Next Article in Journal
A Novel Surface-Exposed Polypeptide Is Successfully Employed as a Target for Developing a Prototype One-Step Immunochromatographic Strip for Specific and Sensitive Direct Detection of Staphylococcus aureus Causing Neonatal Sepsis
Next Article in Special Issue
Effects of Diabetes on Microcirculation and Leukostasis in Retinal and Non-Ocular Tissues: Implications for Diabetic Retinopathy
Previous Article in Journal
Clinical Applications of Patient-Specific 3D Printed Models in Cardiovascular Disease: Current Status and Future Directions
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Acrolein: A Potential Mediator of Oxidative Damage in Diabetic Retinopathy

by
Moaddey Alfarhan
1,2,
Eissa Jafari
1,2 and
S. Priya Narayanan
1,2,*
1
Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia and Charlie Norwood VA Medical Center, Augusta, GA 30912, USA
2
Vision Discovery Institute, Augusta University, Augusta, GA 30912, USA
*
Author to whom correspondence should be addressed.
Biomolecules 2020, 10(11), 1579; https://doi.org/10.3390/biom10111579
Submission received: 11 October 2020 / Revised: 5 November 2020 / Accepted: 17 November 2020 / Published: 20 November 2020

Abstract

:
Diabetic retinopathy (DR) is the leading cause of vision loss among working-age adults. Extensive evidences have documented that oxidative stress mediates a critical role in the pathogenesis of DR. Acrolein, a product of polyamines oxidation and lipid peroxidation, has been demonstrated to be involved in the pathogenesis of various human diseases. Acrolein’s harmful effects are mediated through multiple mechanisms, including DNA damage, inflammation, ROS formation, protein adduction, membrane disruption, endoplasmic reticulum stress, and mitochondrial dysfunction. Recent investigations have reported the involvement of acrolein in the pathogenesis of DR. These studies have shown a detrimental effect of acrolein on the retinal neurovascular unit under diabetic conditions. The current review summarizes the existing literature on the sources of acrolein, the impact of acrolein in the generation of oxidative damage in the diabetic retina, and the mechanisms of acrolein action in the pathogenesis of DR. The possible therapeutic interventions such as the use of polyamine oxidase inhibitors, agents with antioxidant properties, and acrolein scavengers to reduce acrolein toxicity are also discussed.

1. Introduction

Diabetic retinopathy (DR), a complication of diabetes, is a significant public health issue and the leading cause of vision loss among working-age adults [1]. DR accounts for almost 40% of diabetes mellitus complications in patients aged ≥40 years [2]. It is estimated that the prevalence of any form of DR is about 93 million worldwide [3]. DR patients’ quality of life is negatively impacted, affecting their mental health and general vision, and is decreased with the duration of the disease [4]. DR develops in both type 1 and type 2 diabetic patients. It is estimated that individual lifetime risk of DR is up to 90% in patients with type 1 diabetes mellitus and 50–60% in those with type 2 diabetes mellitus [5,6]. The unavailability of effective treatments to prevent the incidence or progression of DR is a major clinical problem. The current therapeutic avenues for DR are anti-VEGF agents, anti-inflammatory drugs, and laser treatment. However, they treat advanced stages of the disease, particularly the vascular damage, and have adverse side effects. Hence there is a great need for the identification of novel therapeutic interventions for DR.
All the components of the retinal neurovascular unit are disrupted by diabetes [7]. The precise mechanisms through which diabetes causes retinal damage and degeneration remain to be fully established. Extensive evidences have documented that oxidative stress plays a critical role in the pathogenesis of DR. Formation of reactive oxygen species (ROS) has been shown to play important roles in DR progression [8]. Several pathways and molecules have been studied to be associated with elevated oxidative stress in the diabetic retina [8,9,10,11,12]. Dysregulation in the polyamine pathway is involved in the pathogenesis of diabetes and diabetic retinopathy [13,14,15]. The oxidative products of polyamine metabolism can induce cellular damage and cell death [16,17,18]. Reactive aldehydes and hydrogen peroxide (H2O2) are generated from the metabolism of polyamines [19]. These reactive aldehydes are spontaneously converted to acrolein [16,20], a potent mediator of oxidative damage in various pathologies [21,22].
Acrolein is involved in different pathologies and plays a vital role in the progression of atherosclerosis [23], cardiovascular disease [24], pulmonary inflammation [25], and kidney disease [26]. Additionally, acrolein is involved in neurodegenerative diseases such as Alzheimer’s disease [27], multiple sclerosis [28], brain infarction [29,30,31,32], and spinal cord injury [33]. Acrolein exposure may cause cognitive problems and may lead to serious neurocognitive effects on humans [34]. Acrolein’s harmful effects are mediated through multiple mechanisms, including DNA damage, inflammation, ROS formation, protein adduction, membrane disruption, endoplasmic reticulum stress, and mitochondrial dysfunction [21]. In vitro studies revealed that injury induced by acrolein was more toxic than ROS [20,35] and other aldehydes produced during polyamine metabolism [36]. This oxidative damage caused by acrolein leads to mitochondrial dysfunction, membrane disruption, and increased apoptosis [21,37,38]. Recently, several investigations have reported the emerging role of acrolein in the progression of DR. The current review summarizes the existing literature on the various sources of acrolein, the impact of acrolein in the generation of oxidative damage in the diabetic retina, and the mechanisms of acrolein action in the pathogenesis of DR.

2. Acrolein: Sources and Metabolism

Acrolein is a highly reactive unsaturated aldehyde. It is found as a contaminant in food, air, and water. The major sources of acrolein include dietary, environmental, and endogenous sources. Acrolein is an environmental pollutant generated in the environment during incomplete combustion of plastic, combustion of petrochemical fuels, and tobacco smoke [39]. In addition, acrolein is produced from overheated vegetable oils and animal fats [40]. Endogenously, acrolein is formed during the peroxidation of polyunsaturated fatty acids [41] or polyamine catabolism [16]. Free radicals generated from ROS react with the polyunsaturated fatty acids (PUFAs), forming lipid radicals and lipid peroxyl radicals. These are highly unstable and are further degraded into reactive, secondary products such as acrolein, 4-hydroxynonenal (4-HNE), and malondialdehyde (MDA) [42,43]. These aldehydes react with cellular proteins, leading to advanced lipoxidation end products (ALEs) formation [42,43,44]. The other endogenous source for acrolein formation is the oxidation of spermine and spermidine [39]. During polyamine oxidation, H2O2 and aldehydes, including 3-acetamidopropanal (3-AAP), amino aldehydes 3-aminopropanal (3-AP), and 4-aminobutanal (4-AB), are formed [36,45]. Spermine and spermidine are converted to spermidine and putrescine, respectively, through back-conversion by SSAT (spermidine/spermine N1-acetyltransferase) to N1-acetylated polyamines. N1-acetylated polyamines are oxidized by polyamine oxidase (PAO) to produce spermidine and putrescine, respectively [46]. In addition to the PAO oxidation pathway, spermine can also be oxidized by spermine oxidase (SMOX) to spermidine directly [46]. Therefore, both PAO and SMOX oxidation pathways are involved in generating acrolein from polyamines [47]. The formation of acrolein through lipid peroxidation and polyamine oxidation pathways are presented in Figure 1A,B.
Acrolein is highly soluble in water and alcohol and can cross the membranes by passive diffusion. The mode of elimination of acrolein is by conjugation with glutathione (GSH) in the liver and N-acetylation of the resultant cysteine conjugate to form S-(3-oxopropyl)-N-acetylcysteine (OPMA) in the kidney. OPMA is reduced to form N-acetyl-S-(3-hydroxypropyl)-l-cysteine (or 3-hydroxypropyl mercapturic acid, 3HPMA), the major urinary metabolite of acrolein, and an oxidative route which yields N-acetyl-S-[2-carboxyethyl]-l-cysteine (or 2-carboxyethyl-mercapturic acid, CEMA), a minor urinary metabolite [39,48,49]. These metabolites, 3HPMA and CEMA offer considerable promise as urinary biomarkers. Oxidation of acrolein can lead to the formation of acrylic acid. Enzyme-mediated epoxidation of acrolein produces glycidaldehyde, which can react with water to yield glyceraldehyde or form a conjugate with GSH. Additional acrolein metabolites detected in urine include 3-hydroxy propionic acid, malonic acid, and N-acetyl-S-(2-carboxy-2-hydroxyethyl)-cysteine [39]. Since acrolein is highly unstable, measurements of acrolein metabolites are useful indicators of its internal levels. Various methods are implemented for the detection of acrolein conjugates or metabolites. Liquid chromatography-tandem mass spectrometry (LC/MS) analysis and high-performance liquid chromatography (HPLC) are widely used for the detection of acrolein metabolites in biological fluids [50,51,52,53]. The presence of acrolein conjugates is studied by slot blot assays [51], ELISA [54,55], or immunochemical methods [56,57].

3. Acrolein in the Progression of Diabetic Retinopathy

3.1. Acrolein and Diabetes

Studies have reported the association of acrolein in both type 1 and type 2 diabetes. Elevated urinary level of conjugated acrolein is reported in patients with type I and type 2 diabetes [58,59]. These clinical studies illustrated that elevated urinary acrolein level is linked to the diabetic stage and significantly correlated to the glycemic status [58,59]. In a group of younger type 1 diabetic patient, the urine level of conjugated acrolein, along with other markers of oxidative stress (pentosidine, pyrraline, and 8-hydroxy-2′-deoxyguanosine), was shown to be significantly elevated than healthy subjects [58]. The study performed by Daimon et al. and colleagues [59] in type 2 diabetic patients showed significantly increased levels of acrolein adduct, pentosidine, and pyrraline in urine samples. Furthermore, glycemic control parameters, such as fasting plasma glucose and HbA1c (hemoglobin A1c, the glycated hemoglobin level), were observed to be correlated with the urinary levels of these markers. The acrolein adduct level was higher in subjects with smoking habits than in those without the habit in the diabetic and nondiabetic groups [59]. In addition, acrolein protein adducts are associated with diabetes-related complications such as diabetic nephropathy [60] and diabetic retinopathy [61].
Acrolein feeding elicited dyslipidemia in a mouse model, with increased circulating cholesterol and triglycerides [62,63]. The mechanism behind this acrolein-induced dyslipidemia involved the processes of lipid synthesis and clearance. Acrolein treatment reduced VLDL clearance via decreasing hepatic lipase activity and downregulating LDL receptors, leading to modulating the VLDL clearance and increasing the circulating cholesterol and triglycerides [62]. Acute acrolein feeding modified the expression of plasma and hepatic genes involved with lipid synthesis and trafficking and increased plasma triglycerides [63]. Diabetes is well known to cause dyslipidemia (a condition with abnormal cholesterol levels and lipid in the blood), which increases lipid peroxidation even in well-controlled diabetic patients [64]. Dyslipidemia and lipid peroxidation can lead to increased acrolein production, thus causing diabetes-related complications [41,64]. In addition to lipid peroxidation, increased activity of polyamine oxidases can also cause acrolein formation. An earlier report has indicated a positive correlation between hyperglycemia and PAO activity and that the activity of PAO is positively correlated to the level of HbA1c [65]. These observations suggest that diabetic complications can possibly be triggered by acrolein produced either via lipid peroxidation or polyamine oxidation. A recent report indicated an age-dependent increase in acrolein conjugate, polyamine oxidation, and SMOX in the liver of human subjects [66].

