age-related macular degeneration (AMD) is the main cause of irreversible blindness in the elderly. Factors related to its genesis such as aging, genetic, environmental, dietetic, and cardiovascular, become the prognosis of this disease unfavorable. Biological markers (biomarkers) in AMD are hardly used; rather, imaging exams such as color fundus photography (CFP), fundus autofluorescence (FAF), optical coherence tomography (OCT), fluorescein angiography and ICG are the selected options for its diagnosis, prognosis, and assessment of the therapeutic efficacy. A biomarker is an indicator of normal biological and pathogenic processes, or pharmacologic responses to a therapeutic intervention [1]. Hence, the analysis of oxidative stress biomarkers in AMD may be valuable considering that oxidative stress is the state where there is an imbalance between the antioxidant defense system and the generation of reactive species [of oxygen (ROS) or nitrogen (RNS)], with the involvement of multiple molecules in the progression of the disease.

It is first necessary to point out that oxidizing and antioxidant substances are generated in a scenario of oxide-reduction reactions, where oxidation implies electron gain and reduction, its loss. Many authors have adopted the term redox system imbalance to refer to oxidative stress [2,3], the main AMD triggering factor. The retina is a tissue exposed to oxidative stress due to its high metabolism, large concentrations of polyunsaturated fatty acid content, exposure to visible light (between 400 - 700 nm) and the presence of photosensitive molecules such as rhodopsin and lipofuscin [4]. The chronic oxidative stress induced by UV-light exposure, along with high ocular oxygen levels, generate ROS and RNS, such as superoxide (O2¯ •), hydrogen peroxide (H2O2), singlet oxygen (1O2), and peroxynitrite (ONOO-) and trigger permanent peroxidation of polyunsaturated lipids in the membrane system of ocular photoreceptor membranes leading to lipid peroxidation–derived protein modifications, which induce damage to retinal pigment epithelial (RPE) cells [5-8]. Nevertheless, the body synthesizes antioxidant enzymes by means of complex mechanisms such as the nuclear factor e2-related factor 2 (Nrf2) activated. The Nrf-2 activation induced by the reactive oxygen species promotes an increase in the expression of antioxidant enzymes, responsible for maintaining the retinal homeostasis and consequent visual function [9]. With aging, the increase of ROS/RNS and the decrease in the expression of antioxidant enzymes are observed, making the macula more susceptible to AMD [10]. The redox imbalance can potentially increase the expression of toxic molecules such as malondialdehyde (MDA) and advanced glycation end products (AGEs), which induce the accumulation of lipofuscin inside the RPE cells [11-13]. The accumulation of intracellular lipofuscin causes dysfunction of RPE cells [14-15], leading to an anomalous deposition of lipids and cholesterol esters in the Bruch’s membrane (BM) [16-17], forming druses and other extracellular deposits [18-19], clinically characterizing the onset of AMD. Lack of cure has encouraged the discovery of new diagnostic, prognostic and therapeutic strategies that may help to prevent or slow the onset and/or progression of AMD. From the onset of AMD (characterized by redox imbalance) to the advanced stage of the disease (characterized by the increase of cytokines, enzymes, and growth factors) several molecules come into play. Hence, this review aims to describe the role of oxidative molecules responsible for triggering AMD and correlate them with their serum, plasma, and urine levels. It is meant to serve as a practical guide for ophthalmologists in their daily practices, with an attempt to assess the possible AMD biomarkers by means of non-invasive, safe, and inexpensive exams, and limited to nongenetic compounds.

Methods of literature search

The data sources used in this review study include Pubmed, MedlinePlus Health Information, and Elsevier Science. We used the following keywords and their synonyms in various combinations: age-related macular degeneration; biomarker; free radicals; aging; antioxidant/pro-oxidant balance; oxidative stress; macromolecular damage. When a specific molecule was identified, the specific risk factor was also used as a keyword in a second PubMed search to identify additional publications. Articles cited in the reference list obtained through this search were also reviewed, whenever relevant.

Oxidative stress of molecules

The reactive species, generated by enzymatic and non-enzymatic systems, have been associated with a large number of physiologic and pathological processes (20). Although they are essential for a variety of cellular defense mechanisms [21,22], ROS (as well as RNS) can cause oxidative damage in biomolecules [lipids, proteins, carbohydrates, and deoxyribonucleic acid (DNA)] when present in larger quantity than their system-mediated neutralization antioxidant defense. Under these conditions, ROS/RNS become unstable, very reactive, and short-lived, preventing the detection of these reactive species in biological samples, such as tissues, fluids, and complex biological systems [23,24]. The suggested option to measure ROS is the use of a species with stable, detectable, and measurable characteristics that can reveal the pathways that may trigger some pathologies [25]. The cellular products resultant from the oxidative damage into lipids, proteins, carbohydrates, and DNA are used as biomarkers of oxidative stress, providing indirect detection of the ROS activity in several diseases [23], including AMD [4, 26-27].

Lipid peroxidation biomarkers

The lipids are organic compounds with essential functions for the human body which include energy storage, membrane integration and structure (phospholipid bilayer), in the biosynthesis process of important substances such as prostaglandins, and as an enzyme cofactor [28,29]. The lipids represent the main target for the ROS [30], especially the glycolipids, phospholipids, and cholesterol [31]. The lipid peroxidation resultant from the oxidation of polyunsaturated fatty acids (PUFAs) produces lipid hydroperoxides and several aldehydes [29,32], with a negative impact in the body, triggering several cellular damages which may lead to multiple diseases [31,33]. The retina is one of the body tissues with the highest concentration of PUFAs, essential for the maintenance of the physiological retinal function and development [34,35]. The photoreceptor outer segments (POS), with high PUFAs (up to 70%), are easily peroxidized due to the presence of high concentrations of oxygen and UV irradiation. These POS are continuously phagocyted and degraded by the RPE cells. Under oxidative stress and inflammation, these lipids become dysfunctional and generate a mix of lipid oxidation products (LOP) which will further form malondialdehyde (MDA) or 4-hydroxy-trans-2-nonenal (HNE), constituting a material source resistant to lysosomal degradation named lipofuscin [8,14,36-39]. The accumulation of non-degradable material in the lysosomes causes intracellular accumulation of phagosomes and autophagosomes, with subsequent reduced phagocytosis and autophagic sequestration. This sequence of events has been demonstrated in an experiment in which the lysosomal dysfunction induced by Bis-retinoid N-retinyl-N-retinylidene ethanolamine (A2E), the major component of lipofuscin, significantly reduced the capacity of cultivated RPE cells to phagocyte the POS and sequester cytoplasmic material [40]. Extracellular deposits of non-degradable material, resultant from the reduced phagocytic capacity, may trigger additional damage, such as apoptosis and formation of drusen [41-44]. Many studies demonstrate the presence of esterified and unesterified cholesterol, triglyceride, and lipoproteins in drusen [19,45-47], which induce the inflammatory response [48]. In this regard, the accumulation of apolipoprotein B100 (apoB100) lipoprotein particles in the BM and RPE cell apoptosis are considered critical events for the triggering of AMD [4,6,19,49-50]. Regarding the systemic biomarkers, several studies have shown that AMD patients have higher lipid peroxidation levels in their plasma [51-52].

Low-density lipoproteins

Oxidized Low-Density Lipoproteins (OxLDL) originate from low density lipoprotein (LDL) in pro-oxidant tissue environment in which lipids and proteins become oxidatively modified causing diseases such as atherosclerosis [53,54]. The retina, particularly the photoreceptor layer rich in unsaturated lipoproteins, is readily susceptible to oxidation by ROS. Lipid oxidation increases with the age and presence of oxidized low-density lipoproteins are particularly relevant to retinal para-inflammation [55]. Studies suggest that OxLDL may also be involved in pathobiological alterations of RPE cells [56-59]. It is important to point out that RPE cells express functional receptors for both LDL (LDL-R) and CD36 [60,61], which take up LDL and OxLDL in large quantities, both in vitro and in vivo [44]. The accumulation of oxidized lipid-protein complexes in the RPE prevents phagosome maturation by blocking phosphatidylinositol 3-kinase (PI3K) recruitment to the phagosomal membrane, leading to delayed processing of internalized POS [62]. Consequently, the oxysterols, products of autoxidation or cholesterol enzymatic oxidation, in OxLDL become cytotoxic to RPE cells [44,62]. Both the alterations of the sensory retina as well as those of RPE, induced by OxLDL, contribute to AMD pathogenesis [44,58]. From these alterations, formation of subretinal deposits may be formed [63] since accumulation of oxidized lipids and lipoproteins has been found in BM and is thought to be an early event in the development of AMD [59,64]. Another study revealed that besides OxLDL, native LDL can potentially up-regulate the expression of vascular endothelial growth factor (VEGF), a major angiogenic and inflammatory factor in RPE cells (56). Nevertheless, it has been demonstrated that the OxLDL treatment decreased human retinal pigment epithelial (ARPE-19) cells viability in a dose-dependent manner, whereas native LDL had no effect. Incubation of ARPE-19 cells with 10 mg/mL OxLDL induced marked apoptosis, compared with untreated control cells. OxLDL also increased VEGF expression and decreased Pigment Epithelium-Derived Factor (PEDF) expression, whereas native LDL had no significant effect. The VEGF-to-PEDF ratio was elevated after OxLDL treatment, suggesting that OxLDL treatment induced cellular changes in ARPE-19 cells that seemed to reflect pathogenic events in neovascular AMD [65]. It was also demonstrated that OxLDL induces apoptosis of human retinal pigment epithelium through activation of ERK-Bax/Bcl-2 signaling pathways [66], and promotes NLRP3 inflammasome activation [67], playing a role in the pathogenesis of AMD, also through this pathway. A study corroborated the reported findings that oxidized LDL increases the expression of inflammatory factors as well as the production of ROS, which could be regulated by the activation of the canonical pathway [68]. It was suggested that OxLDL could promote senescence of RPE cells, inducing outer blood-retinal barrier dysfunction as an early pathogenesis of AMD [69]. Additionally, free-radical oxidation of lipids in LDL produces protein-bound (carboxyalkyl) pyrroles [70], which bind to cellular proteins forming advanced lipoxidation end products (ALEs) and induce an inflammatory response, contributing to the progression of AMD [71,72]. Corroborating this theory, studies report that LDL and OxLDL treatments increase the expression of fibronectin, laminin alpha 1, collagen type IV alpha 2, and transforming growth factor beta-2 (TGF-b2) in RPE cells in vitro, similar to the pathogenic events identified in AMD [58]. It has been shown that OxLDLs are immunohistochemically detected in surgically excised choroidal neovascular (CNV) membranes, revealing those macrophages and RPE in the CNV membranes express cell surface scavenger receptors for oxidized lipoproteins. This study suggested that macrophages may accumulate to take up oxidized lipoproteins in AMD [57]. Experimental studies in vivo and in vitro, inducing RPE to a lipid oxidative damage and chronic inflammation by means of exposure to OxLDL, have been performed to assess the antioxidant and anti-inflammatory effects of several substances [73-77].

