Next Article in Journal
Waste Citrus limon Leaves as Source of Essential Oil Rich in Limonene and Citral: Chemical Characterization, Antimicrobial and Antioxidant Properties, and Effects on Cancer Cell Viability
Next Article in Special Issue
Exogenous CFH Modulates Levels of Pro-Inflammatory Mediators to Prevent Oxidative Damage of Retinal Pigment Epithelial Cells with the At-Risk CFH Y402H Variant
Previous Article in Journal
An Ethyl Acetate Extract of Eryngium carlinae Inflorescences Attenuates Oxidative Stress and Inflammation in the Liver of Streptozotocin-Induced Diabetic Rats
Previous Article in Special Issue
Matrix Metalloproteinase 13 Is Associated with Age-Related Choroidal Neovascularization
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

New Insights on Dietary Polyphenols for the Management of Oxidative Stress and Neuroinflammation in Diabetic Retinopathy

by
Gustavo Bernardes Fanaro
1,
Marcelo Rodrigues Marques
2,
Karin da Costa Calaza
3,
Rafael Brito
4,
André Moreira Pessoni
5,
Henrique Rocha Mendonça
6,
Deborah Emanuelle de Albuquerque Lemos
7,
José Luiz de Brito Alves
7,
Evandro Leite de Souza
7,* and
Marinaldo Pacífico Cavalcanti Neto
6
1
Institute of Health and Biotechnology, Federal University of Amazonas, Manaus 69460000, Amazonas, Brazil
2
Federal Institute of Science and Technology of Maranhão, Zé Doca 65365000, Maranhão, Brazil
3
Department of Neurobiology, Institute of Biology, Fluminense Federal University, Niterói 24210201, Rio de Janeiro, Brazil
4
Department of Cellular and Molecular Biology, Institute of Biology, Fluminense Federal University, Niterói 24210201, Rio de Janeiro, Brazil
5
Faculté de Medicine, Université Laval, Quebec, QC G1V 0A6, Canada
6
Institute of Biodiversity and Sustainability (NUPEM), Federal University of Rio de Janeiro, Macaé 27965045, Rio de Janeiro, Brazil
7
Department of Nutrition, Health Sciences Center, Federal University of Paraíba, João Pessoa 58051900, Paraíba, Brazil
*
Author to whom correspondence should be addressed.
Antioxidants 2023, 12(6), 1237; https://doi.org/10.3390/antiox12061237
Submission received: 15 April 2023 / Revised: 5 June 2023 / Accepted: 6 June 2023 / Published: 8 June 2023
(This article belongs to the Special Issue Oxidative Stress in Retinal Degeneration)

Abstract

:
Diabetic retinopathy (DR) is a neurodegenerative and vascular pathology that is considered one of the leading causes of blindness worldwide, resulting from complications of advanced diabetes mellitus (DM). Current therapies consist of protocols aiming to alleviate the existing clinical signs associated with microvascular alterations limited to the advanced disease stages. In response to the low resolution and limitations of the DR treatment, there is an urgent need to develop more effective alternative therapies to optimize glycemic, vascular, and neuronal parameters, including the reduction in the cellular damage promoted by inflammation and oxidative stress. Recent evidence has shown that dietary polyphenols reduce oxidative and inflammatory parameters of various diseases by modulating multiple cell signaling pathways and gene expression, contributing to the improvement of several chronic diseases, including metabolic and neurodegenerative diseases. However, despite the growing evidence for the bioactivities of phenolic compounds, there is still a lack of data, especially from human studies, on the therapeutic potential of these substances. This review aims to comprehensively describe and clarify the effects of dietary phenolic compounds on the pathophysiological mechanisms involved in DR, especially those of oxidative and inflammatory nature, through evidence from experimental studies. Finally, the review highlights the potential of dietary phenolic compounds as a prophylactic and therapeutic strategy and the need for further clinical studies approaching the efficacy of these substances in DR management.