3.2. Pathogenesis of DR

DR’s incidence and progression are affected by various factors, including duration of diabetes, age, pregnancy, hyperglycemia, hyperlipidemia, obesity, smoking, etc. [10]. It is recognized as a neurovascular disease, and neurodegeneration and vasculopathy are the major hallmarks of DR progression. Diabetes-induced retinal neurodegeneration is characterized by the progressive thinning of the nerve fiber layer, loss of retinal ganglion cells, and alterations in visual function [15,67,68,69,70,71]. While neurodegeneration is considered as an early event, the clinical classification of DR is based on its vascular complications. These include increased vascular permeability, endothelial dysfunction, vascular degeneration, and pathological angiogenesis [72,73]. In addition to neuronal and vascular changes, the diabetic retina exhibits increased inflammatory changes [74,75,76] and glial activation [77,78]. Müller cells, the principal glia of the retina, serve to maintain the extracellular environment’s homeostasis and play an important role in the pathogenesis of DR [79]. In addition, Müller cells are a part of the inner blood–retinal barrier cells and regulate retinal blood flow [79]. It is known that Müller cells become activated in the retina during diabetes, and the expression of the glial fibrillary acidic protein (GFAP) is increased in the Müller cells and is a characteristic feature of the diabetic retina [80]. Several mechanisms/pathways have been demonstrated to be involved in the pathogenesis of DR [81,82]. Oxidative stress has been established as a significant contributor to DR pathogenesis. It has been confirmed that metabolic pathways, including advanced glycation end products (AGEs), polyol, hexosamine, and protein kinase C (PKC), are activated under diabetic conditions. Accumulation of by-products of these pathways can induce oxidative stress through the formation of ROS and nitrogen-oxygen species. Studies from our laboratory are the first to demonstrate polyamine oxidation in mediating neuronal and vascular damage in the retina [83,84]. Studies from our group [15,83] and others [12,85] have shown the involvement of SMOX, a crucial enzyme in the polyamine oxidation pathway in DR. Altered levels of polyamines are reported in the vitreous samples from patients with proliferative DR [86]. Endogenous spermine, a polyamine, demonstrated role in the distribution and regulation of voltage-dependent calcium channels in the diabetic retina [87].

3.3. Acrolein Conjugates in the Diabetic Retina

Studies have shown the upregulation of lipid peroxidation and its metabolites in the serum of DR patients [88,89] as well as in the retinas of experimental diabetic models [90,91], emphasizing the role of lipid peroxidation in the progression of diabetes. Several investigations have demonstrated the involvement of polyamine oxidation and lipid peroxidation as the major pathways of acrolein generation in the diabetic retina [15,92,93]. Diabetic patients with DR are found to have much more lipid peroxidation products compared with those without DR [94]. Acrolein is a strong electrophile and hence shows high reactivity with proteins, DNA, and RNA. Acrolein reacts with the sulfhydryl group of cysteine, the imidazole group of histidine, and the amino group of lysine, to form mainly Michael addition-type adducts or Schiff base cross-links [95,96]. Protein adduction by acrolein can cause significant protein modifications leading to alterations in protein functions. FDP-lysine, also known as a biomarker of acrolein, is formed when acrolein conjugates with lysine residue to form Nε-(3-formyl-3,4-dehydropiperidino)lysine (FDP-lysine) [41]. An earlier study consisting of type 1 and type 2 diabetic patients investigated the correlation between the level of FDP-lysine, (serum and hemoglobin), and the severity of diabetic retinopathy [97]. Compared with the control group, the serum and hemoglobin levels of FDP-lysine were significantly increased in diabetic patients. The levels of hemoglobin FDP-lysine were increased in patients with proliferative retinopathy compared with patients without retinopathy and with nonproliferative retinopathy, however, no association was observed between serum FDP-lysine and severity of DR. FDP-lysine is increased significantly in the vitreous fluids of patients with proliferative diabetic retinopathy (PDR) [98]. Pieces of evidence demonstrated increased levels of FDP-lysine or conjugated acrolein in the retina of diabetic rodents [15,78,99]. In an earlier study, significantly elevated accumulation of FDP-lysine in the Müller cells was observed in the retinas of diabetic animals, and the accumulation was shown to be progressive with the duration of diabetes [99]. Along with other oxidative stress markers, acrolein was reported to be increased in the retinas of diabetic rats [100,101,102]. Altogether, these observations suggest a correlation between acrolein formation and the progression of DR.
Aldehyde dehydrogenases (ALDH) are the enzymes that metabolize aldehydes to lesser toxic compounds [103]. ALDH1A1 is one of the ALDH enzymes that participate in lipid peroxidation detoxifying aldehydes [104,105]. Interestingly, retinas of diabetic rats showed a reduced transcription and expression of ALDH1A1 and reduced activity of ALDH [57]. Acrolein is detoxified when it is conjugated with glutathione (GSH), and this reaction is catalyzed by glutathione S transferase (GST) [39]. However, a reduction in the GSH level was observed in diabetic rat retinas [106]. In an in vitro experimental model, retinal pigment epithelial (RPE) cells showed a reduction in GSH level and a reduction in GST activity when cells were treated with acrolein [107]. These studies suggest the impact of acrolein in elevating oxidative stress in diabetes-induced pathologies in the retina.

4. Mechanism of Action of Acrolein in the Diabetic Retina

Acrolein mediates its function by diverse mechanisms. These include direct mechanisms of acrolein toxicity such as protein and DNA adduction and via indirect mechanisms, including oxidative, mitochondrial, and ER stress [21]. Protein modifications by protein adduction is a major mechanism of acrolein-mediated cellular dysfunction. Both in vitro and in vivo evidence have shown that acrolein causes oxidative damage. In the following section, the central mechanisms of acrolein-induced damages investigated in the diabetic retina are summarized.

4.1. Depletion of Antioxidants

The compromised antioxidant defense system is one of the causes of elevated oxidative stress in the diabetic retina. This is a major mechanism by which acrolein induces cellular oxidative stress and is executed by the formation of acrolein protein adducts. Because of acrolein’s significant reactivity with thiols, GSH is one of the primary targets of acrolein-mediated injury [39]. Even though the reaction of GSH with acrolein is essential for the endogenous removal of acrolein, depletion of GSH reserves reduces the ability to handle additional oxidative stress [108]. Acrolein decreased the levels of antioxidants such as GSH, intracellular glutathione peroxidase (G-Px), GST, and superoxide dismutase (SOD) in RPE cells [109,110,111]. Acrolein exposure also reduced the total antioxidant capacity (T-AOC) and the expression of glutamate-cysteine ligase (GCL), the enzyme that controls GSH production [111]. Moreover, RPE cells exposed to acrolein showed a reduction in nuclear factor-e2-related factor 2 (Nrf2), a regulator of antioxidant response [109,110,111]. GSH expression was shown to be decreased in the rat retinal Müller cell line TR-MUL5 exposed to acrolein treatments [112]. Studies have shown that acrolein depletes the cellular antioxidant levels (including GSH) by conjugating with thiol groups [113,114]. In an experimental model of diabetes, the accumulation of FDP-lysine in Müller cells was increased and associated with an elevated level of heme oxygenase-1 (HO-1) [78], a marker of oxidative stress [115].

4.2. Protein Carbonyl Formation

The formation of protein carbonyls is a major cause of oxidative stress in the diabetic retina. Acrolein can carbonylate proteins to generate reactive carbonyl species [110,116] and increase the oxidative damage by protein carbonylation [96,117]. Recent reports show that protein carbonyl levels are elevated in DR patients [118,119,120] and experimental models [121,122]. The presence of protein carbonyls is also reported in models of photoreceptor degeneration [123] and glaucoma [124]. Acrolein exposure demonstrated to increase the generation of ROS in RPE cells [107,109,110,111] and a rat retinal Müller cell line [112]. The increased oxidative stress in RPE cells caused DNA fragmentation and protein oxidation by increasing protein carbonyl levels [109,110,111]. However, the specific mechanisms by which protein carbonyls are increased in DR needs further investigation.

4.3. Mitochondrial Dysfunction

Oxidative damage-mediated mitochondrial dysfunction is a major mechanism for oxidative stress in DR [125]. While mitochondria are essential for cell growth and energy production, they are also responsible for ROS formation [126]. Acrolein is known as a mitochondrial toxin and can impair mitochondrial respiratory function [38,127]. In RPE cells, acrolein induced mitochondrial dysfunction by decreasing mitochondrial membrane potential and mitochondrial viability and by increasing the intracellular Ca2+ [107,109,110,111]. Additionally, acrolein exposure reduced oxygen consumption, mitochondrial complexes (I, II, and V) activities, and ATP content in RPE cells [107,109,110,111]. Acrolein-induced mitochondrial dysfunction is demonstrated in the rat brain [128,129]. Carbonylation of mitochondrial proteins by acrolein has been shown as a mechanism of neuronal death studied using neuron-like PC12 cells [116].