Another theory correlates the origin of the lipoproteins in BM with plasma LDL particles in the choriocapillaris. Circulating human plasma LDL may enter the RPE through fenestrated junctions in the choriocapillaris endothelium, crossing BM to reach the RPE [44,78]. Considering this hypothesis, the serum results of OxLDL become relevant. The increased LDL in serum was related to the increased risk of AMD whereas increased high-density lipoprotein (HDL) was related to decreased risk [79,80]. A study with 45 patients affected by exudative age-related macular degeneration (E-AMD), compared with 45 sex- and age-matched healthy controls, reported a positive and significant correlation between plasma OxLDL concentration and homocysteine in patients with E-AMD [81]. Conversely, an observational prospective cohort study assessed the relationship between serum OxLDL cholesterol and the incidence of AMD over a 25-year period. A cohort of 2468 participants was selected for OxLDL measurements, revealing that OxLDL was associated with neither a worsening condition along the AMD severity scale, nor with the incidence of late AMD [82]. Another study with fewer participants also did not correlate the serum levels of OxLDL with AMD [83].

Malondialdehyde

Malondialdehyde (MDA), a highly reactive three-carbon dialdehyde produced as a byproduct of PUFA peroxidation, is catalyzed by free radicals [11,84] and has been detected in high levels in several diseases [85]. Its molecule is small, with polar characteristics and highly soluble in water [86]. MDA is one of the most studied biomarkers of oxidative stress, used in several experimental human, animal and even plant models. This biomarker presents mutagenic and cytotoxic effects in the body, important in the assessment of lipid peroxidation [87].

A study reported that the aerobic illumination of human lipofuscin isolated from aged donors leads to formation of hydroperoxides and MDA [88]. Another study confirmed the presence of MDA in RPE [26]. In fact, some studies have also shown that MDA induces RPE dysfunction and VEGF expression in RPE [89-93]. Similarly, MDA was found in the drusen [26,93]. From the epidemiologic perspective, clinical trials have shown the risk of high dietary intake of linoleic acid (LA) for AMD [94,95]. Experimentally, higher dietary intake of LA promoted progression of the CNV membrane in mice with increased MDA levels [92]. Many studies have suggested that MDA levels are higher in the blood of AMD patients than in that of healthy subjects [96-100]. A systematic review and meta-analysis, involving twelve case-control studies with a total of 634 AMD patients and 656 controls without AMD, concluded that there is some evidence of higher levels of MDA in AMD patients compared with healthy controls; however, this result should be interpreted with caution because of extreme between-study heterogeneity and the possible effect of publication bias [101]. It is important to point out that age is an important confounder when assessing the potential role of blood MDA in oxidative stress. Indeed, advanced age is associated with increasing MDA. Although most studies that report MDA as a risk factor for AMD claim that patients and controls were age-matched, the precision of matching and the statistical test of the outcome of matching are seldom shown [101].

Isoprostanes

A great advancement in the assessment of lipid peroxidation was made with the discovery of F2 isoprostanes (F2-IPs). They comprise a class of compounds structurally similar to prostaglandin-F2 (PGF2), which are produced by the peroxidation of the arachidonic acid (AA) and of other PUFAs, such as the linolenic, the eicosapentaenoic (EPA) and the docosahexaenoic acid (DHA) [102-104]. F2-IPs are considered the most sensitive and most stable marker of lipid peroxidation and oxidative stress [105-106]. High levels of F2-IPs in human fluids and tissues were found in atherosclerosis, chronic inflammation, diabetes, pulmonary diseases, Alzheimer’s, and other neurodegenerative disorders [107-109].

At first, it is important to emphasize that the retinal photoreceptors contain the highest concentrations of AA and DHA in relation to any other known membrane [110]. They are highly peroxidizable [111]. Additionally, the formation of F2-IPs is regulated by the oxygen tension, so that the production of isofurans (IsoF), oxidation products generated from the nonenzymatic oxidation of AA (formed instead of F2-IPs in settings of increased oxygen tension), is favored as the oxygen concentration increases [105], which makes the retina an appropriate tissue for the formation of F2-IPs [4]. High levels of this biomarker were related to risk factors for diseases associated with AMD [112], such as smoking [113], and multiple systemic diseases [106,114-115]. It is known that the lipid peroxidation has been widely associated with inflammation [116], one of the main pillars of AMD genesis. While one of the studies did not report any association between early AMD and serum levels of 8-Iso-Prostaglandin F2 [117], another one suggested a significant association of mean plasma levels of IsoFs with AMD [118]. Nevertheless, a more relevant association was observed in a study that included 238 adults with AMD and 390 age- and sex-matched controls without AMD, reporting that higher levels of urinary F2-IPs were associated with AMD [119] and have been used as biomarkers of AMD in several studies [120-123].

4-Hydroxynonenal

4-Hydroxy-2,3-trans-nonenal (4-hydroxynonenal, HNE) or 4-hydroxyhexenal (4-HNE) is highly reactive α,β-unsaturated hydroxyalkenal [123]. 4-HNE is one of the main end products of the lipid peroxidation of the AA and has been widely accepted as an inducer and mediator of the oxidative stress, being involved in the pathogenesis of several degenerative diseases such as Alzheimer’s, atherosclerosis, cataract, and cancer [11,124-125]. In the retina, a large number of proteins contain HNE adducts, suggesting that HNE is the main oxidant of the retina. It has been shown that 4-HNE binds to cellular proteins forming ALEs [71-72], which may induce an inflammatory response, believed to play a role in the pathogenesis of AMD. A significant number of proteins modified by the HNE have been identified in damages induced by light [126-127], age [128-129] and in a model of retinitis pigmentosa in pigs [130]. As an important mediator of oxidative stress, 4-HNE induces lysosome dysregulation, lipofuscin generation and apoptosis of RPE cells [131-133]. It is known that proteins modified by the HNE in the photoreceptors exert their toxic effects on RPE for also inducing angiogenic cytokines [134], such as interleukin-6 (IL-6), interleukin-1-β (IL-1β) and tumor necrosis factor alpha (TNF-α) [135]. In a study, high levels of 4-HNE generated superoxide anion which leads to the apoptotic degeneration of RPE through the activation of NADPH oxidase 4 (NOX4) [136]. However, despite the confirmed participation of HNE in the AMD pathogenesis [39], it was observed that the proteins modified this biomarker and did not accumulate in the neurosensory retina during the progression of the disease, suggesting that the pathways involved in the HNE detoxification or in the removal of the proteins modified by HNE are adequate to prevent its accumulation. Hence, HNE has been considered a little sensitive retinal biomarker for AMD [137]. Conversely, due to the toxicity in ARPE-19) cells, HNE has been used in experimental studies that search for information about antioxidant and anti-proliferative properties of several substances such as the quercetin [138], N-acetyl-cysteine [139], and edaravone [140]. HNE has also been used in experiments that seek a better understanding of AMD mechanism [26,90,139,141-142]. For example, a study investigated the molecular and cellular effects of cigarette smoke exposure on human RPE cells. Exposure of ARPE-19 or primary human RPE cells to cigarette smoke extract (CSE) or hydroquinone (HQ) caused oxidative damage and apoptosis. Evidence of oxidative damage also included increased lipid peroxidation (4-HNE) and mitochondrial superoxide production, as well as a decrease in intracellular glutathione (GSH) [143]. Therefore, HNE plasma levels have been used to assess the effect of oxidant and antioxidant substances in AMD epidemiologic studies [120].

Carboxyethylpyrrole

Carboxyethylpyrrole (CEP) protein adducts belong to a family of 2-(ω-carboxyalkyl) pyrrole adducts generated from the oxidation of PUFA [70], exclusively from DHA compounds [144]. DHA is present in small quantities in most tissues, but it is a major structural lipid of the retina presenting particularly high levels in this neural tissue [145-146]. Although rare in most human tissues, o DHA is the most oxidizable fatty acid in humans and is present in ~80 mol % of the polyunsaturated lipids in POS [110]. DHA may be involved in the permeability, thickness, fluidity, and other properties of the membrane of photoreceptors (146), and its insufficiency is linked to changes in the function of the retina [146-147]. Immunocytochemical analysis showed that CEP was present in photoreceptor rod outer segments and RPE cells in both mouse and human retinas [13,144]. Additionally, it has been reported that CEP protein adducts found in AMD are immunogenic, inducing autoantibody production and inflammation in the retina [13,144,148]. The immunization of mice with CEP-modified mouse serum albumin (CEP-MSA) induced antibodies against CEP and led to inflammatory responses such as the deposit of complement component-3 in BM and macrophage infiltration [149]. CEP is involved in the pathogenesis of both angiogenesis in the retina (wet AMD) [150] and geographic retinal atrophy (dry AMD) [149]. Regarding wet AMD, it has been shown that CEP adducts stimulate angiogenesis in the chick embryo chorioallantoic membrane (CAM) and corneal micropocket assays and exacerbate CNV in a mouse model. These results, coupled with the elevated levels of CEP adducts in AMD tissues, strongly suggest that CEP may play a role in the development of the wet (exudative) form of AMD. Overall, these results suggest that CEP-induced angiogenesis utilizes VEGF-independent pathways and that anti-CEP therapeutic modalities might be of value in limiting CNV in AMD [150]. Confirming the inflammatory and angiogenic condition of CEP, studies revealed that it induces PYD domains-containing protein 3 (NLRP3) inflammasome priming, via Toll-like receptor 2 (TLR2) in macrophages, promoting IL-1β release [151]. Similarly, other studies reported that CEP-adducts cooperate in a highly specific manner to amplify low-grade inflammation mediated by TLR2/TLR1-activating pathogen associated molecular patterns (PAMPs) and increases M1 (pro-inflammatory macrophages that produce TNF-α, IL-12 polarization [152]. Hence, the presence of CEP protein adducts in the outer retina is considered an early marker of high risk for AMD development [13,149,153]. An immunocytochemical analysis of BM/choroid tissue demonstrated higher CEP and carboxymethyllysine immunoreactivity in BM/RPE/choroid tissues of donors with AMD than of donors with normal eyes [13,144]. A study confirmed these findings revealing that CEP protein adducts are more abundant in the tissue of donors with AMD than in those with normal eyes [154]. Consequently, DHA oxidation products may be used as biomarkers to identify susceptibility to AMD [144]. Specifically, CEP markers increase the predictive accuracy of AMD [155], as well as its prognosis [156].

A study with 19 donors revealed that the mean level of anti-CEP immunoreactivity in human plasma with AMD was 1.5-fold higher than in age-paired controls. The serum levels of AMD patients presented a mean of anti-CEP antibody 2.3-fold higher than those of controls. Out of the patients (n = 13) who presented higher levels of antigen and autoantibodies than the control patients without AMD, 92% had AMD [144]. Another study that used a significantly larger sample size (916 AMD and 488 control donors), determined a 1.6-fold increase in mean plasma anti-CEP immunoreactivity and a 1.3-fold increase in mean plasma anti-CEP autoantibody titer in AMD donors with respect to control donors [155]. The plasma analysis of the 58 AMD and 32 control donors also revealed that CEP increased ~86% in the AMD cohort and that in combination with N-ε-carboxymethyllysine (CML) and pentosidine increased the potential use of the biomarker to assess risks of and susceptibility to AMD [156]. Interestingly, proteomic CEP markers alone can distinguish between AMD and normal donors with approximately 76% accuracy and when analyzed together with genomic markers, the discriminatory accuracy increased to about 80% [157].