1. Introduction

Diabetic retinopathy (DR) is the most common complication of diabetes mellitus (DM) and the leading cause of blindness, affecting approximately 95% of patients with type 1 DM and more than 60% of patients with type 2 DM. DR is a public health and socioeconomic burden that could be prevented or reduced by implementing interventions, such as glycemic control, healthy lifestyle behaviors, and dietary modification [1,2,3].
According to data from the International Diabetes Federation (IDF) [4], 537 million people aged 20 to 79 years were diagnosed with diabetes worldwide in 2021, an increase of 16% over the previous two years, and projections for 2030 point to 643 million cases. The alarming global evolution of DM and the increasing development and progression of DR suggest early diagnosis and more effective, accessible, and cost-effective prophylactic and therapeutic measures.
Poor glycemic and blood pressure control and duration of diabetes are significant risk factors for developing DR, and rigorous control of these parameters promotes long-term benefits in delaying or preventing DR [5,6,7,8,9,10,11,12]. Pregnancy and puberty [13,14,15,16], kidney disease [17,18], and a high body mass index (BMI) [19] are also risk factors for DR. Even when these risk factors are under control, some patients still develop diabetic macular edema (DME), the major vision-threatening late complication of DR [20,21].
The nutritional profile is also a determining factor for DR [22,23]. The correlation between the consumption of different dietary components (macro- or micronutrients) [24,25,26] or even specific dietary patterns (e.g., Mediterranean diet) has been investigated in this context [27,28]. Despite the growing body of evidence linking dietary intake and DR, current data are inconclusive, for example, on which dietary components are associated with the protection or susceptibility to DR [22].
Although clinical protocols and nutritional recommendations are well established in treating DM, there is still a lack of specific dietary recommendations for DR [29,30]. In this sense, the development and implementation of specific nutritional protocols and therapies with bioactive compounds may represent an attractive, accessible, and cost-effective strategy in the management of DR [31,32,33].
Despite remarkable progress in understanding the molecular and pathophysiological mechanisms of DR, some gaps remain, including those related to the early stages of DR. Currently, therapeutic strategies, including glycemic and blood pressure control, laser photocoagulation, intravitreal injections of anti-inflammatory or VEGF neutralizing agents, and vitreoretinal surgery are limited to late stages of the disease, reducing the chances of preventing vision loss [34,35,36,37,38]. In addition, not all patients respond satisfactorily to current clinical therapies, suggesting that developing effective strategies for the prevention and treatment of DR is critical [36,38].
Polyphenols are a diverse group of bioactive molecules derived primarily from plant foods, such as fruits, vegetables, whole grains, coffee, nuts, and red wine. These compounds exert health benefits primarily due to their antioxidant and anti-inflammatory properties [39,40]. Experimental and clinical studies on the effects of a polyphenol-rich diet have shown significant modulatory effects on the pathophysiological mechanisms involved in chronic diseases, such as diabetes and cardiovascular and neurodegenerative diseases, suggesting a significant prophylactic and therapeutic potential [41,42]. Given the increase in pro-inflammatory markers and reactive oxygen species (ROS) in DR, dietary intervention or supplementation with polyphenols may be a promising strategy to manage the pathology.
This review aims to comprehensively describe and clarify the effects of dietary polyphenols on the pathophysiological mechanisms involved in DR, especially those related to oxidative and inflammatory mechanisms, as well as to discuss evidence from animal models on the effects of dietary polyphenols as a prophylactic and therapeutic strategy in DR management.