5. Impact of Acrolein on DR Pathogenesis

It is well known that diabetes trigger cellular changes in all component of the retinal neurovascular unit, including neurons, glial cells, and blood vessels [7]. The diabetic retina is characterized by vascular changes, glial activation, and neuronal loss. As illustrated in this review, acrolein is involved in DR pathogenesis and its progressive nature. Possibly, acrolein accumulation might affect each element of the neurovascular retina. The potential mechanisms of acrolein-induced damage in the diabetic retina are presented in Figure 2.

5.1. Inflammation

Inflammation plays a significant role in DR pathogenesis. FDP-lysine accumulation in the diabetic rats was associated with an increase in Müller cells gliosis, upregulation of receptor for advanced glycation end products (RAGE), and calcium-binding protein B (S100B), resulting in the microglial activation and the secretion of inflammatory mediators such as CCL2, IL-1b, and ICAM-1 [78]. Furthermore, in vitro studies demonstrated that the increase in oxidative stress by acrolein exposure increased the protein expression and mRNA level of inflammatory chemokine CXCL1 in rat retinal Müller cell line [112]. The incubation of human Müller cells (MIO-M1) with FDP-lysine human serum albumin increased the mRNA expression of inflammatory molecules, interleukin-6 (IL-6), and tumor necrosis factor-α (TNFα) [99].

5.2. Neurodegeneration

Diabetes deteriorates the visual function, and electroretinogram studies can assess the functional changes. An in vivo analysis illustrated that after 7 weeks of diabetes, rats experienced a reduction in a- and b-waves and increased the summed oscillatory potential [78]. An elevation in the FDP-lysine level accompanied these alterations in diabetes-induced retinal function. These results might indicate a crucial role of acrolein in the diabetes-induced alterations in visual function. Studies have shown that treatment with 2-hydrazino-4,6-dimethylpyrimidine, an acrolein scavenger, and MDL72527 (N, N′-Bis(2,3-butadienyl)-1,4-butanediamine dihydrochloride), a PAO/SMOX inhibitor, improved the visual function in diabetic animals [15,78]. Immunofluorescence evidence from our laboratory has shown an increase in conjugated acrolein levels in the ganglion cell layer and inner nuclear layer in STZ diabetic mice [15]. This increase was associated with a reduction in the retinal function and retinal thinning [15]. These observations support that acrolein is involved in the neurodegenerative process in the diabetic retina. Treatment with acrolein showed a toxic effect on rat retinal ganglion cell line and was more harmful than other aldehydes produced during polyamine metabolism [36].

5.3. Vascular Damage and Blood–Retinal Barrier Integrity

One of the most critical components of the neurovascular unit is the vascular compartment. Dong et al. reported that FDP-lysine is accumulated in endothelial cells and pericytes of PDR patients [130]. The accumulation of FDP-lysine was associated with increased vascular density in patients with PDR [130]. To further confirm the involvement of FDP-lysine in the vascular changes, human retinal microvascular endothelial cells (HRMECs) were treated with a sublethal dose of acrolein, and an increase in cell proliferation and the expression of HO-1 were observed [130]. Incubation with a high dose of acrolein decreased the cell viability of HRMECs [130]. Studies performed by Murata et al. showed that FDP-lysine levels in the endothelial cells of fibrovascular tissues from PDR patients and that treatment of rat retinal capillary endothelial cell line (TR-iBRB2) with acrolein reduced GSH levels in a dose-dependent manner and caused cellular toxicity [13]. RPE cells are involved in the pathogenesis of DR as they maintain the external blood–retinal barrier and produce VEGF and transforming growth factor β (TGFβ). Both VEGF and TGFβ, especially VEGF, plays a vital role in the development of diabetic retinopathy [131,132]. Treatment of ARPE-19 cells with acrolein upregulated VEGF, and TGFβ signaling pathway and these effects further increased when cells were cultured in high glucose medium [61]. Acrolein exposure reduced the viability of RPE cells and mitochondrial membrane potential [109,110,111].

5.4. Müller Glial Dysfunction

Studies using experimental models of DR have shown that FDP-lysine is accumulated in retinal Müller cells of diabetic animals [78,99]. The increase in FDP-lysine was associated with Müller glial dysfunction [78]. The upregulation of FDP-lysine in Müller cells could be because they are the primary polyamine storage in the retina [133]. Cell culture studies showed that small doses of acrolein slightly increased the viability TR-MUL5 cells, while the higher doses of acrolein reduced the viability significantly [112]. Acrolein exposure increased the mRNA and the protein expression of inflammatory chemokine CXCL1 in TR-MUL5 cells [112]. Additionally, the increase in CXCL1 by acrolein treatment induced cell migration of TR-MUL5 cells [112]. An earlier study has shown that incubation of human serum albumin (HSA)-conjugated FDP-lysine increased the apoptosis of MIO-M1 cells [99]. It also upregulated VEGF transcription [99], which plays an important role in the angiogenesis and development of proliferative DR [131]. Based on these observations on the impact of acrolein on the components of the blood–retinal barrier (BRB), it is suggested to play a curial role in regulating vascular permeability and neovascularization.
Figure 3 summarizes the molecular targets of acrolein in the development of DR.

6. Strategies to Reduce Acrolein Toxicity

Current treatments for DR focus on the late stage of the disease and have side-effects. Based on the increasing evidence on acrolein’s implication in DR pathogenesis, strategies to eliminate or reduce its toxicity are of very high importance. The use of agents with antioxidant properties, SMOX/PAO inhibitors, and acrolein-scavenging agents may offer potential intervention strategies. MDL 72527 is an irreversible inhibitor for polyamine oxidases [134]. The inhibition of SMOX/PAO function with MDL 72527 treatment in diabetic mice decreased the presence of conjugated acrolein in the retina [15]. This reduction by MDL 72527 treatment was associated with improved neuronal survival and retinal function in diabetic mice [15].
Several classes of natural products/synthetic compounds have shown to possess potential as acrolein-trapping agents. These include sulfur (thiol)-containing compounds, a group of nitrogen-containing compounds, and the naturally occurring phenolic compounds with acrolein-scavenging properties [135]. Thiol containing acrolein scavengers are compounds such as GSH, l-cysteine, 2-mercaptoethane sulfonate (MESNA), 2,6-Dithiopurine, and lipoic acid. Pretreatment with alpha-lipoic acid (LA), hydroxytyrosol (HTS), and α-tocopherol protected against the reduction in cell viability caused by acrolein in RPE cells [109,110,111]. These compounds also reduced the mitochondrial dysfunction caused by acrolein exposure [109,110,111]. Furthermore, pretreatment with LA or HTS inhibited the acrolein-induced increase in intracellular Ca2+ levels and protected against acrolein-induced oxidative damage by reducing ROS and increasing the cellular antioxidants in RPE cells [109,110,111]. The increased expression of CXCL1 induced by acrolein TR-MUL5 cells was prevented by N-acetylcysteine (NAC) [112]. Many nitrogen (amino)-containing compounds are being used as acrolein scavengers. These include hydralazine, carnosine, aminoguanidine, pyridoxamine, edaravone, and glycyl-histidyl-lysine. A potent acrolein scavenger, 2-hydrazino-4,6-dimethylpyrimidine (2-HDP), significantly decreases the presence of FDP-lysine in the retina of diabetic rats [78]. Moreover, 2-HDP also reduced the activation of microglia and inflammation markers in the retina of diabetic rats. Furthermore, treatment with 2-HDP also attenuated Müller cells gliosis, oxidative stress, distribution of potassium channels, and visual function in the diabetic rats [78]. While acrolein scavengers are still emerging in the field of vision, they are widely investigated in other central nervous system disorders. Treatment with hydralazine, an acrolein scavenger, and an antihypertensive drug demonstrated reduced neuroinflammation and neuropathic pain in a rat model of diabetes [136]. Hydralazine improved behavioral outcome and reduced myelin damage in an experimental model of multiple sclerosis [50,137] and reduced the neural apoptosis, brain edema, and neurological functional deficits in an experimental model of intracerebral hemorrhage [138]. Phenelzine, an FDA-approved monoamine oxidase inhibitor with acrolein-scavenging properties, showed neuroprotection in the experimental models of traumatic brain injury [139,140], multiple sclerosis [141], and spinal cord injury [142]. There are naturally occurring polyphenols with acrolein-scavenging properties. These include epicatechin, cyanomaclurin, theaflavin, phloretin, and phloridzin [135]. However, preclinical studies are yet to be conducted to demonstrate their acrolein-detoxifying properties. Further studies focused on the impact of acrolein-scavenging agents are needed in models of diabetic retinopathy. While defense against oxidative stress is thought to be a major event involved in the inhibition of acrolein’s harmful effects, compounds with both antioxidant activity and acrolein-scavenging capacity should be more impactful.

7. Conclusions and Future Prospects

Diabetic retinopathy is a major complication of diabetes, and the unavailability of effective treatment strategies to reduce DR progression is a major problem. Recent studies have shown an emerging role of acrolein, an unsaturated aldehyde and a mediator of oxidative damage, in DR pathogenesis. Polyamine oxidation and lipid peroxidation are the major pathways of acrolein formation in the diabetic retina. Acrolein elevates oxidative stress and mediates cellular damage and dysfunction by mechanisms such as the depletion of antioxidants, formation of protein carbonyls, and mitochondrial dysfunction. Acrolein is shown to be involved in diabetes-induced alterations in the neurovascular retina, such as neurodegeneration, glial dysfunction, vascular injury, and inflammation.
Further investigations are needed to understand better the specific mechanisms of acrolein-induced changes in the diabetic retina. Pharmacological treatment that neutralizes acrolein or inhibits acrolein formation may serve as promising therapeutic options for DR treatment. A comprehensive understanding of the mechanisms involved in acrolein-induced retinal damage in the diabetic retina may help establish acrolein scavenging as a novel therapeutic intervention for diabetic retinopathy.

Author Contributions

M.A. prepared the initial draft of the review. E.J. helped with drafting and editing. S.P.N. revised and compiled the final version. All authors have read and agreed to the final version of the manuscript.

Funding

This work was supported by a grant from the National Institute of Health (R01EY028569) to SPN and funds from the University of Georgia.