Protein oxidation biomarkers protein carbonyl

Protein carbonyl content is an index of the amount of oxidative damage to proteins due to the direct attack of free radicals or modification of proteins by the carbohydrate oxidation products or PUFA. The carbonylated proteins are composed of a carbon atom double-bonded to an oxygen atom, commonly found in determined functional groups, named aldehydes and ketones [158-160]. The binding of carbonyl groups in residues of protein amino acids is a major hallmark for oxidative modification [161]. Carbonyl protein, considered the most widely studied marker of protein oxidation, has been used in several publications as a parameter of oxidative stress [162-164], due to the chemical stability of the carbonyl group, its oxidation-irreversible products and irreparable changes induced by the group, although the cells display native enzymatic systems that eliminate the changed proteins and maintain homeostasis and cell survival [165]. When these enzymatic systems fail, the carbonylated proteins accumulate in the cells, interrupting their functions [166]. Concentration increase, and consequent accumulation in the body, is associated to smoking and aging, as well as to the development of several pathologies such as Alzheimer’s, Parkinson, Huntington, and respiratory syndromes [160,167-168]. The plasma biological samples are the most used analytical methods for the detection of the carbonyl groups [158]. Values of protein carbonyl groups in plasma significantly higher than in CG have been observed in patients with wet AMD [169]. Other studies have shown similar results, reporting significant increase of protein carbonyl groups in AMD when compared to control group [170-171]. It has also been demonstrated that aging significantly affects the antioxidant status and oxidative damage in AMD patients when compared to controls, and an increase of protein carbonyl groups was identified in both early- and late-AMD patients [99]. Conversely, a study that sought to examine the relationship between inflammation, oxidative stress, and endothelial dysfunction markers with a 20-year cumulative incidence of early AMD did not find a relationship between total carbonyl content (TCC) and early AMD [117].

Biomarkers of carbohydrate oxidation

AGEs comprise a heterogeneous group of compounds derived from a non-enzymatic reaction named glycation, Maillard reaction, a spontaneous post-translational modification in which a carbonyl group of reducing sugars is covalently bound to proteins, lipids and nucleic acids [171-172] The α-dicarbonyls can react with lysine and arginine functional groups on proteins, leading to the formation of stable AGE compounds, such as CML, a lysine modification, and pentosidine, a fluorescent lysine-arginine cross-link, generated by oxidation of carbohydrate [173]. Their physiological serum level is in the range of 2–10 Lg/mL and increases with age [174]. AGEs, generated from oxidized carbohydrate products, promote inflammation in many general age-related diseases such as Alzheimer's disease, atherosclerosis, diabetes, osteoarthritis [175-177], and represent a risk factor for AMD development [178]. CML, a major circulating advanced glycation end- product, was the first AGE to be found in AMD BM and drusen [179]. Further studies reported that CML increases with age in BM [180-181]. Other studies have identified glycoxidation products in subretinal membranes of patients with AMD [178,182], whereas the study that assessed the macula of human donor retinas from normal eyes and eyes with early AMD and advanced AMD with GA demonstrated that RAGE and AGE were elevated on RPE and photoreceptor cells in early and advanced dry AMD [183], especially in RPE overlying drusen-like deposits on BM [184]. Other studies confirm the presence of AGEs also in the drusen, as well as in the BM, RPE, and choroidal extracellular matrix of elderly people [13,185]. AGEs influence the profiles of the ARPE-19 expression and may contribute to the decrease in the degrative capacity of the lysosomal enzyme and increase in the lipofuscin accumulation. AGEs formation in the BM may have important consequences for RPE dysfunction related to aging and may damage the outer retina [186]. AGEs stimulate RPE cells to secrete different anti/proinflammatory factors, which trigger the para-inflammation state in RPE cells, that is, short-term adaptive RPE cell reaction on AGE stimulation [187]; however chronic RPE exposure to AGE favors deregulation of RPE function and leads to photoreceptors and RPE cells degeneration and atrophy [181]. Oxidative protein modifications like CML, elevated in AMD BM, stimulate neovascularization in vivo, suggesting possible roles in CNV [188]. Corroborating the findings of this study, it has been demonstrated that AGEs can stimulate the proliferation of choroid endothelial cells, the expression of matrix metalloproteinase type 2, and growth factors such as VEGF [189]. AGEs undergo endocytosis and are removed by macrophages [190]. The failure in macrophage recruitment may lead to increased RPE exposure with AGEs and damage retinal tissue in the pathological angiogenesis process [187]. Several different receptors for AGEs have been discovered, one of which, RAGE, initiates the intracellular signaling that disrupts cellular function through its recognition and binding of AGEs. RAGE is a member of the immunoglobulin superfamily of receptors [191-192]. A study with 300 early AMD patients, 300 patients with exudative AMD, and 800 healthy controls revealed a significant association between RAGE gene rs1800624 and rs1800625 polymorphisms and AMD risk [193]. AGE-R3, also known as galectin-3, is elevated in AMD BM [194]. In vitro studies have shown that AGE-RAGE binding on macrophages and microglia leads to oxidant stress and activation of the nuclear factor kappa β (NF-κβ) [170,195-196]. Activation of NF-κβ induces an increase in the expression of genes associated with inflammatory cytokines, enzymes, and adhesion molecules, which, in turn, are closely related to AMD [197]. NF-κβ sites control cellular expression of RAGE, linking RAGE to the inflammatory response [198]. A study performed in vitro in ARPE-19 cells revealed that AGEs increased the NF-κβ activation resulting in pro-apoptotic changes in ARPE-19 cells and that, along with OxLDL, homocysteine (Hcy), homocysteine thiolactone (HCTL), they act as pro-oxidant metabolites in RPE that promote AMD through oxidative stress, inflammation, chemotaxis, and neovascularization [72].

The plasma analysis of 58 AMD and 32 control donors demonstrated that CEP was elevated ~86% in the AMD cohort and that, in combination with CML and pentosidine, it increases the potential use of the biomarker in the risk evaluation of and susceptibility to AMD [199]. Another study revealed that the mean level of anti-CEP immunoreactivity in AMD human plasma (n = 19 donors) was 1.5-fold higher than in the age-matched controls (n = 19 donors). Similarly, the serum from AMD patients demonstrated mean titers of anti-CEP autoantibody 2.3-fold higher than controls. It has also been observed that out of individuals (n=13) presenting both antigen and autoantibody levels above the mean for non-AMD controls, 92% had AMD. These results suggest that together CEP immunoreactivity and autoantibody titer may have a diagnostic role in predicting AMD susceptibility (144). Another analysis called skin autofluorescence (AF), a non-invasive marker for AGE in tissues, identified an increase in patients with neovascular AMD, suggesting that AMD is accompanied by enhanced systemic AGE accumulation, which may indicate a role in the pathophysiology of AMD [200]. However, a large population-based cohort involving 4,907 older adults revealed that the higher serum CML concentration had no significant cross-sectional association with prevalent AMD) [201].

Biomarkers of nucleic acid oxidation

Aging is associated with slowing down of the efficiency repair of DNA, and consequent accumulation of DNA damage [202-203], which leads to dysregulation of the cell function, therefore, to aging [206]. This oxidative damage to DNA may induce harmful genetic alterations [205]. It is known that the oxidative damage to DNA is processed by means of the cellular DNA repair systems, whose efficiency may reduce with age [205]. These concepts are valid for all body cells, including the cells of the retina [206]. It has been shown that AMD patients present a significantly higher level of endogenous DNA damage in their peripheral blood lymphocytes (PBL) than the control patients and that oxidative damage to DNA has contributed to this increase. Simultaneously, a higher sensitivity to hydrogen peroxide and to ultraviolet light, as well as a slower kinetics of DNA damage repair induced by these mutagenic were observed in AMD patients when compared to control patients. Additionally, AMD patients presented a larger extent to DNA damage than the mean of the general population [206].

8-Hydroxy-29-Deoxyguanosine

Oxidation of the nucleic acids, by means of the reaction of the hydroxyl radical on the 8-carbon of guanine results in the formation of 8-OHdG. This compound, which presents the molecular formula of C10H13O5, is a highly mutagenic molecule, with stable characteristics. 8-OHdG is one of the most representative products of oxidative damage to DNA [207]. It can cross the cell membranes and may be detected in urine, serum, plasma, saliva, and tissue samples [33,208]. The increase in its levels has been observed in many systemic diseases, as well as in several human organs in age-related processes [209-210]. An age-related increase in oxidative DNA damage product in the intraocular fluid has been reported, providing further support that oxidative DNA damage is associated with aging [211]. Corroborating this hypothesis, a higher level of 8-OHdG in RPE and choroid of adult rats was reported when compared with young control groups [212].

In AMD experimental models, the increase of 8-OHdG in the retinal tissue was induced by the sub-retinal injection of human lipid hydroperoxide (HpODE), a product derived from the sub-macular region of elderly and AMD patients [213]. It may also be induced genetically [214], using H2O2 in cultured ARPE19 cells [215], induced by light [216-217] or the exposure to chronic cigarette smoke [218]. In regard to smoking, one of the main modifiable AMD factors, it has been demonstrated that exposure of ARPE-19 cells to a cigarette smoke concentrate (CSC) not only enhanced ROS levels but also induced 8-OHdG DNA lesions [219].

8- OHdG has been used as a marker of the oxidative stress in experiments that aimed to assess the antioxidant effects of substances such as vitamin D, associated with diphlorethohydroxycarmalol [215], astaxanthin [217] and a nutritional complex [220], as well as to assess the neuroprotective effect of SUN N8075 [221]. With this evidence, it is possible to affirm that 8-OHdG, the major product of oxidative DNA damage [222], is associated with AMD [148,170]. This hypothesis is strengthened by findings of increased levels in the 8- OHdG aqueous humor of AMD patients when compared with those in control groups [223-234]. The same results were observed in the blood serum of AMD patients, who presented increased levels of 8-OHdG when compared with non-AMD cohorts [170]. The serum levels of 8-OHdG measured in early- and late-AMD patients versus healthy controls showed that both early- and late-AMD patients had higher 8-OHdG levels than healthy controls (99).

Total oxidant status (TOS)

Total oxidant status (TOS) and total antioxidant status (TAS) and oxidative stress index (OSI) are oxidative stress parameters used to evaluate the overall oxidative stress status in the body [225]. An imbalance between TOS and TAS has been proposed to be responsible for the increased lipid, protein and DNA damage observed in AMD patients [170]. Serum (or plasma) concentrations of different oxidant species can be measured in laboratories separately, but the measurements are time-consuming, labor-intensive, and costly and require complicated techniques [226]. Since the measurement of different oxidant molecules separately is not practical and their oxidant effects are additive, the TOS of a sample is measured and it is named total peroxide (TP), serum oxidation activity, reactive oxygen metabolites (ROM) or some other synonyms [227]. A study with 156 early-AMD patients, 80 wet-late AMD, 72 dry-late AMD and 207 healthy controls reported that a significantly increased oxidative damage was associated with AMD patients >60 years of age of both genders when compared with controls. Both early- and late-AMD patients presented higher TOS levels than healthy controls [225]. Corroborating these findings, other studies also observed a significant increase in TOS levels in the sera of AMD patients when compared to controls [170,228-229].