2. Retina

The retina transduces and preprocesses the information via an intraretinal circuit and sends it via the optic nerve to the visual nuclei of the brain [43]. The mature vertebrate retina is organized into layers [44,45], such as (i) the outer nuclear layer (ONL), composed of the cell bodies of rod and cone photoreceptors, located closest to the retinal pigmented epithelium (RPE); (ii) the inner nuclear layer (INL), composed of the cell bodies of interneurons-horizontal, bipolar, amacrine, and displaced ganglion cells-located between the ONL; and (iii) the ganglion cell layer (GCL). The GCL is located near the vitreous body of the retina and consists of ganglion cells, displaced amacrine cells, and some light-sensitive ganglion cells. Interspersed between these nuclear layers are the outer plexiform layer (OPL) and the inner plexiform layer (IPL), where synaptic contacts occur. Ganglion cells are the projection neurons, and their axons form the nerve fiber layer (NFL) within the retina and give rise to the optic nerve as they exit the retina [44,46].
Photoreceptors communicate with the rod- or cone-specific bipolar cells and horizontal cells in the OPL. Bipolar cells synapse with ganglion cells in the IPL. This circuit is named the vertical/radial pathway, and all cells use glutamate as a neurotransmitter [44,47]. Horizontal and amacrine cells may modulate the radial pathway in the OPL and IPL and mediate the processing of light/image stimuli, contrast, and detection of movement direction [48].
The retinal vasculature consists of the choroidal and intraretinal blood vessels responsible for the blood supply of the outer and inner retina, respectively. In humans, the inner retina presents a peripapillary plexus in the innermost part of the NFL, accompanied by inner (in the GCL) and outer (in the IPL, INL to OPL) intraretinal beds. Interestingly, the specialized region for high visual acuity in primates, the fovea, is avascular, which minimizes light distortion [49].
In addition to neurons, the retina contains three types of glial cells, Müller cells, astrocytes, and microglia. Müller glia, which comprises 90% of retinal glial cells, have a cell body in the center of the INL and radial processes that extend through all retina layers. Consistent with the multiple roles that Müller glia play in retinal homeostasis and physiology, dysfunction of these cells in DR may be related to retinal dysfunction and severity of DR [47,48,50,51]. Figure 1 shows a schematic drawing of the retina.