Acknowledgments

The scholarships provided by the Jazan University, Saudi Arabia to M.A. and E.J. are acknowledged.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Cheung, N.; Mitchell, P.; Wong, T.Y. Diabetic retinopathy. Lancet 2010, 376, 124–136. [Google Scholar] [CrossRef]
  2. Sabanayagam, C.; Banu, R.; Chee, M.L.; Lee, R.; Wang, Y.X.; Tan, G.; Jonas, J.B.; Lamoureux, E.L.; Cheng, C.Y.; Klein, B.E.K.; et al. Incidence and progression of diabetic retinopathy: A systematic review. Lancet Diabetes Endocrinol. 2019, 7, 140–149. [Google Scholar] [CrossRef]
  3. Yau, J.W.; Rogers, S.L.; Kawasaki, R.; Lamoureux, E.L.; Kowalski, J.W.; Bek, T.; Chen, S.J.; Dekker, J.M.; Fletcher, A.; Grauslund, J.; et al. Global prevalence and major risk factors of diabetic retinopathy. Diabetes Care 2012, 35, 556–564. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Pereira, D.M.; Shah, A.; D’Souza, M.; Simon, P.; George, T.; D’Souza, N.; Suresh, S.; Baliga, M.S. Quality of Life in People with Diabetic Retinopathy: Indian Study. J. Clin. Diagn. Res. 2017, 11, NC01–NC06. [Google Scholar] [CrossRef] [PubMed]
  5. Klein, R.; Klein, B.E.; Moss, S.E.; Davis, M.D.; DeMets, D.L. The Wisconsin epidemiologic study of diabetic retinopathy. II. Prevalence and risk of diabetic retinopathy when age at diagnosis is less than 30 years. Arch. Ophthalmol. 1984, 102, 520–526. [Google Scholar] [CrossRef]
  6. Klein, R.; Knudtson, M.D.; Lee, K.E.; Gangnon, R.; Klein, B.E. The Wisconsin Epidemiologic Study of Diabetic Retinopathy: XXII the twenty-five-year progression of retinopathy in persons with type 1 diabetes. Ophthalmol. 2008, 115, 1859–1868. [Google Scholar] [CrossRef] [Green Version]
  7. Gardner, T.W.; Davila, J.R. The neurovascular unit and the pathophysiologic basis of diabetic retinopathy. Graefes. Arch. Clin. Exp. Ophthalmol. 2017, 255, 1–6. [Google Scholar] [CrossRef]
  8. Cecilia, O.M.; Jose Alberto, C.G.; Jose, N.P.; Ernesto German, C.M.; Ana Karen, L.C.; Luis Miguel, R.P.; Ricardo Raul, R.R.; Adolfo Daniel, R.C. Oxidative Stress as the Main Target in Diabetic Retinopathy Pathophysiology. J. Diabetes Res. 2019, 2019, 8562408. [Google Scholar] [CrossRef] [Green Version]
  9. Martins, B.; Amorim, M.; Reis, F.; Ambrosio, A.F.; Fernandes, R. Extracellular Vesicles and MicroRNA: Putative Role in Diagnosis and Treatment of Diabetic Retinopathy. Antioxidants 2020, 9, 705. [Google Scholar] [CrossRef]
  10. Narayanan, S.P.; Shosha, E.; Palani, C.D. Spermine oxidase: A promising therapeutic target for neurodegeneration in diabetic retinopathy. Pharmacol. Res. 2019, 147, 104299. [Google Scholar] [CrossRef]
  11. Shosha, E.; Fouda, A.Y.; Narayanan, S.P.; Caldwell, R.W.; Caldwell, R.B. Is the Arginase Pathway a Novel Therapeutic Avenue for Diabetic Retinopathy? J. Clin. Med. 2020, 9, 425. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Wu, M.Y.; Yiang, G.T.; Lai, T.T.; Li, C.J. The Oxidative Stress and Mitochondrial Dysfunction during the Pathogenesis of Diabetic Retinopathy. Oxid. Med. Cell Longev. 2018, 2018, 3420187. [Google Scholar] [CrossRef] [PubMed]
  13. Murata, M.; Noda, K.; Kawasaki, A.; Yoshida, S.; Dong, Y.; Saito, M.; Dong, Z.; Ando, R.; Mori, S.; Saito, W.; et al. Soluble Vascular Adhesion Protein-1 Mediates Spermine Oxidation as Semicarbazide-Sensitive Amine Oxidase: Possible Role in Proliferative Diabetic Retinopathy. Curr. Eye Res. 2017, 42, 1674–1683. [Google Scholar] [CrossRef] [PubMed]
  14. Wu, D.; Noda, K.; Murata, M.; Liu, Y.; Kanda, A.; Ishida, S. Regulation of Spermine Oxidase through Hypoxia-Inducible Factor-1alpha Signaling in Retinal Glial Cells under Hypoxic Conditions. Investig. Ophthalmol. Vis. Sci. 2020, 61, 52. [Google Scholar] [CrossRef] [PubMed]
  15. Liu, F.; Saul, A.B.; Pichavaram, P.; Xu, Z.; Rudraraju, M.; Somanath, P.R.; Smith, S.B.; Caldwell, R.B.; Narayanan, S.P. Pharmacological Inhibition of Spermine Oxidase Reduces Neurodegeneration and Improves Retinal Function in Diabetic Mice. J. Clin. Med. 2020, 9, 340. [Google Scholar] [CrossRef] [Green Version]
  16. Stewart, M.T.; Dunston, T.T.; Woster, P.M.; Casero, R.A., Jr. Polyamine catabolism and oxidative damage. J. Biol. Chem. 2018, 293, 18736–18745. [Google Scholar] [CrossRef] [Green Version]
  17. Agostinelli, E.; Belli, F.; Molinari, A.; Condello, M.; Palmigiani, P.; Vedova, L.D.; Marra, M.; Seiler, N.; Arancia, G. Toxicity of enzymatic oxidation products of spermine to human melanoma cells (M14): Sensitization by heat and MDL 72527. Biochim. Biophys. Acta 2006, 1763, 1040–1050. [Google Scholar] [CrossRef] [Green Version]
  18. Zahedi, K.; Huttinger, F.; Morrison, R.; Murray-Stewart, T.; Casero, R.A.; Strauss, K.I. Polyamine Catabolism Is Enhanced after Traumatic Brain Injury. J. Neurotrauma 2010, 27, 515–525. [Google Scholar] [CrossRef]
  19. Pegg, A.E. Mammalian Polyamine Metabolism and Function. IUBMB Life 2009, 61, 880–894. [Google Scholar] [CrossRef]
  20. Sharmin, S.; Sakata, K.; Kashiwagi, K.; Ueda, S.; Iwasaki, S.; Shirahata, A.; Igarashi, K. Polyamine cytotoxicity in the presence of bovine serum amine oxidase. Biochem. Biophys. Res. Commun. 2001, 282, 228–235. [Google Scholar] [CrossRef] [Green Version]
  21. Moghe, A.; Ghare, S.; Lamoreau, B.; Mohammad, M.; Barve, S.; McClain, C.; Joshi-Barve, S. Molecular mechanisms of acrolein toxicity: Relevance to human disease. Toxicol. Sci. 2015, 143, 242–255. [Google Scholar] [CrossRef] [PubMed]
  22. Ambaw, A.; Zheng, L.; Tambe, M.A.; Strathearn, K.E.; Acosta, G.; Hubers, S.A.; Liu, F.; Herr, S.A.; Tang, J.; Truong, A.; et al. Acrolein-mediated neuronal cell death and alpha-synuclein aggregation: Implications for Parkinson’s disease. Mol. Cell Neurosci. 2018, 88, 70–82. [Google Scholar] [CrossRef] [PubMed]
  23. Park, Y.S.; Taniguchi, N. Acrolein induces inflammatory response underlying endothelial dysfunction: A risk factor for atherosclerosis. Ann. N. Y. Acad. Sci. 2008, 1126, 185–189. [Google Scholar] [CrossRef] [PubMed]
  24. DeJarnett, N.; Conklin, D.J.; Riggs, D.W.; Myers, J.A.; O’Toole, T.E.; Hamzeh, I.; Wagner, S.; Chugh, A.; Ramos, K.S.; Srivastava, S.; et al. Acrolein exposure is associated with increased cardiovascular disease risk. J. Am. Heart Assoc. 2014, 3. [Google Scholar] [CrossRef] [Green Version]
  25. Sun, Y.; Ito, S.; Nishio, N.; Tanaka, Y.; Chen, N.; Isobe, K. Acrolein induced both pulmonary inflammation and the death of lung epithelial cells. Toxicol. Lett. 2014, 229, 384–392. [Google Scholar] [CrossRef]
  26. Sindhu, K.K. Uremic toxins: Some thoughts on acrolein and spermine. Ren. Fail. 2016, 38, 1755–1758. [Google Scholar] [CrossRef]
  27. Dang, T.N.; Arseneault, M.; Murthy, V.; Ramassamy, C. Potential role of acrolein in neurodegeneration and in Alzheimer’s disease. Curr. Mol. Pharmacol. 2010, 3, 66–78. [Google Scholar]
  28. Tully, M.; Shi, R. New insights in the pathogenesis of multiple sclerosis--role of acrolein in neuronal and myelin damage. Int. J. Mol. Sci. 2013, 14, 20037–20047. [Google Scholar] [CrossRef] [Green Version]
  29. Uemura, T.; Watanabe, K.; Ishibashi, M.; Saiki, R.; Kuni, K.; Nishimura, K.; Toida, T.; Kashiwagi, K.; Igarashi, K. Aggravation of brain infarction through an increase in acrolein production and a decrease in glutathione with aging. Biochem. Biophys. Res. Commun. 2016, 473, 630–635. [Google Scholar] [CrossRef]
  30. Saiki, R.; Park, H.; Ishii, I.; Yoshida, M.; Nishimura, K.; Toida, T.; Tatsukawa, H.; Kojima, S.; Ikeguchi, Y.; Pegg, A.E.; et al. Brain infarction correlates more closely with acrolein than with reactive oxygen species. Biochem. Biophys. Res. Commun. 2011, 404, 1044–1049. [Google Scholar] [CrossRef]
  31. Saiki, R.; Nishimura, K.; Ishii, I.; Omura, T.; Okuyama, S.; Kashiwagi, K.; Igarashi, K. Intense correlation between brain infarction and protein-conjugated acrolein. Stroke 2009, 40, 3356–3361. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Igarashi, K.; Uemura, T.; Kashiwagi, K. Acrolein toxicity at advanced age: Present and future. Amino Acids 2018, 50, 217–228. [Google Scholar] [CrossRef] [PubMed]