Conclusion

At molecular level, the cell products resultant from the oxidative damage in lipids, proteins, carbohydrates, and DNA induce an up-regulated expression of inflammatory cytokines, adhesion molecules, matrix metalloproteinase type 2, VEGF, decreased PEDF expression, NLRP3 inflammasome activation, and NF-κβ activation. These molecular alterations are associated with the increase in the inflammatory response, endothelial activation, macrophage infiltration and consequently stimulate the proliferation of choroid endothelial cells. At structural level, the deposit of complement component-3 in BM and macrophage infiltration, drusen formation, photoreceptors and RPE cells degeneration and atrophy, and the subretinal neovascular membrane are observed. Regarding the use of the oxidation products as biomarkers in serum, plasma, and urine samples, it has been observed that the molecules resultant from the oxidative damage of lipids such as MDA, F2-IPs and CEP, as well as the molecules derived from the DNA oxidative damage, 8-OHdG, and TOS revealed an important association with AMD, and may be considered biomarkers of this disease. It is important to point out that because of the presence of the blood-retinal barrier, biomarkers might be only locally dysregulated inside the eye with no measurable systemic effect [230]. Additionally, aging is a relevant point of misunderstanding, as some molecules besides presenting up-regulation in AMD, are also naturally increased with aging.

Conflicts of interest

None

Criteria for inclusion in the authors/contributors list

Active participation in the research, article drafting and revision.