3. Diabetic Retinopathy

DR is a multifactorial pathology that results from a dysfunction in a variety of retinal cells, including Müller glia, ganglion cells, endothelial cells, and photoreceptors, as well as optic nerve damage and, later, retinal neuronal cell death and vascular lesions [52,53]. Although neural retina/optic nerve changes occur before vascular alterations in both human and animal models, DR is a chronic microvascular disease characterized by vasculopathy, hyperpermeability, hypoperfusion, and neoangiogenesis of slow progression, accompanied by vitreous hemorrhage, retinal detachment, diabetic macular edema process (DME), and/or macular ischemia, resulting in neural abnormalities and retinal deficits [20,54,55]. Although DME and macular ischemia can occur at any stage of the disease, they are more common in the later stages of DR [54].
Chronic hyperglycemia promotes the thickening of the retinal capillary basement membrane, impairs adherence of tight junctions between endothelial cells, and makes blood vessels more permeable. These events define the process known as the blood–retinal barrier (BRB) breakdown. This phenomenon is accompanied by decreased pericytes and mesenchymal cells surrounding endothelial cells, providing structural support for the vessel wall and vascular repair processes [56,57]. The loss of pericytes leads to voids in the capillary walls. In response, capillary endothelial cell growth is induced toward the inner part of the vessel wall, favoring the formation of cell clusters that obstruct the vessel lumen and the appearance of hemorrhagic spots and microaneurysms. This combination of damage compromises vascular function and the oxygen and nutrient supply to the retinal tissue, leading to ischemia, increased synthesis, and release of pro-angiogenic factors and neovascularization [55,57].
DR is classified based on pathophysiological characteristics and vascular manifestations during the clinical course, the initial phase, known as non-proliferative DR (NPDR), and the advanced phase, known as proliferative DR (PDR). The presence of new blood vessels characterizes the PDR stage [58].
NPDR is characterized by morphological changes and vascular damage, including alterations in the microvasculature and an increase in vascular caliber. During this phase, hyperglycemia causes hemodynamic dysfunction, increased permeability, microaneurysms, hemorrhages, and exudates. These manifestations generally progress slowly, and some patients may be asymptomatic. As the disease progresses, the frequency of hemorrhagic events increases, disrupting the retina’s oxygen supply and metabolic demands and increasing the risk of ischemic events. Fundus examination may reveal whitish spots known as “cotton-wool spots” that indicate ischemic processes or retinal infarction [56,59,60]. Thus, the abnormal angiogenic process leads to a more advanced and critical PDR [55,61].
Approximately half of NPDR patients progress to PDR in less than one year [62]. The main feature of PDR is the appearance of new blood vessels because of the local accumulation of vascular growth factors caused by prolonged hypoxia, followed by the formation of ischemic zones. These vessels are delicate, composed entirely of the endothelium, and grow along the retina and into the vitreous without initially causing symptoms or vision loss. However, because their walls are thin and fragile, the vessels can rupture and release blood into the vitreous cavity, causing severe vision loss and even blindness [55,63]. Neovascularization is usually accompanied by fibrous tissue growth, which is visible on ophthalmoscopy [62].
Although DR has been predominantly considered a microvascular disease, increasing evidence suggests that retinal dysfunction and degeneration are critical features associated with disease progression and that early neural injury occurs before vascular damage in DR [64]. Data from electroretinograms of diabetic patients show a decrease in the oscillatory potential, which would be the first observable change in the retina of these individuals [65]. Other parameters are altered in individuals with DM without vascular changes, such as contrast sensitivity, color vision, and oscillatory potentials [53]. Histologic analysis of clinical and postmortem specimens confirmed alterations in retinal structural parameters, such as changes in the thickness of different retinal layers, neuronal death, and increased oxidative stress before vascular changes visible with fundoscopy, supporting the concept of an early neurodegenerative phase of DR preceding NPDR [65,66,67,68].
In this sense, a prospective and longitudinal human study of 45 patients with type 1 DM, without or with minimal DR, reported significant and progressive impairment of both the nerve fiber layer (NFL) and the ganglion cell (GC)/inner plexiform layer, independent of glycated hemoglobin, age, and sex [64]. To confirm these findings, eye samples from six deceased patients with DM, without or with minimal DR, were compared to eyes from donors of a similar age without DM. The first group showed a significantly thinner NFL but no difference in GC density. Surprisingly, the data showed that despite the neuronal changes observed in the DR group, there was no difference in retinal capillary density between the groups. These findings were confirmed in two experimental diabetes mouse models. Recently, other studies using optical coherence tomography showed decreased retinal thickness (INL/GCL and NFL) in diabetic patients without signs of DR, indicating neurodegeneration before vascular changes [64,69,70,71,72]. These results suggest that retinal diabetic neuropathy is not necessarily a consequence of retinal ischemia. It develops gradually, independent of glycated hemoglobin, age, and sex, before the onset of microvasculopathy.
All retinal cells (e.g., ganglion, bipolar, amacrine, and photoreceptor cells) show altered function before visible microvascular lesions [53]. Therefore, the description of DR as a mere “microvascular pathology” is an oversimplified and flawed characterization that hinders the exploration of novel therapeutic approaches based, for example, on intervention on neurodegenerative pathways [73].