  33. Luo, J.; Uchida, K.; Shi, R. Accumulation of acrolein-protein adducts after traumatic spinal cord injury. Neurochem. Res. 2005, 30, 291–295. [Google Scholar] [CrossRef] [PubMed]
  34. Khoramjouy, M.; Naderi, N.; Kobarfard, F.; Heidarli, E.; Faizi, M. An Intensified Acrolein Exposure Can Affect Memory and Cognition in Rat. Neurotox. Res. 2020. [Google Scholar] [CrossRef]
  35. Yoshida, M.; Tomitori, H.; Machi, Y.; Hagihara, M.; Higashi, K.; Goda, H.; Ohya, T.; Niitsu, M.; Kashiwagi, K.; Igarashi, K. Acrolein toxicity: Comparison with reactive oxygen species. Biochem. Biophys. Res. Commun. 2009, 378, 313–318. [Google Scholar] [CrossRef]
  36. Wood, P.L.; Khan, M.A.; Moskal, J.R. The concept of “aldehyde load” in neurodegenerative mechanisms: Cytotoxicity of the polyamine degradation products hydrogen peroxide, acrolein, 3-aminopropanal, 3-acetamidopropanal and 4-aminobutanal in a retinal ganglion cell line. Brain Res. 2007, 1145, 150–156. [Google Scholar] [CrossRef]
  37. Luo, J.; Shi, R. Acrolein induces axolemmal disruption, oxidative stress, and mitochondrial impairment in spinal cord tissue. Neurochem. Int. 2004, 44, 475–486. [Google Scholar] [CrossRef]
  38. Shi, R.; Rickett, T.; Sun, W. Acrolein-mediated injury in nervous system trauma and diseases. Mol. Nutr. Food Res. 2011, 55, 1320–1331. [Google Scholar] [CrossRef]
  39. Stevens, J.F.; Maier, C.S. Acrolein: Sources, metabolism, and biomolecular interactions relevant to human health and disease. Mol. Nutr. Food Res. 2008, 52, 7–25. [Google Scholar] [CrossRef] [Green Version]
  40. Abraham, K.; Andres, S.; Palavinskas, R.; Berg, K.; Appel, K.E.; Lampen, A. Toxicology and risk assessment of acrolein in food. Mol. Nutr. Food Res. 2011, 55, 1277–1290. [Google Scholar] [CrossRef]
  41. Uchida, K.; Kanematsu, M.; Morimitsu, Y.; Osawa, T.; Noguchi, N.; Niki, E. Acrolein is a product of lipid peroxidation reaction. Formation of free acrolein and its conjugate with lysine residues in oxidized low density lipoproteins. J. Biol. Chem. 1998, 273, 16058–16066. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Negre-Salvayre, A.; Coatrieux, C.; Ingueneau, C.; Salvayre, R. Advanced lipid peroxidation end products in oxidative damage to proteins. Potential role in diseases and therapeutic prospects for the inhibitors. Br. J. Pharmacol. 2008, 153, 6–20. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Gallelli, C.A.; Calcagnini, S.; Romano, A.; Koczwara, J.B.; de Ceglia, M.; Dante, D.; Villani, R.; Giudetti, A.M.; Cassano, T.; Gaetani, S. Modulation of the Oxidative Stress and Lipid Peroxidation by Endocannabinoids and Their Lipid Analogues. Antioxidants 2018, 7, 93. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Sousa, B.C.; Pitt, A.R.; Spickett, C.M. Chemistry and analysis of HNE and other prominent carbonyl-containing lipid oxidation compounds. Free Radic. Biol. Med. 2017, 111, 294–308. [Google Scholar] [CrossRef] [PubMed]
  45. O’Brien, P.J.; Siraki, A.G.; Shangari, N. Aldehyde sources, metabolism, molecular toxicity mechanisms, and possible effects on human health. Crit. Rev. Toxicol. 2005, 35, 609–662. [Google Scholar] [CrossRef] [PubMed]
  46. Casero, R.A.; Pegg, A.E. Polyamine catabolism and disease. Biochem. J. 2009, 421, 323–338. [Google Scholar] [CrossRef] [Green Version]
  47. Uemura, T.; Nakamura, M.; Sakamoto, A.; Suzuki, T.; Dohmae, N.; Terui, Y.; Tomitori, H.; Casero, R.A., Jr.; Kashiwagi, K.; Igarashi, K. Decrease in acrolein toxicity based on the decline of polyamine oxidases. Int. J. Biochem. Cell Biol. 2016, 79, 151–157. [Google Scholar] [CrossRef]
  48. Burcham, P.C. Acrolein and Human Disease: Untangling the Knotty Exposure Scenarios Accompanying Several Diverse Disorders. Chem. Res. Toxicol. 2017, 30, 145–161. [Google Scholar] [CrossRef]
  49. Carmella, S.G.; Chen, M.; Zhang, Y.; Zhang, S.; Hatsukami, D.K.; Hecht, S.S. Quantitation of acrolein-derived (3-hydroxypropyl)mercapturic acid in human urine by liquid chromatography-atmospheric pressure chemical ionization tandem mass spectrometry: Effects of cigarette smoking. Chem. Res. Toxicol. 2007, 20, 986–990. [Google Scholar] [CrossRef] [Green Version]
  50. Tully, M.; Tang, J.; Zheng, L.; Acosta, G.; Tian, R.; Hayward, L.; Race, N.; Mattson, D.; Shi, R. Systemic Acrolein Elevations in Mice with Experimental Autoimmune Encephalomyelitis and Patients With Multiple Sclerosis. Front. Neurol. 2018, 9, 420. [Google Scholar] [CrossRef]
  51. Wang, T.W.; Liu, J.H.; Tsou, H.H.; Liu, T.Y.; Wang, H.T. Identification of acrolein metabolites in human buccal cells, blood, and urine after consumption of commercial fried food. Food Sci. Nutr. 2019, 7, 1668–1676. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Vatsalya, V.; Kong, M.; Gobejishvili, L.; Chen, W.Y.; Srivastava, S.; Barve, S.; McClain, C.; Joshi-Barve, S. Urinary acrolein metabolite levels in severe acute alcoholic hepatitis patients. Am. J. Physiol. Gastrointest. Liver Physiol. 2019, 316, G115–G122. [Google Scholar] [CrossRef] [PubMed]
  53. Igarashi, K.; Yoshida, M.; Waragai, M.; Kashiwagi, K. Evaluation of dementia by acrolein, amyloid-beta and creatinine. Clin. Chim. Acta 2015, 450, 56–63. [Google Scholar] [CrossRef] [PubMed]
  54. Yoshida, M.; Tomitori, H.; Machi, Y.; Katagiri, D.; Ueda, S.; Horiguchi, K.; Kobayashi, E.; Saeki, N.; Nishimura, K.; Ishii, I.; et al. Acrolein, IL-6 and CRP as markers of silent brain infarction. Atherosclerosis 2009, 203, 557–562. [Google Scholar] [CrossRef] [PubMed]
  55. Tomitori, H.; Usui, T.; Saeki, N.; Ueda, S.; Kase, H.; Nishimura, K.; Kashiwagi, K.; Igarashi, K. Polyamine oxidase and acrolein as novel biochemical markers for diagnosis of cerebral stroke. Stroke 2005, 36, 2609–2613. [Google Scholar] [CrossRef]
  56. Takamatsu, M.; Fukase, K.; Oka, R.; Kitazume, S.; Taniguchi, N.; Tanaka, K. A Reduction-Based Sensor for Acrolein Conjugates with the Inexpensive Nitrobenzene as an Alternative to Monoclonal Antibody. Sci. Rep. 2016, 6, 35872. [Google Scholar] [CrossRef] [PubMed]
  57. McDowell, R.E.; McGahon, M.K.; Augustine, J.; Chen, M.; McGeown, J.G.; Curtis, T.M. Diabetes Impairs the Aldehyde Detoxifying Capacity of the Retina. Investig. Ophthalmol. Vis. Sci. 2016, 57, 4762–4771. [Google Scholar] [CrossRef] [Green Version]
  58. Tsukahara, H.; Sekine, K.; Uchiyama, M.; Kawakami, H.; Hata, I.; Todoroki, Y.; Hiraoka, M.; Kaji, M.; Yorifuji, T.; Momoi, T.; et al. Formation of advanced glycosylation end products and oxidative stress in young patients with type 1 diabetes. Pediatr. Res. 2003, 54, 419–424. [Google Scholar] [CrossRef] [Green Version]
  59. Daimon, M.; Sugiyama, K.; Kameda, W.; Saitoh, T.; Oizumi, T.; Hirata, A.; Yamaguchi, H.; Ohnuma, H.; Igarashi, M.; Kato, T. Increased urinary levels of pentosidine, pyrraline and acrolein adduct in type 2 diabetes. Endocr. J. 2003, 50, 61–67. [Google Scholar] [CrossRef] [Green Version]
  60. Suzuki, D.; Miyata, T. Carbonyl stress in the pathogenesis of diabetic nephropathy. Intern. Med. 1999, 38, 309–314. [Google Scholar] [CrossRef] [Green Version]