References

  1. Biomarkers Definitions Working G (2001) Biomarkers and surrogate endpoints: preferred definitions and conceptual framework. Clin Pharmacol Ther 69: 89e95. [Crossref]
  2. Poli G, Schaur RJ, Siems WG, Leonarduzzi G (2008) 4-hydroxynonenal: a membrane lipid oxidation product of medicinal interest. Med Res Rev 28: 569-631. [Crossref]
  3. Santos CX, Tanaka LY, Wosniak J, Laurindo FR (2009) Mechanisms and implications of reactive oxygen species generation during the unfolded protein response: roles of endoplasmic reticulum oxidoreductases, mitochondrial electron transport, and NADPH oxidase. Antioxid Redox Signal 11: 2409-2427. [Crossref]
  4. Beatty S, Koh H, Phil M, Henson D, Boulton M (2000) The role of oxidative stress in the pathogenesis of age-related macular degeneration. Surv Ophthalmol 45: 115-134. [Crossref]
  5. Coleman HR, Chan CC, Ferris FL, Chew EY (2008) Age-related macular degeneration. Lancet. 372: 1835-1845.
  6. Winkler BS, Boulton ME, Gottsch JD, Sternberg P (1999) Oxidative damage and age-related macular degeneration. Mol Vis 5: 32. [Crossref]
  7. Goldberg J, Flowerdew G, Smith E, Brody JA, Tso MO (1988) Factors associated with age-related macular degeneration. An analysis of data from the first National Health and Nutrition Examination Survey. Am J Epidemiol. 128: 700-10.
  8. Berman ER, Rothman MS (1980) Lipofuscin of human retinal pigment epithelium. J Ophthalmol 90: 783-791. [Crossref]
  9. Datta S, Cano M, Ebrahimi K, Wang L, Handa JT (2017) The impact of oxidative stress and inflammation on RPE degeneration in non-neovascular AMD. Prog Retin Eye Res 60: 201-218. [Crossref]
  10. Frank RN, Amin RH, Puklin JE (1999) Antioxidant enzymes in the macular retinal pigment epithelium of eyes with neovascular age-related macular degeneration. Am J Ophthalmol 127: 694-709. [Crossref]
  11. Esterbauer H, Schaur RJ, Zollner H (1991) Chemistry and biochemistry of 4-hydroxynonenal, malondialdehyde, and related aldehydes. Free Radic Biol Med 11: 81-128. [Crossref]
  12. Shimokawa I, Higami Y, Horiuchi S, Iwasaki M, Ikeda T (1998) Advanced glycation end products in adrenal lipofuscin. J Gerontol A Biol Sci Med Sci 53: B49-B51. [Crossref]
  13. Crabb JW, Miyagi M, Gu X, Shadrach K, West KA, et al. (2002) Drusen proteome analysis: an approach to the etiology of age-related macular degeneration. Proc Natl Acad Sci USA 99: 14682-14687. [Crossref]
  14. Shamsi FA, Boulton M (2001) Inhibition of RPE lysosomal and antioxidant activity by the age pigment lipofuscin. Invest Ophthalmol Vis Sci 42: 3041-3046. [Crossref]
  15. Mata NL, Weng J, Travis GH (2000) Biosynthesis of a major lipofuscin fluorophore in mice and humans with ABCR-mediated retinal and macular degeneration. Proc Natl Acad Sci USA 9: 7154-7159. [Crossref]
  16. Ruberti JW, Curcio CA, Millican CL, Menco BP, Huang JD, et al. (2003) Quick-freeze/deep-etch visualization of age-related lipid accumulation in Bruch's membrane. Invest Ophthalmol Vis Sci 44: 1753-1759. [Crossref]
  17. Pauleikhoff D, Harper CA, Marshall J, Bird AC (1990) Aging changes in Bruch’s membrane: a histochemical and morphologic study. Ophthalmology 97: 171-178. [Crossref]
  18. Curcio CA, Millican CL (1999) Basal linear deposit and large drusen are specific for early age-related maculopathy. Arch Ophthalmol 117: 329-339.
  19. Curcio CA, Millican CL, Bailey T, Kruth HS (2001) Accumulation of cholesterol with age in human Bruch's membrane. Invest Ophthalmol Vis Sci 42: 265-274.
  20. Aldini G, Dalle-Donne I, Facino RM, Milzani A, Carini M (2007) Intervention strategies to inhibit protein carbonylation by lipoxidation-derived reactive carbonyls. Med Res Rev 27: 817-868.
  21. Touyz RM, Schiffrin EL (2004) Reactive oxygen species in vascular biology: implications in hypertension. Histochem Cell Biol. 122: 339-352. [Crossref]
  22. Oneschuk D, Younus J (2008) Natural health products and cancer chemotherapy and radiation therapy. Oncol Rev 1: 233-242.
  23. Ogino K, Wang DH (2007) Biomarkers of oxidative/nitrosative stress: an approach to disease prevention. Acta Med Okayama 61: 181-189. [Crossref]
  24. Siwek M, Sowa-Kucma M, Dudek D, Styczen K, Szewczyk B, et al. (2013) Oxidative stress markers in affective disorders. Pharmacol Rep 65: 1558-1571. [Crossref]
  25. Dalle-Donne I, Rossi R, Colombo R, Giustarini D, Milzani A (2006) Biomarkers of oxidative damage in human disease. Clin Chem 52: 601-623. [Crossref]
  26. Schutt F, Bergmann M, Holz FG, Kopitz J (2003) Proteins modified by malondialdehyde, 4-hydroxynonenal, or advanced glycation end products in lipofuscin of human retinal pigment epithelium. Invest Ophthalmol Vis Sci 44: 3663-3668. [Crossref]
  27. Handa JT, Cano M, Wang L, Datta S, Liu T (2017) Lipids, oxidized lipids, oxidation-specific epitopes, and Age-related Macular Degeneration. Biochim Biophys Acta Mol Cell Biol Lipids 1862: 430-440. [Crossref]
  28. Dabrowska M. Zielinska A, Nowak I (2015) Lipid oxidation products as a potential health and analytical problem. Chemik 69: 89-94.
  29. Yin H, Xu L, Porter NA (2011) Free radical lipid peroxidation: mechanisms and analysis. Chem Rev 111: 5944-5972.
  30. Niki E (2009) Lipid peroxidation: physiological levels and dual biological effects. Free Radic Biol Med 47: 469-484. [Crossref]
  31. Ayala A, Muñoz MF, Argüelles S (2014) Lipid peroxidation: production, metabolism, and signaling mechanisms of malondialdehyde and 4-hydroxy-2-nonenal. Oxid Med Cell Longev 2014: 360438. [Crossref]
  32. Uchida K (2015) Aldehyde adducts generated during lipid peroxidation modification of proteins. Free Radic Res 49: 896-904. [Crossref]
  33. Frijhoff J, Winyard PG, Zarkovic N, Davies SS, Stocker R, et al. (2015) Clinical relevance of biomarkers of oxidative stress. Antioxid Redox Signal 23: 1144-1170. [Crossref]
  34. Berdeaux O, Acar N (2011) Very-long-chain polyunsaturated fatty acids in the retina: Analysis and clinical relevance in physiological and pathological conditions. OCL 18: 284-290.
  35. Agbaga MP, Mandal MN, Anderson RE (2010) Retinal very long-chain PUFAs: new insights from studies on ELOVL4 protein. J Lipid Res 51: 1624-1642. [Crossref]
  36. Holz FG, Schutt F, Kopitz J, et al. Inhibition of lysosomal degradative functions in RPE cells by a retinoid component of lipofuscin. Invest Ophthalmol Vis Sci 1999; 40:737-43.
  37. Katz ML. Incomplete proteolysis may contribute to lipofuscin accumulation in the retinal pigment epithelium. Adv Exp Med Biol.1989; 266:109-16.
  38. Kaemmerer E, Schutt F, Krohne TU, Holz FG, Kopitz J (2007) Effects of lipid peroxidation-related protein modifications on RPE lysosomal functions and POS phagocytosis. Invest Ophthalmol Vis Sci 48: 1342-1347. [Crossref]
  39. Kopitz J, Holz FG, Kaemmerer E, Schutt F (2004) Lipids and lipid peroxidation products in the pathogenesis of age-related macular degeneration. Biochimie 86: 825-831.
  40. Bergmann M, Schütt F, Holz FG, Kopitz J (2004) Inhibition of the ATP-driven proton pump in RPE lysosomes by the major lipofuscin fluorophore A2-E may contribute to the pathogenesis of age-related macular degeneration. FASEB J 18: 562-564.
  41. Martinez CM, Ayala S, Coquet A, Lepinay M, Michel O, et al. (1990) Malignant cell autolysis caused by intracytoplasmic liberation of lysosomal enzymes. Cell Biol Int Rep 14: 255-266.
  42. Mullins RF, Russell SR, Anderson DH, Hageman GS (2000) Drusen associated with aging and age-related macular degeneration contain proteins common to extracellular deposits associated with atherosclerosis, elastosis, amyloidosis, and dense deposit disease. FASEB J 14: 835-846. [Crossref]
  43. Holz FG, Wolfensberger TJ, Piguet B, Gross-Jendroska M, Wells JA, et al. (1994) Bilateral macular drusen in age-related macular degeneration. Prognosis and risk factors. Ophthalmology 101: 1522-1528. [Crossref]
  44. Gordiyenko N, Campos M, Lee JW, Fariss RN, Sztein J, et al. (2004) RPE cells internalize low-density lipoprotein (LDL) and oxidized LDL (oxLDL) in large quantities in vitro and in vivo. Invest Ophthalmol Vis Sci 45: 2822-2829. [Crossref]
  45. Huang JD, Curcio CA, Johnson M (2008) Morphometric analysis of lipoprotein-like particle accumulation in aging human macular Bruch's membrane. Invest Ophthalmol Vis Sci 49: 2721-2727. [Crossref]
  46. Li CM, Chung BH, Presley JB, Malek G, Zhang X, et al. (2006) Apolipoprotein localization in isolated drusen and retinal apolipoprotein gene expression. Invest Ophthalmol Vis Sci 47: 3119-3128. [Crossref]
  47. Wang L, Leitner WP, Rivest AJ, Staples MK, Radeke MJ, et al. (2010) Abundant lipid and protein components of drusen. PLoS One 5: e10329
  48. Anderson DH, Mullins RF, Hageman GS, Johnson LV (2002) A role for local inflammation in the formation of drusen in the aging eye. Am J Ophthalmol 134: 411-431.
  49. Dunaief JL, Dentchev T, Ying GS, Milam AH (2002) The role of apoptosis in age-related macular degeneration. Arch Ophthalmol 120: 1435-1442. [Crossref]
  50. Li CM, Chung BH, Presley JB, Malek G, Zhang X, et al. (2005) Lipoprotein-like particles and cholesteryl esters in human Bruch's membrane: initial characterization. Invest Ophthalmol Vis Sci 46: 2576-2586. [Crossref]
  51. Evereklioglu C, Er H, Doganay S, Cekmen M, Turkoz Y, et al. (2003) Nitric oxide and lipid peroxidation are increased and associated with decreased antioxidant enzyme activities in patients with age-related macular degeneration. Doc Ophthalmol 106: 129-136. [Crossref]
  52. Nowak M, Swietochowska E, Wielkoszynski T, Marek B, Karpe J, et al. (2003) Changes in blood antioxidants and several lipid peroxidation products in women with age-related macular degeneration. Eur J Ophthalmol 13: 281-286. [Crossref]
  53. Levitan I, Volkov S, Subbaiah PV (2010) Oxidized LDL: diversity, patterns of recognition, and pathophysiology. Antioxid Redox Signal 13: 39-75. [Crossref]
  54. Steinberg D (1997) Low density lipoprotein oxidation and its pathobiological significance. J Biol Chem 272: 20963-20966.
  55. Xu H, Chen M, Forrester JV (2009) Para-inflammation in the aging retina. Prog Retin Eye Res 28: 348-368. [Crossref]
  56. Lee SJ, Kim JH, Kim JH, Chung MJ, Wen Q, et al. (2007) Human apolipoprotein E2 transgenic mice show lipid accumulation in retinal pigment epithelium and altered expression of VEGF and bFGF in the eyes. J Microbiol Biotechnol 17: 1024-1030. [Crossref]
  57. Kamei M, Yoneda K, Kume N, Suzuki M, Itabe H, et al. (2007) Scavenger receptors for oxidized lipoprotein in age-related macular degeneration. Invest Ophthalmol Vis Sci 48: 1801-1807. [Crossref]
  58. Yu AL, Lorenz RL, Haritoglou C, Kampik A, Welge-Lussen U (2009) Biological effects of native and oxidized low-density lipoproteins in cultured human retinal pigment epithelial cells. Exp Eye Res 88: 495-503. [Crossref]
  59. Yamada Y, Tian J, Yang Y, Cutler RG, Wu T, et al. (2008) Oxidized low density lipoproteins induce a pathologic response by retinal pigmented epithelial cells. J Neurochem 105: 1187-1197. [Crossref]
  60. Hayes KC, Lindsey S, Stephan ZF, Brecker D (1989) Retinal pigment epithelium possesses both LDL and scavenger receptor activity. Invest Ophthalmol Vis Sci. 30: 225-232. [Crossref]
  61. Ryeom SW, Silverstein RL, Scotto A, Sparrow JR (1996) Binding of anionic phospholipids to retinal pigment epithelium may be mediated by the scavenger receptor CD36. J Biol Chem 271: 20536-20539. [Crossref]
  62. Hoppe G, Marmorstein AD, Pennock EA, Hoff HF (2001) Oxidized low density lipoprotein-induced inhibition of processing of photoreceptor outer segments by RPE. Invest Ophthalmol Vis Sci 42: 2714-2720. [Crossref]
  63. Picard E, Houssier M, Bujold K, Sapieha P, Lubell W, et al. (2010) CD36 plays an important role in the clearance of oxLDL and associated age-dependent sub-retinal deposits. Aging (Albany NY) 2: 981-989. [Crossref]