4. Cellular and Molecular Pathways Involved with Oxidative Stress and Inflammatory Response during DR

The retina is susceptible to oxidative stress due to its high polyunsaturated fatty acids (PUFA) concentration and rapid oxygen and glucose uptake. Another critical element in the pathophysiology of DR is inflammation. The oxidative stress induces ROS production, the root cause of neuropathy and retinopathy [74].
Glucose is the primary precursor for energy production in the cell. The citric acid cycle requires glycolysis-derived pyruvate to produce the high-energy electron carriers FADH2 and NADH, which fuel the electron transport required for ATP production. At the end of this electron transport chain, oxygen molecules serve as electron acceptors, limiting reactions that would otherwise impair the function of other cellular components. High glucose levels cause increased glycolysis rate, pyruvate levels, and FADH2 and NADH production. When these electron carriers reach the upper limit of the voltage gradient across the inner mitochondrial membrane, electron transfer through complex III is impaired, pushing electrons back to coenzyme Q, which mediates superoxide (O2) production [75]. Superoxide belongs to a class of reactive oxygen species (ROS). ROS are highly reactive, mainly due to unpaired electrons in the outer shell of their atoms, causing dysfunction in enzymes and altering the interaction properties of proteins, lipids, sugars, and nucleic acids. O2 can be converted by superoxide dismutase activity to the non-radical ROS, hydrogen peroxide (H2O2). H2O2 alters protein interactions or function through cysteine oxidation. H2O2 can be converted to hydroxyl radical (OH.) by the Fenton reaction, as well as to hypochlorous acid (HOCl) when targeted by neutrophil myeloperoxidase during an inflammatory process [76,77]. Indeed, diabetes-induced neuroinflammation showed increased NO synthase (NOS2) levels in the retina. The reaction of nitric oxide (NO) with O2 forms the family member of the reactive nitrogen species peroxynitrite (ONOO−). ROS damages DNA leading to the activation of DNA repair enzymes, such as poly (adenosine diphosphate (ADP)-ribose) polymerase. This enzyme produces ADP-ribose, which inhibits glycerol-3-phosphate dehydrogenase (GAPDH), which is responsible for a step in the glycolytic pathway. The result is an accumulation of glycolytic intermediates used by the polyol and hexosamine pathways, advanced glycation end products (AGEs), and upregulation of the PKC signaling pathway [78].
All these pathways increase ROS and form a positive feedback loop to increase oxidative stress in the cell. Among many ROS-generating systems, NADPH oxidase (NOX) activity seems particularly important for generating cytoplasmic ROS, especially for neovascularization [79]. NOX is a group of enzymes that catalyzes the production of O2 by transferring an electron from NADPH to O2. NOX4, a subtype of NOX, can also produce H2O2. NOX activity is upregulated by several signals produced during the progression of DR, including tumor necrosis factor-alpha (TNF-α), several interleukins, and AGE signaling. Therefore, NOX appears to be important in advance of DR, and inhibition of NOX has emerged as a promising therapeutic target for DR [80,81]. Thus, oxidative stress is a constitutive problem and a hallmark of DM.
As mentioned above, hyperglycemic conditions induce the production of AGEs. Briefly, AGEs are generated by the reduction in amino acids, lipids, or nucleic acids by glucose, triggering the formation of Schiff bases. Rearrangements of Schiff bases lead to forming Amadori products, which have reactive carbonyl groups. These carbonyl groups interact with amino, sulfhydryl, and guanidine functional groups, denaturing and cross-linking target proteins. Finally, reactions of Amadori products with arginine or lysine within target proteins lead to the formation of stable AGEs. For example, AGE can signal through specific receptors, the receptor for advanced glycation end products (RAGE), mediating paracrine and autocrine signaling involved in the pathology of diabetic retinopathy [82].
AGEs and oxidative stress activate retinal micro- and macroglia during diabetic retinopathy [83]. Once activated, microglia produced interleukin (IL)-1β, IL-6, IL-8, and TNFα under the control of NF-kappaB (NF-kB) [84,85,86,87,88,89]. Müller glia is activated in DR, leading to increased secretion of IL-6, IL-8, VEGF, INF-γ, TNF-α, IL-1β, monocyte chemoattractant protein-1 (MCP-1), and EGF [90,91]. In addition, galectin-3 is upregulated in Müller glia in diabetic rats, and the absence of galectin-3 reduces the number of astrocytes and macrophage-like cells, both quiescent and activated, in the optic nerve and protects against ganglion cell death [92]. Retinal astrocytes are consistently activated by oxidative damage, increasing their secretion of IL-6, MCP-1, and MIP-2 under the control of p38MAPK and NF-kB signaling [93]. In addition, the AGE receptor mediates astrocyte activation biased toward a pro-inflammatory profile, increasing NF-kB and complement C3 expression, mediating synaptic loss [94].
All these secreted mediators orchestrate various pathological events leading to persistent inflammation, ROS production, neurodegeneration, blood–retinal barrier permeability, and defective neovascularization, which are hallmarks of diabetic retinopathy. TNF-α and galectin-3 mediate apoptotic cell death of retinal ganglion cells [92,95]. Both IL-1β and TNF-α increased the content of ICAM-1 on the endothelial surface, allowing monocyte diapedesis through VLA4 and CD18 binding [96]. These monocytes are attracted to the retina by MCP-1 secreted by astrocytes and Müller glia and differentiate into macrophages once in the retinal tissue [97]. IL-8 recruits neutrophils, which release myeloperoxidase granules, further contributing to the maintenance of oxidative stress [98,99]. These leukocyte recruitments rely on the luminal exposure of MIP-2, which stimulates the upregulation of leukocyte integrins during diapedesis [100]. Oxidative damage and IL-6 stimulate VEGF production by Müller glia and endothelial cells, respectively [101]. VEGF decreases occludin content in endothelial cells, thereby increasing the permeability of the blood–retinal barrier, leading to retinal edema and degeneration [102]. To worsen the scenario, VEGF and EGF stimulate neovascularization, expanding the area covered by defective capillaries in the proliferative phase of diabetic retinopathy [103]. Thus, neuroinflammation and oxidative stress produce each other in a positive feedback loop, contributing to neuronal death and vascular pathology of diabetic retinopathy [53].