  61. Grigsby, J.; Betts, B.; Vidro-Kotchan, E.; Culbert, R.; Tsin, A. A possible role of acrolein in diabetic retinopathy: Involvement of a VEGF/TGFbeta signaling pathway of the retinal pigment epithelium in hyperglycemia. Curr. Eye Res. 2012, 37, 1045–1053. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Conklin, D.J.; Barski, O.A.; Lesgards, J.F.; Juvan, P.; Rezen, T.; Rozman, D.; Prough, R.A.; Vladykovskaya, E.; Liu, S.; Srivastava, S.; et al. Acrolein consumption induces systemic dyslipidemia and lipoprotein modification. Toxicol. Appl. Pharmacol. 2010, 243, 1–12. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Conklin, D.J.; Prough, R.A.; Juvan, P.; Rezen, T.; Rozman, D.; Haberzettl, P.; Srivastava, S.; Bhatnagar, A. Acrolein-induced dyslipidemia and acute-phase response are independent of HMG-CoA reductase. Mol. Nutr. Food Res. 2011, 55, 1411–1422. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. De Souza Bastos, A.; Graves, D.T.; de Melo Loureiro, A.P.; Junior, C.R.; Corbi, S.C.T.; Frizzera, F.; Scarel-Caminaga, R.M.; Camara, N.O.; Andriankaja, O.M.; Hiyane, M.I.; et al. Diabetes and increased lipid peroxidation are associated with systemic inflammation even in well-controlled patients. J. Diabetes Complicat. 2016, 30, 1593–1599. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Bjelakovic, G.; Beninati, S.; Bjelakovic, B.; Sokolovic, D.; Jevtovic, T.; Stojanovic, I.; Rossi, S.; Tabolacci, C.; Kocic, G.; Pavlovic, D.; et al. Does polyamine oxidase activity influence the oxidative metabolism of children who suffer of diabetes mellitus? Mol. Cell Biochem. 2010, 341, 79–85. [Google Scholar] [CrossRef] [PubMed]
  66. Uemura, T.; Akasaka, Y.; Ikegaya, H. Correlation of polyamines, acrolein-conjugated lysine and polyamine metabolic enzyme levels with age in human liver. Heliyon 2020, 6, e05031. [Google Scholar] [CrossRef] [PubMed]
  67. Harrison, W.W.; Bearse, M.A., Jr.; Ng, J.S.; Jewell, N.P.; Barez, S.; Burger, D.; Schneck, M.E.; Adams, A.J. Multifocal electroretinograms predict onset of diabetic retinopathy in adult patients with diabetes. Investig. Ophthalmol. Vis. Sci. 2011, 52, 772–777. [Google Scholar] [CrossRef] [Green Version]
  68. Kern, T.S.; Barber, A.J. Retinal ganglion cells in diabetes. J. Physiol. 2008, 586, 4401–4408. [Google Scholar] [CrossRef]
  69. Van Dijk, H.W.; Verbraak, F.D.; Stehouwer, M.; Kok, P.H.; Garvin, M.K.; Sonka, M.; DeVries, J.H.; Schlingemann, R.O.; Abramoff, M.D. Association of visual function and ganglion cell layer thickness in patients with diabetes mellitus type 1 and no or minimal diabetic retinopathy. Vision Res. 2011, 51, 224–228. [Google Scholar] [CrossRef] [Green Version]
  70. Ziccardi, L.; Parisi, V.; Picconi, F.; Di Renzo, A.; Lombardo, M.; Frontoni, S.; Parravano, M. Early and localized retinal dysfunction in patients with type 1 diabetes mellitus studied by multifocal electroretinogram. Acta Diabetol. 2018, 55, 1191–1200. [Google Scholar] [CrossRef]
  71. Lieth, E.; Gardner, T.W.; Barber, A.J.; Antonetti, D.A.; Penn State Retina Research Group. Retinal neurodegeneration: Early pathology in diabetes. Clin. Exp. Ophthalmol. 2000, 28, 3–8. [Google Scholar] [CrossRef]
  72. Barcelona, P.F.; Sitaras, N.; Galan, A.; Esquiva, G.; Jmaeff, S.; Jian, Y.; Sarunic, M.V.; Cuenca, N.; Sapieha, P.; Saragovi, H.U. p75NTR and Its Ligand ProNGF Activate Paracrine Mechanisms Etiological to the Vascular, Inflammatory, and Neurodegenerative Pathologies of Diabetic Retinopathy. J. Neurosci. 2016, 36, 8826–8841. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Caldwell, R.B.; Zhang, W.; Romero, M.J.; Caldwell, R.W. Vascular dysfunction in retinopathy-an emerging role for arginase. Brain Res. Bull. 2010, 81, 303–309. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Zhang, W.; Liu, H.; Rojas, M.; Caldwell, R.W.; Caldwell, R.B. Anti-inflammatory therapy for diabetic retinopathy. Immunotherapy 2011, 3, 609–628. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Hachana, S.; Pouliot, M.; Couture, R.; Vaucher, E. Diabetes-Induced Inflammation and Vascular Alterations in the Goto-Kakizaki Rat Retina. Curr. Eye Res. 2020, 45, 965–974. [Google Scholar] [CrossRef]
  76. Al-Shabrawey, M.; Rojas, M.; Sanders, T.; Behzadian, A.; El-Remessy, A.; Bartoli, M.; Parpia, A.K.; Liou, G.; Caldwell, R.B. Role of NADPH oxidase in retinal vascular inflammation. Investig. Ophthalmol. Vis. Sci. 2008, 49, 3239–3244. [Google Scholar] [CrossRef]
  77. Suzumura, A.; Kaneko, H.; Funahashi, Y.; Takayama, K.; Nagaya, M.; Ito, S.; Okuno, T.; Hirakata, T.; Nonobe, N.; Kataoka, K.; et al. n-3 Fatty Acid and Its Metabolite 18-HEPE Ameliorate Retinal Neuronal Cell Dysfunction by Enhancing Muller BDNF in Diabetic Retinopathy. Diabetes 2020, 69, 724–735. [Google Scholar] [CrossRef]
  78. McDowell, R.E.; Barabas, P.; Augustine, J.; Chevallier, O.; McCarron, P.; Chen, M.; McGeown, J.G.; Curtis, T.M. Muller glial dysfunction during diabetic retinopathy in rats is reduced by the acrolein-scavenging drug, 2-hydrazino-4,6-dimethylpyrimidine. Diabetologia 2018, 61, 2654–2667. [Google Scholar] [CrossRef] [Green Version]
  79. Bringmann, A.; Pannicke, T.; Grosche, J.; Francke, M.; Wiedemann, P.; Skatchkov, S.N.; Osborne, N.N.; Reichenbach, A. Muller cells in the healthy and diseased retina. Prog. Retin. Eye Res. 2006, 25, 397–424. [Google Scholar] [CrossRef]
  80. Coughlin, B.A.; Feenstra, D.J.; Mohr, S. Muller cells and diabetic retinopathy. Vis. Res. 2017, 139, 93–100. [Google Scholar] [CrossRef]
  81. Chakravarthy, H.; Devanathan, V. Molecular Mechanisms Mediating Diabetic Retinal Neurodegeneration: Potential Research Avenues and Therapeutic Targets. J. Mol. Neurosci. 2018, 66, 445–461. [Google Scholar] [CrossRef] [PubMed]
  82. Zhang, J.R.; Sun, H.J. Roles of circular RNAs in diabetic complications: From molecular mechanisms to therapeutic potential. Gene 2020, 763, 145066. [Google Scholar] [CrossRef] [PubMed]
  83. Narayanan, S.P.; Xu, Z.; Putluri, N.; Sreekumar, A.; Lemtalsi, T.; Caldwell, R.W.; Caldwell, R.B. Arginase 2 deficiency reduces hyperoxia-mediated retinal neurodegeneration through the regulation of polyamine metabolism. Cell Death Dis. 2014, 5, e1075. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Patel, C.; Xu, Z.; Shosha, E.; Xing, J.; Lucas, R.; Caldwell, R.W.; Caldwell, R.B.; Narayanan, S.P. Treatment with polyamine oxidase inhibitor reduces microglial activation and limits vascular injury in ischemic retinopathy. Biochim. Biophys. Acta 2016, 1862, 1628–1639. [Google Scholar] [CrossRef]
  85. Seghieri, G.; Gironi, A.; Niccolai, M.; Mammini, P.; Alviggi, L.; De Giorgio, L.A.; Caselli, P.; Bartolomei, G. Serum spermidine oxidase activity in patients with insulin-dependent diabetes mellitus and microvascular complications. Acta. Diabetol. Lat. 1990, 27, 303–308. [Google Scholar] [CrossRef]
  86. Nicoletti, R.; Venza, I.; Ceci, G.; Visalli, M.; Teti, D.; Reibaldi, A. Vitreous polyamines spermidine, putrescine, and spermine in human proliferative disorders of the retina. Br. J. Ophthalmol. 2003, 87, 1038–1042. [Google Scholar] [CrossRef] [Green Version]
  87. Matsushita, K.; Fukumoto, M.; Kobayashi, T.; Kobayashi, M.; Ishizaki, E.; Minami, M.; Katsumura, K.; Liao, S.D.; Wu, D.M.; Zhang, T.; et al. Diabetes-induced inhibition of voltage-dependent calcium channels in the retinal microvasculature: Role of spermine. Investig. Ophthalmol. Vis. Sci. 2010, 51, 5979–5990. [Google Scholar] [CrossRef] [Green Version]
  88. Kesavulu, M.M.; Giri, R.; Kameswara Rao, B.; Apparao, C. Lipid peroxidation and antioxidant enzyme levels in type 2 diabetics with microvascular complications. Diabetes Metab. 2000, 26, 387–392. [Google Scholar]
  89. Kedziora-Kornatowska, K.Z.; Luciak, M.; Blaszczyk, J.; Pawlak, W. Lipid peroxidation and activities of antioxidant enzymes in erythrocytes of patients with non-insulin dependent diabetes with or without diabetic nephropathy. Nephrol. Dial. Transpl. 1998, 13, 2829–2832. [Google Scholar] [CrossRef] [Green Version]
  90. Al-Shabrawey, M.; Bartoli, M.; El-Remessy, A.B.; Ma, G.; Matragoon, S.; Lemtalsi, T.; Caldwell, R.W.; Caldwell, R.B. Role of NADPH oxidase and Stat3 in statin-mediated protection against diabetic retinopathy. Investig. Ophthalmol. Vis. Sci. 2008, 49, 3231–3238. [Google Scholar] [CrossRef]
  91. Al-Shabrawey, M.; Bartoli, M.; El-Remessy, A.B.; Platt, D.H.; Matragoon, S.; Behzadian, M.A.; Caldwell, R.W.; Caldwell, R.B. Inhibition of NAD(P)H oxidase activity blocks vascular endothelial growth factor overexpression and neovascularization during ischemic retinopathy. Am. J. Pathol. 2005, 167, 599–607. [Google Scholar] [CrossRef] [Green Version]