  64. Spaide RF, Ho-Spaide WC, Browne RW, Armstrong D (1999) Characterization of peroxidized lipids in Bruch’s membrane. Retina 19: 141-147.
  65. Yin L, Wu X, Gong Y, Shi Y, Qiu Y, et al. (2011) OX-LDL up-regulates the vascular endothelial growth factor-to-pigment epithelium-derived factor ratio in human retinal pigment epithelial cells. Curr Eye Res 36: 379-385. [Crossref]
  66. Yating Q, Yuan Y, Wei Z, Qing G, Xingwei W, et al. (2015) Oxidized LDL induces apoptosis of human retinal pigment epithelium through activation of ERK-Bax/Bcl-2 signaling pathways. Curr Eye Res 40: 415-422. [Crossref]
  67. Gnanaguru G, Choi AR, Amarnani D, D'Amore PA (2016) Oxidized lipoprotein uptake through the CD36 receptor activates the NLRP3 inflammasome in human retinal pigment epithelial cells. Invest Ophthalmol Vis Sci 57: 4704-4712. [Crossref]
  68. Zhou T, Hu Y, Chen Y, Zhou KK, Zhang B, et al. (2010) The pathogenic role of the canonical Wnt pathway in age-related macular degeneration. Invest Ophthalmol Vis Sci 51: 4371-4379. [Crossref]
  69. Kim JH, Lee SJ, Kim KW, Yu YS, Kim JH (2012) Oxidized low density lipoprotein-induced senescence of retinal pigment epithelial cells is followed by outer blood-retinal barrier dysfunction. Int J Biochem Cell Biol 44: 808-814. [Crossref]
  70. Kaur K, Salomon RG, O'Neil J, Hoff HF (1997) (Carboxyalkyl)pyrroles in human plasma and oxidized low-density lipoproteins. Chem Res Toxicol 10: 1387-1396. [Crossref]
  71. Pamplona R (2011) Advanced lipoxidation end-products. Chem Biol Interact 192: 14-20.
  72. Thorpe SR, Baynes JW (2003) Maillard reaction products in tissue proteins: new products and new perspectives. Amino Acids 25: 275-281. [Crossref]
  73. Mao K, Shu W, Qiu Q, Gu Q, Wu X. Salvianolic acid A protects retinal pigment epithelium from OX-LDL-induced inflammation in an age-related macular degeneration model. Discov Med 23: 129-147. [Crossref]
  74. AnandBabu K, Sen P, Angayarkanni N (2019) Oxidized LDL, homocysteine, homocysteine thiolactone and advanced glycation end products act as pro-oxidant metabolites inducing cytokine release, macrophage infiltration and pro-angiogenic effect in ARPE-19 cells. PLoS One 14: e0216899. [Crossref]
  75. Mao K, Shu W, Liu L, Gu Q, Qiu Q, et al. (2017) Salvianolic acid a inhibits OX-LDL effects on exacerbating choroidal neovascularization via downregulating CYLD. Oxid Med Cell Longev 2017: 6210694. [Crossref]
  76. Biswas L, Zeng Z, Graham A, Shu X (2020) Gypenosides mediate cholesterol efflux and suppress oxidized LDL induced inflammation in retinal pigment epithelium cells. Exp Eye Res 191: 107931.
  77. Tian B, Al-Moujahed A, Bouzika P, Hu Y, Notomi S, et al. (2017) Atorvastatin promotes phagocytosis and attenuates pro-inflammatory response in human retinal pigment epithelial cells. Sci Rep 7: 2329. [Crossref]
  78. Yin L, Shi Y, Liu X, Zhang H, Gong Y, et al. (2012) A rat model for studying the biological effects of circulating LDL in the choriocapillaris-BrM-RPE complex. Am J Pathol 180: 541-549.
  79. Tan JS, Mitchell P, Smith W, Wang JJ (2007) Cardiovascular risk factors and the long-term incidence of age-related macular degeneration: the Blue Mountains Eye Study. Ophthalmology 114: 1143-1150. [Crossref]
  80. Chakravarthy U, Wong TY, Fletcher A, Piault E, Evans C, et al. (2010) Clinical risk factors for age-related macular degeneration: a systematic review and meta- analysis. BMC Ophthalmol 10: 31. [Crossref]
  81. Javadzadeh A, Ghorbanihaghjo A, Bahreini E, Rashtchizadeh N, Argani H, et al. (2010) Plasma oxidized LDL and thiol-containing molecules in patients with exudative age-related macular degeneration. Mol Vis 16: 2578-2584. [Crossref]
  82. Klein R, Lee KE, Tsai MY, Cruickshanks KJ, Gangnon RE, et al. (2019) Oxidized low-density lipoprotein and the incidence of age-related macular degeneration. Ophthalmology 126: 752-758. [Crossref]
  83. Simonelli F, Zarrilli F, Mazzeo S, Verde V, Romano N, et al. (2002) Serum oxidative and antioxidant parameters in a group of Italian patients with age-related maculopathy. Clin Chim Acta 320: 111-115. [Crossref]
  84. Njie-Mbye YF, Kulkarni-Chitnis M, Opere CA, Barrett A, Ohia SE (2013) Lipid peroxidation: pathophysiological and pharmacological implications in the eye. Front Physiol 4: 366. [Crossref]
  85. Amara A, Constans J, Chaugier C, Sebban A, Dobourg L, et al. (1995) Autoantibodies to malondialdehyde-modified epitope in connective tissue diseases and vasculitides. Clin Exp Immunol 101: 233-238. [Crossref]
  86. Mateos R, Lecumberri E, Ramos S, Goya L, Bravo L (2005) Determination of malondialdehyde (MDA) by high-performance liquid chromatography in serum and liver as a biomarker for oxidative stress. Application to a rat model for hypercholesterolemia and evaluation of the effect of diets rich in phenolic antioxidants from fruits. J Chromatogr B Analyt Technol Biomed Life Sci 827: 76-82. [Crossref]
  87. Barbosa KBF, Costa NMB, Alfenas RCG, Paula SO, Minin VPR, et al. (2008) Estresse oxidativo: avaliação de marcadores. Nutrire Rev Soc Bras Aliment Nutr 33: 111-128.
  88. Rozanowska M, Jarvis-Evans J, Korytowski W, Boulton ME, Burke JM, et al. (1995) Blue light-induced reactivity of retinal age pigment: in vitro generation of oxygen-reactive species. J Biol Chem 270: 18825-18830. [Crossref]
  89. Bergmann M, Holz F, Kopitz J (2011) Lysosomal stress and lipid peroxidation products induce VEGF-121 and VEGF-165 expression in ARPE-19 cells. Graefes Arch Clin Exp Ophthalmol 249: 1477-1483. [Crossref]
  90. Krohne TU, Stratmann NK, Kopitz J, Holz FG (2010) Effects of lipid peroxidation products on lipofuscinogenesis and autophagy in human retinal pigment epithelial cells. Exp Eye Res 90: 465-471.
  91. Krohne TU, Kaemmerer E, Holz FG, Kopitz J (2010) Lipid peroxidation products reduce lysosomal protease activities in human retinal pigment epithelial cells via two different mechanisms of action. Exp Eye Res 90: 261-266.
  92. Ye F, Kaneko H, Hayashi Y, Takayama K, Hwang SJ, et al. (2016) Malondialdehyde induces autophagy dysfunction and VEGF secretion in the retinal pigment epithelium in age-related macular degeneration. Free Radic Biol Med 94: 121-134. [Crossref]
  93. Weismann D, Hartvigsen K, Lauer N, Bennett KL, Scholl HP, et al. (2011) Complement factor H binds malondialdehyde epitopes and protects from oxidative stress. Nature 478: 76-81. [Crossref]
  94. Tan JS, Wang JJ, Flood V, Mitchell P (2009) Dietary fatty acids and the 10-year incidence of age-related macular degeneration: the Blue Mountains Eye Study. Arch Ophthalmol 127: 656-665. [Crossref]
  95. Seddon JM, Cote J, Rosner B (2003) Progression of age-related macular degeneration: association with dietary fat, transunsaturated fat, nuts, and fish intake. Arch Ophthalmol 121: 1728-1737. [Crossref]
  96. Shen XL, Jia JH, Zhao P, Fan R, Pan XY, et al. (2012) Changes in blood oxidative and antioxidant parameters in a group of Chinese patients with age-related macular degeneration. J Nutr Health Aging 16: 201-204. [Crossref]
  97. Totan Y, Cekic O, Borazan M, Uz E, Sogut S, et al. (2001) Plasma malondialdehyde and nitric oxide levels in age related macular degeneration. Br J Ophthalmol 85: 1426-1428. [Crossref]
  98. Yildirim O, Ates NA, Tamer L, Muslu N, Ercan B, et al. (2004) Changes in antioxidant enzyme activity and malondialdehyde level in patients with age-related macular degeneration. Ophthalmologica 218: 202-206. [Crossref]
  99. Venza I, Visalli M, Cucinotta M, Teti D, Venza M (2012) Association between oxidative stress and macromolecular damage in elderly patients with age-related macular degeneration. Aging Clin Exp Res 24: 21-27. [Crossref]
  100. Zor RK, Ersan S, Küçük E, Yildirim G, Sari I (2020) Serum malondialdehyde, monocyte chemoattractant protein-1, and vitamin C levels in wet type age-related macular degeneration patients. Ther Adv Ophthalmol 12: 2515841420951682.
  101. Pinna A, Boscia F, Paliogiannis P, Carru C, Zinellu A (2020) Malondialdehyde levels in patients with age-related macular degeneration: A Systematic Review and Meta-analysis. Retina 40: 195-203. [Crossref]
  102. Montuschi P, Barnes P, Roberts LJ (2007) Insights into oxidative stress: the isoprostanes. Curr Med Chem 14: 703-717. [Crossref]
  103. Roberts LJ II, Morrow JD (1997) The generation and actions of isoprostanes. Biochim Biophys Acta 1345: 121-135. [Crossref]
  104. Rokach J, Khanapure SP, Hwang SW, Adiyaman M, Lawson JA, et al. (1997) The iso- prostanes: A perspective. Prostaglandins 54: 823-851. [Crossref]
  105. Roberts LJ 2nd, Fessel JP (2004) The biochemistry of the isoprostane, neuroprostane, and isofuran pathways of lipid peroxidation. Chem Phys Lipids 128: 173-186. [Crossref]
  106. Fessel JP, Jackson Roberts L (2005) Isofurans: novel products of lipid peroxidation that define the occurrence of oxidant injury in settings of elevated oxygen tension. Antioxid Redox Signal 7: 202-209. [Crossref]
  107. Bauer J, Ripperger A, Frantz S, Ergün S, Schwedhelm E, et al. (2014) Pathophysiology of isoprostanes in the cardiovascular system: implications of isoprostane-mediated thromboxane A2 receptor activation. Br J Pharmacol 171: 3115-3131. [Crossref]
  108. Galano JM, Lee YY, Oger C, Vigor C, Vercauteren J, et al. (2017) Isoprostanes, neuroprostanes and phytoprostanes: An overview of 25years of research in chemistry and biology. Prog Lipid Res 68: 83-108.
  109. Morrow JD (2005) Quantification of isoprostanes as indices of oxidant stress and the risk of atherosclerosis in humans. Arterioscler Thromb Vasc Biol 25: 279-286. [Crossref]
  110. Fliesler SJ, Anderson RE (1983) Chemistry and metabolism of lipids in the vertebrate retina. Prog Lipid Res 22: 79-131. [Crossref]
  111. Cosgrove JP, Church DF, Pryor WA (1987) The kinetics of the autoxidation of polyunsaturated fatty acids. Lipids 22: 299-304. [Crossref]
  112. Torres RJAT, Maia M, Muccioli C, Winter G, Souza GK, et al. (2009) Modifiable factors of age-related macular degeneration. Arq Bras Oftalmol 72: 406-412.
  113. Morrow JD, Frei B, Longmire AW, Gaziano JM, Lynch SM, et al. (1995) Increase in circulating products of lipid peroxidation (F2-isoprostanes) in smokers. Smoking as a cause of oxidative damage. N Engl J Med 332: 1198-1203. [Crossref]
  114. Morrow JD, Roberts LJ (2002) The isoprostanes: their role as an index of oxidant stress status in human pulmonary disease. Am J Respir Crit Care Med 166: S25-S30. [Crossref]
  115. Davies SS, Roberts LJ 2nd (2011) F2-isoprostanes as an indicator and risk factor for coronary heart disease. Free Radic Biol Med 50: 559-566. [Crossref]
  116. Freigang S (2016) The regulation of inflammation by oxidized phospholipids. Eur J Immunol 46: 1818-1825. [Crossref]
  117. Klein R, Myers CE, Cruickshanks KJ, Gangnon RE, Danforth LG, et al. (2014) Markers of inflammation, oxidative stress, and endothelial dysfunction and the 20-year cumulative incidence of early age-related macular degeneration: the Beaver Dam Eye Study. JAMA Ophthalmol 132: 446-455. [Crossref]
  118. Brantley MA, Jr, Osborn MP, Sanders BJ, Rezaei KA, Lu P, et al. (2012) Plasma biomarkers of oxidative stress and genetic variants in age-related macular degeneration. Am J Ophthalmol 153: 460-467.
  119. Sabanayagam C, Lye WK, Januszewski A, Banu Binte Mohammed Abdul R, Cheung GCM, et al. (2017) Urinary Isoprostane Levels and Age-Related Macular Degeneration. Invest Ophthalmol Vis Sci 58: 2538-2543.
  120. Leung HH, Ng AL, Durand T, Kawasaki R, Oger C, et al. (2019) Increase in omega-6 and decrease in omega-3 polyunsaturated fatty acid oxidation elevates the risk of exudative AMD development in adults with Chinese diet. Free Radic Biol Med 145: 349-356. [Crossref]
  121. Leung HH, Galano JM, Crauste C, Durand T, Lee JC (2020) Combination of lutein and zeaxanthin, and DHA regulated polyunsaturated fatty acid oxidation in H2O2-Stressed retinal cells. Neurochem Res 45: 1007-1019. [Crossref]
  122. Brantley MA Jr, Osborn MP, Sanders BJ, Rezaei KA, Lu P, et al. (2012) The short-term effects of antioxidant and zinc supplements on oxidative stress biomarker levels in plasma: a pilot investigation. Am J Ophthalmol 153: 1104-1109. [Crossref]
  123. Schauenstein E, Esterbauer H, Jaag G, Taufer M (1964) The effect of aldehydes on normal and malignant cells. 1st report: hydroxy-octenal, a new fat-aldehyde. Monatshefte Fur Chemie 95: 180-183.
  124. Long EK, Picklo MJ (2010) Trans-4-hydroxy-2-hexenal, a product of n-3 fatty acid peroxidation: make some room HNE. J Free Radic Biol Med 49: 1-8. [Crossref]
  125. Uchida K (2003) 4-Hydroxy-2-nonenal: a product and mediator of oxidative stress. Prog Lipid Res 42: 318-343. [Crossref]
  126. Tanito M, ElliottMH, KotakeY, AndersonRE (2005) Protein modifications by 4-hydroxynonenal and 4-hydroxyhexenal in light-exposed rat retina. Invest Ophthalmol Vis Sci 46: 3859-3868. [Crossref]
  127. Rex TS, Tsui I, Hahn P, Maguire AM, Duan D, et al. (2004) Adenovirus-mediated delivery of catalase to retinal pigment epithelial cells protects neighboring photoreceptors from photo-oxidative stress. Hum Gene Ther 15: 960-967. [Crossref]
  128. Kapphahn RJ, Giwa BM, Berg KM, Roehrich H, Feng X, et al. (2006) Retinal proteins modified by 4-hydroxynonenal: identification of molecular targets. Exp Eye Res 83: 165-175. [Crossref]
  129. Louie JL, Kapphahn RJ, Ferrington DA (2002) Proteasome function and protein oxidation in the aged retina. Exp Eye Res 75: 271-284. [Crossref]
  130. Shen J, Yang X, Dong A, Petters RM, Peng YW, et al. (2005) Oxidative damage is a potential cause of cone cell death in retinitis pigmentosa. J Cell Physiol 203: 457-464.
  131. Ambati J, Atkinson JP, Gelfand BD (2013) Immunology of age-related macular degeneration. Nat Rev Immunol 13: 438-451. [Crossref]
  132. Bonet-Ponce L, Saez-Atienzar S, da Casa C, Sancho-Pelluz J, Barcia JM, et al. (2016) Rotenone induces the formation of 4-Hydroxynonenal aggresomes. Role of ROS-mediated tubulin hyperacetylation and autophagic flux disruption. Mol Neurobiol 53: 6194-6208. [Crossref]
  133. Wihlmark U, Wrigstad A, Roberg K, Brunk UT, Nilsson SE (1996) Formation of lipofuscin in cultured retinal pigment epithelial cells exposed to pre-oxidized photoreceptor outer segments. APMIS 104: 272-279. [Crossref]
  134. Higgins GT, Wang JH, Dockery P, Cleary PE, Redmond HP (2003) Induction of angiogenic cytokine expression in cultured RPE by ingestion of oxidized photoreceptor outer segments. Invest Ophthalmol Vis Sci 44: 1775-1782. [Crossref]
  135. Yang HJ, Hu R, Sun H, Bo Chen, Li X, et al. (2019) 4-HNE induces proinflammatory cytokines of human retinal pigment epithelial cells by promoting extracellular efflux of HSP70. Exp Eye Res 188: 107792. [Crossref]
  136. Bae D, Gautam J, Jang H, Banskota S, Lee SY, et al. (2018) Protective effects of 6-ureido/thioureido-2,4,5-trimethylpyridin-3-ols against 4-hydroxynonenal-induced cell death in adult retinal pigment epithelial-19 cells. Bioorg Med Chem Lett 28: 107-112. [Crossref]
  137. Ethen CM, Reilly C, Feng X, Olsen TW, Ferrington DA (2007) Age-related macular degeneration and retinal protein modification by 4-hydroxy-2-nonenal. Invest Ophthalmol Vis Sci 48: 3469-3479. [Crossref]
  138. Hytti M, Piippo N, Salminen A, Honkakoski P, Kaarniranta K, et al. (2015) Quercetin alleviates 4-hydroxynonenal-induced cytotoxicity and inflammation in ARPE-19 cells. Exp Eye Res 132: 208-215.
  139. Hara R, Inomata Y, Kawaji T, Sagara N, Inatani M, et al. (2010) Suppression of choroidal neovascularization by N-acetyl-cysteine in mice. Curr Eye Res 35: 1012-1020. [Crossref]
  140. Masuda T, Shimazawa M, Takata S, Nakamura S, Tsuruma K, et al. (2016) Edaravone is a free radical scavenger that protects against laser-induced choroidal neovascularization in mice and common marmosets. Exp Eye Res 146: 196-205. [Crossref]
  141. Zhou J, Cai B, Jang YP, Pachydaki S, Schmidt AM, et al. (2005) Mechanisms for the induction of HNE- MDA- and AGE-adducts, RAGE and VEGF in retinal pigment epithelial cells. Exp Eye Res 80: 567-580. [Crossref]
  142. Ayalasomayajula SP, Kompella UB (2002) Induction of vascular endothelial growth factor by 4-hydroxynonenal and its prevention by glutathione precursors in retinal pigment epithelial cells. Eur J Pharmacol 449: 213-220. [Crossref]
  143. Bertram KM, Baglole CJ, Phipps RP, Libby RT (2009) Molecular regulation of cigarette smoke induced-oxidative stress in human retinal pigment epithelial cells: implications for age-related macular degeneration. Am J Physiol Cell Physiol 297: C1200-C1210. [Crossref]
  144. Gu X, Meer SG, Miyagi M, Rayborn ME, Hollyfield JG, et al. (2003) Carboxyethylpyrrole protein adducts and autoantibodies, biomarkers for age-related macular degeneration. J Biol Chem 278: 42027-42035. [Crossref]
  145. Connor WE, Neuringer M (1988) The effects of n-3 fatty acid deficiency and repletion upon the fatty acid composition and function of the brain and retina. Prog Clin Biol Res 282: 275-294. [Crossref]
  146. SanGiovanni JP, Chew EY (2005) The role of omega-3 long-chain polyunsaturated fatty acids in health and disease of the retina. Prog Retin Eye Res 24: 87-138. [Crossref]
  147. Querques G, Souied EH (2014) The role of omega-3 and micronutrients in age-related macular degeneration. Surv Ophthalmol 59: 532-539.
  148. Abokyi S, To CH, Lam TT, Tse DY (2020) Central role of oxidative stress in age-related macular degeneration: Evidence from a review of the molecular mechanisms and animal models. Oxid Med Cell Longev 2020: 7901270. [Crossref]
  149. Hollyfield JG, Bonilha VL, Rayborn ME, Yang X, Shadrach KG, et al. (2008) Oxidative damage-induced inflammation initiates age-related macular degeneration. Nat Med 14: 194-198. [Crossref]
  150. Ebrahem Q, Renganathan K, Sears J, Vasanji A, Gu X, et al. (2006) Carboxyethylpyrrole oxidative protein modifications stimulate neovascularization: Implications for age-related macular degeneration. Proc Natl Acad Sci U S A 103: 13480-13484.
  151. Doyle SL, Campbell M, Ozaki E, Salomon RG, Mori A, et al. (2012) NLRP3 has a protective role in age-related macular degeneration through the induction of IL-18 by drusen components. Nat Med 18: 791-798. [Crossref]
  152. Saeed AM, Duffort S, Ivanov D, Wang H, Laird JM, et al. (2014) The oxidative stress product carboxyethylpyrrole potentiates TLR2/TLR1 inflammatory signaling in macrophages. PLoS One 9: e106421. [Crossref]
  153. Hollyfield JG, Perez VL, Salomon RG (2010) A hapten generated from an oxidation fragment of docosahexaenoic acid is sufficient to initiate age-related macular degeneration. Mol Neurobiol 41: 290-298. [Crossref]
  154. Hollyfield JG, Salomon RG, Crabb JW (2003) Proteomic approaches to understanding age-related macular degeneration. Adv Exp Med Biol 533: 83-89.
  155. Gu J, Pauer GJ, Yue X, Narendra U, Sturgill GM, et al. (2009) Assessing susceptibility to age-related macular degeneration with proteomic and genomic biomarkers. Mol Cell Proteomics 8: 1338-1349. [Crossref]
  156. Lu L, Gu X, Hong L, Laird J, Jaffe K, et al. (2009) Synthesis and structural characterization of carboxyethylpyrrole-modified proteins: mediators of age-related macular degeneration. Bioorg Med Chem 17: 7548-7561.
  157. Ni J, Yuan X, Gu J, Yue X, Gu X, et al. (2009) Plasma protein pentosidine and carboxymethyllysine, biomarkers for age-related macular degeneration. Mol Cell Proteomics 8: 1921-1933. [Crossref]
  158. Del Bo' C, Martini D, Porrini M, Klimis-Zacas D, Riso P (2015) Berries and oxidative stress markers: an overview of human intervention studies. Food Funct 6: 2890-2917. [Crossref]
  159. Di Domenico F, Coccia R, Butterfield DA, Perluigi M (2011) Circulating biomarkers of protein oxidation for Alzheimer disease: expectations within limits. Biochim Biophys Acta 1814: 1785-1795. [Crossref]
  160. Thanan R, Oikawa S, Hiraku Y, Ohnishi S, Ma N, et al. (2014) Oxidative stress and its significant roles in neurodegenerative diseases and cancer. Int J Mol Sci 16: 193-217. [Crossref]
  161. Levine RL, Garland D, Oliver CN, Amici A, Climent I, et al. (1990) Determination of carbonyl content in oxidatively modified proteins. Methods Enzymol 186: 464-478. [Crossref]
  162. Davies MJ, Fu S, Wang H, Dean RT (1999) Stable markers of oxidant damage to proteins and their application in the study of human disease. Free Radic Biol Med 27: 1151-1163. [Crossref]
  163. Dalle-Donne I, Giustarini D, Colombo R, Rossi R, Milzani A (2003) Protein carbonylation in human diseases. Trends Mol Med 9: 169-176. [Crossref]
  164. Levin RL (2002) Carbonyl modified proteins in cellular regulation, aging, and disease. Free Radic Biol Med 32: 790-796. [Crossref]
  165. Bulteau AL, Szweda LI, Friguet B (2006) Mitochondrial protein oxidation and degradation in response to oxidative stress and aging. Exp Gerontol 41: 653-657. [Crossref]
  166. Stadtman ER, Levine RL (2000) Protein oxidation. Ann N Y Acad Sci 899: 191-208. [Crossref]
  167. Phaniendra A, Jestadi DB, Periyasamy L (2015) Free radicals: properties, sources, targets, and their implication in various diseases. Indian J Clin Biochem 30: 11-26. [Crossref]
  168. Trnkova L, Drsata J, Bousova I (2015) Oxidation as an important factor of protein damage: Implications for Maillard reaction. J Biosci 40: 419-439.
  169. Zafrilla P, Losada M, Perez A, Caravaca G, Mulero J (2013) Biomarkers of oxidative stress in patients with wet age related macular degeneration. J Nutr Health Aging 17: 219-222. [Crossref]
  170. Totan Y, Yagci R, Bardak Y, Ozyurt H, Kendir F, et al. (2009) Oxidative macromolecular damage in age-related macular degeneration. Curr Eye Res 34: 1089-1093. [Crossref]
  171. Schmidt AM, Hori O, Brett J, Yan SD, Wautier JL, et al. (1994) Cellular receptors for advanced glycation end products. Implications for induction of oxidant stress and cellular dysfunction in the pathogenesis of vascular lesions. Arterioscler Thromb 14: 1521-1528. [Crossref]
  172. Brownlee M (1995) Advanced protein glycosylation in diabetes and aging. Annu Rev Med 46: 223-234. [Crossref]
  173. Ahmed MU, Thorpe SR, Baynes JW (1986) Identification of N-ε-carboxymethyllysine as a degradation product of fructoselysine in glycated protein. J Biol Chem 261: 4889-4894. [Crossref]
  174. Yamagishi S, Adachi H, Takeuchi M, Enomoto M, Furuki K, et al. (2007) Serum level of advanced glycation end-products (AGEs) is an independent determinant of plasminogen activator inhibitor-1 (PAI-1) in nondiabetic general population. Horm Metab Res 39: 845-848. [Crossref]
  175. Lue LF, Yan SD, Stern DM, Walker DG (2005) Preventing activation of receptor for advanced glycation endproducts in Alzheimer's disease. Curr Drug Targets CNS Neurol Disord 4: 249-266. [Crossref]
  176. Harja E, Bu DX, Hudson BI, Chang JS, Shen X, et al. (2008) Vascular and inflammatory stresses mediate atherosclerosis via RAGE and its ligands in apoE-/- mice. J Clin Invest 118: 183-194. [Crossref]
  177. Gul A, Rahman MA, Hasnain SN (2009) Role of fructose concentration on cataractogenesis in senile diabetic and non-diabetic patients. Graefes Arch Clin Exp Ophthalmol 247: 809-814.
  178. McCarty CA, Mukesh BN, Fu CL, Mitchell P, Taylor HR (2001) Risk factors for age-related maculopathy: the visual impairment project. Arch Ophthalmol 119: 1455-1462. [Crossref]