  92. Obrosova, I.G.; Fathallah, L.; Greene, D.A. Early changes in lipid peroxidation and antioxidative defense in diabetic rat retina: Effect of DL-alpha-lipoic acid. Eur. J. Pharmacol. 2000, 398, 139–146. [Google Scholar] [CrossRef]
  93. Curtis, T.M.; Hamilton, R.; Yong, P.H.; McVicar, C.M.; Berner, A.; Pringle, R.; Uchida, K.; Nagai, R.; Brockbank, S.; Stitt, A.W. Muller glial dysfunction during diabetic retinopathy in rats is linked to accumulation of advanced glycation end-products and advanced lipoxidation end-products. Diabetologia 2011, 54, 690–698. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Pan, H.Z.; Zhang, H.; Chang, D.; Li, H.; Sui, H. The change of oxidative stress products in diabetes mellitus and diabetic retinopathy. Br. J. Ophthalmol. 2008, 92, 548–551. [Google Scholar] [CrossRef]
  95. Pizzimenti, S.; Ciamporcero, E.; Daga, M.; Pettazzoni, P.; Arcaro, A.; Cetrangolo, G.; Minelli, R.; Dianzani, C.; Lepore, A.; Gentile, F.; et al. Interaction of aldehydes derived from lipid peroxidation and membrane proteins. Front Physiol. 2013, 4, 242. [Google Scholar] [CrossRef] [Green Version]
  96. Zarkovic, N.; Cipak, A.; Jaganjac, M.; Borovic, S.; Zarkovic, K. Pathophysiological relevance of aldehydic protein modifications. J. Proteomics 2013, 92, 239–247. [Google Scholar] [CrossRef]
  97. Zhang, X.; Lai, Y.; McCance, D.R.; Uchida, K.; McDonald, D.M.; Gardiner, T.A.; Stitt, A.W.; Curtis, T.M. Evaluation of N (epsilon)-(3-formyl-3,4-dehydropiperidino)lysine as a novel biomarker for the severity of diabetic retinopathy. Diabetologia 2008, 51, 1723–1730. [Google Scholar] [CrossRef] [Green Version]
  98. Noda, K.; Murata, M.; Saito, W.; Kanda, A.; Ishida, S. Accumulation of Acrolein-conjugated Protein in the Vitreous Fluid of Proliferative Diabetic Retinopathy. Investig. Ophthalmol. Vis. Sci. 2017, 58, 2514. [Google Scholar]
  99. Yong, P.H.; Zong, H.; Medina, R.J.; Limb, G.A.; Uchida, K.; Stitt, A.W.; Curtis, T.M. Evidence supporting a role for N-(3-formyl-3,4-dehydropiperidino)lysine accumulation in Muller glia dysfunction and death in diabetic retinopathy. Mol. Vis. 2010, 16, 2524–2538. [Google Scholar]
  100. Huang, J.; Li, X.; Li, M.; Li, J.; Xiao, W.; Ma, W.; Chen, X.; Liang, X.; Tang, S.; Luo, Y. Mitochondria-targeted antioxidant peptide SS31 protects the retinas of diabetic rats. Curr. Mol. Med. 2013, 13, 935–945. [Google Scholar] [CrossRef]
  101. Yeh, P.T.; Wang, L.C.; Chang, S.W.; Yang, W.S.; Yang, C.M.; Yang, C.H. Effect of Fenofibrate on the Expression of Inflammatory Mediators in a Diabetic Rat Model. Curr. Eye Res. 2019, 44, 1121–1132. [Google Scholar] [CrossRef] [PubMed]
  102. Schaefer, S.; Kajimura, M.; Tsuyama, S.; Uchida, K.; Sato, E.; Inoue, M.; Suematsu, M.; Watanabe, K. Aberrant utilization of nitric oxide and regulation of soluble guanylate cyclase in rat diabetic retinopathy. Antioxid. Redox Signal. 2003, 5, 457–465. [Google Scholar] [CrossRef] [PubMed]
  103. Ahmed Laskar, A.; Younus, H. Aldehyde toxicity and metabolism: The role of aldehyde dehydrogenases in detoxification, drug resistance and carcinogenesis. Drug Metab. Rev. 2019, 51, 42–64. [Google Scholar] [CrossRef] [PubMed]
  104. Yoval-Sanchez, B.; Rodriguez-Zavala, J.S. Differences in susceptibility to inactivation of human aldehyde dehydrogenases by lipid peroxidation byproducts. Chem. Res. Toxicol. 2012, 25, 722–729. [Google Scholar] [CrossRef]
  105. Makia, N.L.; Bojang, P.; Falkner, K.C.; Conklin, D.J.; Prough, R.A. Murine hepatic aldehyde dehydrogenase 1a1 is a major contributor to oxidation of aldehydes formed by lipid peroxidation. Chem. Biol. Interact. 2011, 191, 278–287. [Google Scholar] [CrossRef] [Green Version]
  106. Wang, Y.; Tao, J.; Jiang, M.; Yao, Y. Apocynin ameliorates diabetic retinopathy in rats: Involvement of TLR4/NF-kappaB signaling pathway. Int. Immunopharmacol. 2019, 73, 49–56. [Google Scholar] [CrossRef]
  107. Li, Y.; Zou, X.; Cao, K.; Xu, J.; Yue, T.; Dai, F.; Zhou, B.; Lu, W.; Feng, Z.; Liu, J. Curcumin analog 1, 5-bis (2-trifluoromethylphenyl)-1, 4-pentadien-3-one exhibits enhanced ability on Nrf2 activation and protection against acrolein-induced ARPE-19 cell toxicity. Toxicol. Appl. Pharmacol. 2013, 272, 726–735. [Google Scholar] [CrossRef]
  108. Kehrer, J.P.; Biswal, S.S. The molecular effects of acrolein. Toxicol. Sci. 2000, 57, 6–15. [Google Scholar] [CrossRef]
  109. Jia, L.; Liu, Z.; Sun, L.; Miller, S.S.; Ames, B.N.; Cotman, C.W.; Liu, J. Acrolein, a Toxicant in Cigarette Smoke, Causes Oxidative Damage and Mitochondrial Dysfunction in RPE Cells: Protection by (R)-α-Lipoic Acid. Investig. Ophthalmol. Vis. Sci. 2007, 48, 339. [Google Scholar] [CrossRef] [Green Version]
  110. Liu, Z.; Sun, L.; Zhu, L.; Jia, X.; Li, X.; Jia, H.; Wang, Y.; Weber, P.; Long, J.; Liu, J. Hydroxytyrosol protects retinal pigment epithelial cells from acrolein-induced oxidative stress and mitochondrial dysfunction. J. Neurochem. 2007, 103, 2690–2700. [Google Scholar] [CrossRef] [Green Version]
  111. Feng, Z.; Liu, Z.; Li, X.; Jia, H.; Sun, L.; Tian, C.; Jia, L.; Liu, J. alpha-Tocopherol is an effective Phase II enzyme inducer: Protective effects on acrolein-induced oxidative stress and mitochondrial dysfunction in human retinal pigment epithelial cells. J. Nutr. Biochem. 2010, 21, 1222–1231. [Google Scholar] [CrossRef] [PubMed]
  112. Murata, M.; Noda, K.; Yoshida, S.; Saito, M.; Fujiya, A.; Kanda, A.; Ishida, S. Unsaturated Aldehyde Acrolein Promotes Retinal Glial Cell Migration. Investig. Ophthalmol. Vis. Sci. 2019, 60, 4425–4435. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  113. Furuhata, A.; Nakamura, M.; Osawa, T.; Uchida, K. Thiolation of protein-bound carcinogenic aldehyde. An electrophilic acrolein-lysine adduct that covalently binds to thiols. J. Biol. Chem. 2002, 277, 27919–27926. [Google Scholar] [CrossRef] [Green Version]
  114. Qin, W.S.; Deng, Y.H.; Cui, F.C. Sulforaphane protects against acrolein-induced oxidative stress and inflammatory responses: Modulation of Nrf-2 and COX-2 expression. Arch. Med. Sci. 2016, 12, 871–880. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  115. Grochot-Przeczek, A.; Dulak, J.; Jozkowicz, A. Heme oxygenase-1 in neovascularisation: A diabetic perspective. Thromb. Haemost. 2010, 104, 424–431. [Google Scholar] [CrossRef]
  116. Dasgupta, A.; Zheng, J.; Bizzozero, O.A. Protein carbonylation and aggregation precede neuronal apoptosis induced by partial glutathione depletion. ASN Neuro. 2012, 4. [Google Scholar] [CrossRef] [Green Version]
  117. Galligan, J.J.; Smathers, R.L.; Fritz, K.S.; Epperson, L.E.; Hunter, L.E.; Petersen, D.R. Protein carbonylation in a murine model for early alcoholic liver disease. Chem. Res. Toxicol. 2012, 25, 1012–1021. [Google Scholar] [CrossRef] [Green Version]
  118. Gehl, Z.; Bakondi, E.; Resch, M.D.; Hegedus, C.; Kovacs, K.; Lakatos, P.; Szabo, A.; Nagy, Z.; Virag, L. Diabetes-induced oxidative stress in the vitreous humor. Redox Biol. 2016, 9, 100–103. [Google Scholar] [CrossRef] [Green Version]
  119. Loukovaara, S.; Koivunen, P.; Ingles, M.; Escobar, J.; Vento, M.; Andersson, S. Elevated protein carbonyl and HIF-1alpha levels in eyes with proliferative diabetic retinopathy. Acta Ophthalmol. 2014, 92, 323–327. [Google Scholar] [CrossRef]
  120. Mohammad, G.; Alam, K.; Nawaz, M.I.; Siddiquei, M.M.; Mousa, A.; Abu El-Asrar, A.M. Mutual enhancement between high-mobility group box-1 and NADPH oxidase-derived reactive oxygen species mediates diabetes-induced upregulation of retinal apoptotic markers. J. Physiol. Biochem. 2015, 71, 359–372. [Google Scholar] [CrossRef]
  121. Liu, W.Y.; Liou, S.S.; Hong, T.Y.; Liu, I.M. Protective Effects of Hesperidin (Citrus Flavonone) on High Glucose Induced Oxidative Stress and Apoptosis in a Cellular Model for Diabetic Retinopathy. Nutrients 2017, 9, 1312. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  122. Kumar, M.P.; Mamidala, E.; Al-Ghanim, K.A.; Al-Misned, F.; Mahboob, S. Evaluation of the andrographolides role and its indoleamine 2,3-dioxygenase inhibitory potential and attendant molecular mechanism against STZ-induced diabetic rats. Saudi J. Biol. Sci. 2020, 27, 713–719. [Google Scholar] [CrossRef] [PubMed]
  123. Song, H.; Vijayasarathy, C.; Zeng, Y.; Marangoni, D.; Bush, R.A.; Wu, Z.; Sieving, P.A. NADPH Oxidase Contributes to Photoreceptor Degeneration in Constitutively Active RAC1 Mice. Investig. Ophthalmol. Vis. Sci. 2016, 57, 2864–2875. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Yang, X.; Hondur, G.; Tezel, G. Antioxidant Treatment Limits Neuroinflammation in Experimental Glaucoma. Investig. Ophthalmol. Vis. Sci. 2016, 57, 2344–2354. [Google Scholar] [CrossRef] [Green Version]
  125. Kowluru, R.A.; Mishra, M. Oxidative stress, mitochondrial damage and diabetic retinopathy. Biochim. Biophys. Acta 2015, 1852, 2474–2483. [Google Scholar] [CrossRef] [Green Version]
  126. Dan Dunn, J.; Alvarez, L.A.; Zhang, X.; Soldati, T. Reactive oxygen species and mitochondria: A nexus of cellular homeostasis. Redox. Biol. 2015, 6, 472–485. [Google Scholar] [CrossRef]
  127. Sun, L.; Luo, C.; Long, J.; Wei, D.; Liu, J. Acrolein is a mitochondrial toxin: Effects on respiratory function and enzyme activities in isolated rat liver mitochondria. Mitochondrion 2006, 6, 136–142. [Google Scholar] [CrossRef]
  128. Picklo, M.J.; Montine, T.J. Acrolein inhibits respiration in isolated brain mitochondria. Biochim. Biophys. Acta 2001, 1535, 145–152. [Google Scholar] [CrossRef] [Green Version]
  129. Luo, J.; Shi, R. Acrolein induces oxidative stress in brain mitochondria. Neurochem. Int. 2005, 46, 243–252. [Google Scholar] [CrossRef]
  130. Dong, Y.; Noda, K.; Murata, M.; Yoshida, S.; Saito, W.; Kanda, A.; Ishida, S. Localization of Acrolein-Lysine Adduct in Fibrovascular Tissues of Proliferative Diabetic Retinopathy. Curr. Eye Res. 2017, 42, 111–117. [Google Scholar] [CrossRef]
  131. Gupta, N.; Mansoor, S.; Sharma, A.; Sapkal, A.; Sheth, J.; Falatoonzadeh, P.; Kuppermann, B.; Kenney, M. Diabetic retinopathy and VEGF. Open Ophthalmol. J. 2013, 7, 4–10. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  132. Wheeler, S.E.; Lee, N.Y. Emerging Roles of Transforming Growth Factor beta Signaling in Diabetic Retinopathy. J. Cell Physiol. 2017, 232, 486–489. [Google Scholar] [CrossRef] [PubMed]
  133. Biedermann, B.; Skatchkov, S.N.; Brunk, I.; Bringmann, A.; Pannicke, T.; Bernstein, H.G.; Faude, F.; Germer, A.; Veh, R.; Reichenbach, A. Spermine/spermidine is expressed by retinal glial (Muller) cells and controls distinct K+ channels of their membrane. Glia 1998, 23, 209–220. [Google Scholar] [CrossRef]
  134. Seiler, N.; Duranton, B.; Raul, F. The polyamine oxidase inactivator MDL 72527. Prog. Drug Res. 2002, 59, 1–40. [Google Scholar] [CrossRef]
  135. Zhu, Q.; Sun, Z.; Jiang, Y.; Chen, F.; Wang, M. Acrolein scavengers: Reactivity, mechanism and impact on health. Mol. Nutr. Food Res. 2011, 55, 1375–1390. [Google Scholar] [CrossRef]
  136. Yao, L.; Wu, Y.T.; Tian, G.X.; Xia, C.Q.; Zhang, F.; Zhang, W. Acrolein Scavenger Hydralazine Prevents Streptozotocin-Induced Painful Diabetic Neuropathy and Spinal Neuroinflammation in Rats. Anat. Rec. 2017, 300, 1858–1864. [Google Scholar] [CrossRef] [Green Version]
  137. Leung, G.; Sun, W.; Zheng, L.; Brookes, S.; Tully, M.; Shi, R. Anti-acrolein treatment improves behavioral outcome and alleviates myelin damage in experimental autoimmune encephalomyelitis mouse. Neuroscience 2011, 173, 150–155. [Google Scholar] [CrossRef] [Green Version]
  138. Wu, X.; Cui, W.; Guo, W.; Liu, H.; Luo, J.; Zhao, L.; Guo, H.; Zheng, L.; Bai, H.; Feng, D.; et al. Acrolein Aggravates Secondary Brain Injury After Intracerebral Hemorrhage Through Drp1-Mediated Mitochondrial Oxidative Damage in Mice. Neurosci. Bull. 2020, 36, 1158–1170. [Google Scholar] [CrossRef]
  139. Cebak, J.E.; Singh, I.N.; Hill, R.L.; Wang, J.A.; Hall, E.D. Phenelzine Protects Brain Mitochondrial Function In Vitro and In Vivo following Traumatic Brain Injury by Scavenging the Reactive Carbonyls 4-Hydroxynonenal and Acrolein Leading to Cortical Histological Neuroprotection. J. Neurotrauma 2017, 34, 1302–1317. [Google Scholar] [CrossRef] [Green Version]
  140. Hill, R.L.; Singh, I.N.; Wang, J.A.; Hall, E.D. Effects of Phenelzine Administration on Mitochondrial Function, Calcium Handling, and Cytoskeletal Degradation after Experimental Traumatic Brain Injury. J. Neurotrauma 2019, 36, 1231–1251. [Google Scholar] [CrossRef]
  141. Musgrave, T.; Benson, C.; Wong, G.; Browne, I.; Tenorio, G.; Rauw, G.; Baker, G.B.; Kerr, B.J. The MAO inhibitor phenelzine improves functional outcomes in mice with experimental autoimmune encephalomyelitis (EAE). Brain Behav. Immun. 2011, 25, 1677–1688. [Google Scholar] [CrossRef] [PubMed]
  142. Chen, Z.; Park, J.; Butler, B.; Acosta, G.; Vega-Alvarez, S.; Zheng, L.; Tang, J.; McCain, R.; Zhang, W.; Ouyang, Z.; et al. Mitigation of sensory and motor deficits by acrolein scavenger phenelzine in a rat model of spinal cord contusive injury. J. Neurochem. 2016, 138, 328–338. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Figure 1. Diagrammatic representation of acrolein formation through polyamine oxidation pathway (A) and lipid peroxidation pathway (B). PAO: polyamine oxidase; SMOX: spermine oxidase; SSAT: spermidine/spermine N1-acetyltransferase; ALEs: advanced lipid peroxidation end products; MDA: malondialdehyde; ROS: reactive oxygen species; 4-HNE: 4-Hydroxynonenal.
Figure 1. Diagrammatic representation of acrolein formation through polyamine oxidation pathway (A) and lipid peroxidation pathway (B). PAO: polyamine oxidase; SMOX: spermine oxidase; SSAT: spermidine/spermine N1-acetyltransferase; ALEs: advanced lipid peroxidation end products; MDA: malondialdehyde; ROS: reactive oxygen species; 4-HNE: 4-Hydroxynonenal.
Biomolecules 10 01579 g001
Figure 2. Schematic representation of the potential mechanisms of action of acrolein in the diabetic retina.
Figure 2. Schematic representation of the potential mechanisms of action of acrolein in the diabetic retina.
Biomolecules 10 01579 g002
Figure 3. The molecular targets of acrolein in diabetic retinopathy (DR) development. GSH, glutathione; HO-1, heme oxygenase-1, Nrf2, nuclear factor erythroid 2-related factor 2; CCL1, chemokine (C-C motif) ligand 1; CXCL1, C-X-C motif chemokine ligand 1; CCL2, chemokine (C-C motif) ligand 2; ICAM-1, intercellular adhesion molecule 1; IL-6, interleukin 6; IL-1β, interleukin 1 beta, TNF-α, tumor necrosis factor alpha; VEGF, vascular endothelial growth factor and TGF-β, transforming growth factor beta.
Figure 3. The molecular targets of acrolein in diabetic retinopathy (DR) development. GSH, glutathione; HO-1, heme oxygenase-1, Nrf2, nuclear factor erythroid 2-related factor 2; CCL1, chemokine (C-C motif) ligand 1; CXCL1, C-X-C motif chemokine ligand 1; CCL2, chemokine (C-C motif) ligand 2; ICAM-1, intercellular adhesion molecule 1; IL-6, interleukin 6; IL-1β, interleukin 1 beta, TNF-α, tumor necrosis factor alpha; VEGF, vascular endothelial growth factor and TGF-β, transforming growth factor beta.
Biomolecules 10 01579 g003
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Alfarhan, M.; Jafari, E.; Narayanan, S.P. Acrolein: A Potential Mediator of Oxidative Damage in Diabetic Retinopathy. Biomolecules 2020, 10, 1579. https://doi.org/10.3390/biom10111579

AMA Style

Alfarhan M, Jafari E, Narayanan SP. Acrolein: A Potential Mediator of Oxidative Damage in Diabetic Retinopathy. Biomolecules. 2020; 10(11):1579. https://doi.org/10.3390/biom10111579

Chicago/Turabian Style

Alfarhan, Moaddey, Eissa Jafari, and S. Priya Narayanan. 2020. "Acrolein: A Potential Mediator of Oxidative Damage in Diabetic Retinopathy" Biomolecules 10, no. 11: 1579. https://doi.org/10.3390/biom10111579

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