  179. Ishibashi T, Murata T, Hangai M, Nagai R, Horiuchi S, et al. (1998) Advanced glycation end products in age-related macular degeneration. Arch Ophthalmol 116: 1629-1632. [Crossref]
  180. Handa JT, Verzijl N, Matsunaga H, Aotaki-Keen A, Lutty GA, et al. (1999) Increase in the advanced glycation end product pentosidine in Bruch’s membrane with age. Invest Ophthalmol Vis Sci 40: 775-779.
  181. Glenn JV, Beattie JR, Barrett L, Frizzell N, Thorpe SR, et al. (2007) Confocal Raman microscopy can quantify advanced glycation end product (AGE) modifications in Bruch's membrane leading to accurate, nondestructive prediction of ocular aging. FASEB J 21: 3542-3552. [Crossref]
  182. Hammes HP, Hoerauf H, Alt A, Schleicher E, Clausen JT, et al. (1999) N(epsilon)(carboxymethyl)lysin and the AGE receptor RAGE colocalize in age-related macular degeneration. Invest Ophthalmol Vis Sci 40: 1855-1859. [Crossref]
  183. Howes KA, Liu Y, Dunaief JL, Milam A, Frederick JM, et al. (2004) Receptor for advanced glycation end products and age-related macular degeneration. Invest. Ophthalmol Vis Sci 45: 3713-3720. [Crossref]
  184. Yamada Y, Ishibashi K, Ishibashi K, Bhutto IA, Tian J, et al. (2006) The expression of advanced glycation endproduct receptors in rpe cells associated with basal deposits in human maculas. Exp Eye Res 82: 840-848. [Crossref]
  185. Farboud B, Aotaki-Keen A, Miyata T, Hjelmeland LM, Handa JT (1999) Development of a polyclonal antibody with broad epitope specificity for advanced glycation endproducts and localization of these epitopes in Bruch's membrane of the aging eye. Mol Vis 5: 11. [Crossref]
  186. Glenn JV, Mahaffy H, Wu K, Smith G, Nagai R, et al. (2009) Advanced glycation end product (AGE) accumulation on Bruch's membrane: links to age-related RPE dysfunction. Invest Ophthalmol Vis Sci 50: 441-451. [Crossref]
  187. Lin T, Walker GB, Kurji K, Fang E, Law G, et al. (2013) Parainflammation associated with advanced glycation endproduct stimulation of RPE in vitro: implications for age-related degenerative diseases of the eye. Cytokine 62: 369-381. [Crossref]
  188. Okamoto T, Tanaka S, Stan AC, Koike T, Kase M, et al. (2002) Advanced glycation end products induce angiogenesis in vivo. Microvasc Res 63: 186-195. [Crossref]
  189. Hoffmann S, Friedrichs U, Eichler W, Rosenthal A, Wiedemann P (2002) Advanced glycation end products induce choroidal endothelial cell proliferation, matrix metalloproteinase-2 and VEGF upregulation in vitro. Graefes Arch Clin Exp Ophthalmol 240: 996-1002. [Crossref]
  190. Ohgami N, Nagai R, Ikemoto M, Arai H, Kuniyasu A, et al. (2001) CD36, a member of class B scavenger receptor family, is a receptor for advanced glycation end products. Ann N Y Acad Sci 947: 350-355. [Crossref]
  191. Schmidt AM, Vianna M, Gerlach M, Brett J, Ryan J, et al. (1992) Isolation and characterization of two binding proteins for advanced glycosylation end products from bovine lung which are present on the endothelial cell surface. J Biol Chem 267: 14987-14997. [Crossref]
  192. Neeper M, Schmidt AM, Brett J, Yan SD, Wang F, et al. (1992) Cloning and expression of a cell surface receptor for advanced glycosylation end products of proteins. J Biol Chem 267: 14998-15004. [Crossref]
  193. Banevicius M, Vilkeviciute A, Kriauciuniene L, Liutkeviciene R, Deltuva VP (2018) The association between variants of Receptor for Advanced Glycation End Products (RAGE) gene polymorphisms and age-related macular degeneration. Med Sci Monit 24: 190-199. [Crossref]
  194. Yuan X, Gu X, Crabb JS, Yue X, Shadrach K, et al. (2010) Quantitative proteomics: comparison of the macular Bruch membrane/choroid complex from age-related macular degeneration and normal eyes. Mol Cell Proteomics 9: 1031-1046. [Crossref]
  195. Lander HM, Tauras JM, Ogiste JS, Hori O, Moss RA, et al. (1997) Activation of the receptor for advanced glycation end products triggers a p21(ras)-dependent mitogen-activated protein kinase pathway regulated by oxidant stress. J Biol Chem 272: 17810-17814. [Crossref]
  196. Yan SD, Schmidt AM, Anderson GM, Zhang J, Brett J, et al. (1994) Enhanced cellular oxidant stress by the interaction of advanced glycation end products with their receptors/binding proteins. J Biol Chem 269: 9889-9897. [Crossref]
  197. Torres RJA, Torres R, Luchini A, Ferreira ALA (2021) Transcription factor NF-κβ and molecules derived from its activation in age-related macular degeneration”. IJMS. 2021; 8 (March).
  198. Li J, Schmidt AM (1997) Characterization and functional analysis of the promoter of RAGE, the receptor for advanced glycation end products. J Biol Chem 272: 16498-16506. [Crossref]
  199. Ni J, Yuan X, Gu J, Yue X, Gu X, et al. (2009) Clinical genomic and proteomic AMD study group. plasma protein pentosidine and carboxymethyllysine, biomarkers for age-related macular degeneration. Mol Cell Proteomics 8: 1921-1933.
  200. Mulder DJ, Bieze M, Graaff R, Smit AJ, Hooymans JM (2010) Skin autofluorescence is elevated in neovascular age-related macular degeneration. Br J Ophthalmol 94: 622-625.
  201. Semba RD, Cotch MF, Gudnason V, Eiríksdottir G, Harris TB, et al. (2014) Serum carboxymethyllysine, an advanced glycation end product, and age-related macular degeneration: the Age, Gene/Environment Susceptibility-Reykjavik Study. JAMA Ophthalmol 132: 464-470. [Crossref]
  202. Ksiazek K, Piatek K, Witowski J (2008) Impaired response to oxidative stress in senescent cells may lead to accumulation of DNA damage in mesothelial cells from aged donors. Biochem Biophys Res Commun 373: 335-339. [Crossref]
  203. Xu G, Herzig M, Rotrekl V, Walter CA (2008) Base excision repair, aging and health span. Mech Ageing Dev 129: 366-382. [Crossref]
  204. Alexander P (1967) The role of DNA lesions in the processes leading to aging in mice. Symp Soc Exp Biol 21: 29-50. [Crossref]
  205. Baynes JW (2002) The Maillard hypothesis on aging: time to focus on DNA. Ann N Y Acad Sci 959: 360-367. [Crossref]
  206. Szaflik JP, Janik-Papis K, Synowiec E, Ksiazek D, Zaras M, et al. (2009) DNA damage and repair in age-related macular degeneration. Mutat Res 669: 169-176. [Crossref]
  207. Kasai H (1997) Analysis of a form of oxidative DNA damage, 8-hydroxy-2'-deoxyguanosine, as a marker of cellular oxidative stress during carcinogenesis. Mutat Res 387: 147-163. [Crossref]
  208. Colak E (2008) New markers of oxidative damage to macromolecules. J Med Biochem 27: 1-16.
  209. Dabrowska N, Wiczkowski A (2017) Analytics of oxidative stress markers in the early diagnosis of oxygen DNA damage. Adv Clin Exp Med 26: 155-166. [Crossref]
  210. Kondo T, Ohshima T, Ishida Y (2001) Age-dependent expression of 8-hydroxy- 2' -deoxyguanosine in human pituitary gland. Histochem J 33: 647-651. [Crossref]
  211. Van Remmen H, Hamilton ML, Richardson A (2003) Oxidative damage to DNA and aging. Exerc Sport Sci Rev 31: 149-153. [Crossref]
  212. Wang AL, Lukas TJ, Yuan M, Neufeld AH (2008) Increased mitochondrial DNA damage and down-regulation of DNA repair enzymes in aged rodent retinal pigment epithelium and choroid. Mol Vis 14: 644-651. [Crossref]
  213. Kim SY, Kambhampati SP, Bhutto IA, McLeod DS, Lutty GA, et al. (2021) Evolution of oxidative stress, inflammation and neovascularization in the choroid and retina in a subretinal lipid induced age-related macular degeneration model. Exp Eye Res 203: 108391.
  214. Zhang Y, Cross SD, Stanton JB, Marmorstein AD, Le YZ, et al. (2017) Early AMD-like defects in the RPE and retinal degeneration in aged mice with RPE-specific deletion of Atg5 or Atg7. Mol Vis 23: 228-241.
  215. Park C, Lee H, Hong SH, Kim JH, Park SK, et al. (2019) Protective effect of diphlorethohydroxycarmalol against oxidative stress-induced DNA damage and apoptosis in retinal pigment epithelial cells. Cutan Ocul Toxicol 38: 298-308. [Crossref]
  216. Kim GH, Kim HI, Paik SS, Jung SW, Kang S, et al. (2016) Functional and morphological evaluation of blue light-emitting diode-induced retinal degeneration in mice. Graefes Arch Clin Exp Ophthalmol 254: 705-716. [Crossref]
  217. Otsuka T, Shimazawa M, Nakanishi T, Ohno Y, Inoue Y, et al. (2013) Protective effects of a dietary carotenoid, astaxanthin, against light-induced retinal damage. J Pharmacol Sci 123: 209-218.
  218. Fujihara M, Nagai N, Sussan TE, Biswal S, Handa JT (2008) Chronic cigarette smoke causes oxidative damage and apoptosis to retinal pigmented epithelial cells in mice. PLoS One 3: e3119.
  219. Marazita MC, Dugour A, Marquioni-Ramella MD, Figueroa JM, Suburo AM (2016) Oxidative stress-induced premature senescence dysregulates VEGF and CFH expression in retinal pigment epithelial cells: Implications for Age-related Macular Degeneration. Redox Biol 7: 78-87. [Crossref]
  220. Hernandez M, Recalde S, González-Zamora J, Bilbao-Malavé V, Sáenz de Viteri M, et al. (2021) Anti-inflammatory and anti-oxidative synergistic effect of Vitamin D and nutritional complex on retinal pigment epithelial and endothelial cell lines against age-related macular degeneration. Nutrients 13: 1423. [Crossref]
  221. Ojino K, Shimazawa M, Ohno Y, Otsuka T, Tsuruma K, et al. (2014) Protective effect of SUN N8075, a free radical scavenger, against excessive light-induced retinal damage in mice. Biol Pharm Bull 37: 424-430. [Crossref]
  222. Valavanidis A, Vlachogianni T, Fiotakis C (2009) 8-hydroxy-2' -deoxyguanosine (8-OHdG): A critical biomarker of oxidative stress and carcinogenesis. J Environ Sci Health C Environ Carcinog Ecotoxicol Rev 27: 120-139.
  223. Ma Z, Liu J, Li J, Jiang H, Kong J (2020) Klotho levels are decreased and associated with enhanced oxidative stress and inflammation in the aqueous humor in patients with exudative age-related macular degeneration. Ocul Immunol Inflamm 13: 1-8. [Crossref]
  224. Lau LI, Liu CJ, Wei YH (2010) Increase of 8-hydroxy-2'-deoxyguanosine in aqueous humor of patients with exudative age-related macular degeneration. Invest Ophthalmol Vis Sci 51: 5486-5490. [Crossref]
  225. Wu R, Feng J, Yang Y, Dai C, Lu A, et al. (2017) Significance of serum total oxidant/antioxidant status in patients with colorectal cancer. PLoS One 12: e0170003. [Crossref]
  226. Tarpey MM, Wink DA, Grisham MB (2004) Methods for detection of reactive metabolites of oxygen and nitrogen: in vitro and in vivo considerations. Am J Physiol Regul Integr Comp Physiol 286: R431-R444. [Crossref]
  227. Erel O (2005) A new automated colorimetric method for measuring total oxidant status. Clin Biochem 38: 1103-1111. [Crossref]
  228. Ugurlu N, Asik MD, Yülek F, Neselioglu S, Cagil N (2013) Oxidative stress and anti-oxidative defence in patients with age-related macular degeneration. Curr Eye Res 38: 497-502. [Crossref]
  229. Elbay A, Ozer OF, Akkan JCU, Celik U, Kutlutürk I, et al. (2017) Comparison of serum thiol-disulphide homeostasis and total antioxidant-oxidant levels between exudative age-related macular degeneration patients and healthy subjects. Int Ophthalmol 37: 1095-1101. [Crossref]

Editorial Information

Editor-in-Chief

James B. Aguayo-Martel
California Northstate University College of Medicine
USA

Article Type

Research Article

Publication history

Received date: August 25, 2021
Accepted date: October 15, 2021
Published date: October 19, 2021

Copyright

©2021 Torres RJ. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.

Citation

Torres RJ, Luchini A, Torres RJ, Ferreira LA (2021) Potential molecular biomarkers of oxidative stress in agerelated macular degeneration. New Front Ophthalmol 7: DOI: 10.15761/NFO.1000259

Corresponding author

Rogil Jose de Almeida Torres

Rua Emiliano Perneta 390, Conjunto 1407, CEP 80420-080, Brazil

E-mail : bhuvaneswari.bibleraaj@uhsm.nhs.uk

No data.