5. Dietary Polyphenols: A Potential Strategy for Management of Diabetic Retinopathy?

Early studies have demonstrated the importance of consuming foods that contain substances with antioxidant properties for the prevention of cardiovascular disease, cancer, and age-related degenerative brain disorders [104,105,106]. Natural products and plant extracts with antioxidant properties, low toxicity, and known health-promoting properties have been suggested as possible treatments for eye diseases [107,108,109,110,111,112,113]. Although polyphenols offer several advantages, it is crucial to consider certain factors when evaluating their physiological effects. Some polyphenols have low water solubility and limited oral bioavailability, requiring higher doses, which are significant barriers to their broader use in therapies [114,115]. Furthermore, their physicochemical stability, gastrointestinal absorption, and metabolism are crucial to ensure their efficacy [116,117]. In this context, given the wide range of biological activities associated with these molecules (Figure 2), it seems plausible to study not only the potential health benefits but also to develop strategies and tools (pharmacotechnical and biotechnological) to overcome the limitations mentioned above, accompanied by research on the molecular mechanisms of dietary polyphenols and/or their supplementation in diseases affecting the human eye, such as DR.

5.1. Ferulic Acid

Ferulic acid (FA) was first isolated from Ferula foetid in 1866. FA is a polyphenol of the hydroxycinnamic acid (HCA) family, derived from secondary metabolites of lignin biosynthesis from phenylalanine and tyrosine, found in the free or conjugated form [118,119,120]. FA has shown various health biological and therapeutic properties, including anti-inflammatory, antioxidant, antibacterial, anti-diabetic, anti-carcinogenic, anti-aging, and neuroprotective [121].
FA is a potent antioxidant by neutralizing free radicals and enhancing the cellular stress response by upregulating cytoprotective systems, such as heme oxygenase-1, heat shock protein 70, extracellular signal-regulated kinase ½, and the proto-oncogene Akt. In addition, FA can suppress the expression and/or activity of cytotoxic enzymes, such as cyclooxygenase-2 (COX-2), caspases, and inducible nitric oxide synthase. (iNOS) [122]. In animal models of hepatocytic and aortic inflammation and ulcerative colitis, FA treatment inhibited the production of the inflammatory cytokines IL-6, IL-1, and TNF-α. The ability of hydroxycinnamic acids, including FA, to inhibit the NF-kB pathway and suppress the expression of COX-2 and iNOS may be a representative anti-inflammatory mechanism [123]. Using optical coherence tomography and electroretinography, FA or its analog ethyl ferulate attenuated morphologic and functional retinal degeneration in mice, as evidenced by reductions in outer retinal and choroidal layer thickness [124].
Retinal degeneration in DR is typically accompanied by glia-mediated neuroinflammation. FA supplementation reduces neuroinflammation by suppressing microglial activation in mice [125]. The effect on human retinal pigment epithelial cells has also been studied. A recent study demonstrated that FA exerts a potent protective effect against oxidative stress-induced retinal damage. The authors showed that preincubation of human retinal pigment epithelial (ARPE-19) cells in culture with FA for 24 h significantly attenuated apoptosis induced by four h of H2O2 challenge by upregulating Bcl-2 and downregulating Bax [126].
Zhu et al. [127] investigated the anti-apoptotic effect of FA in RPE cells (ARPE-19) exposed to high glucose and its cytoprotective effects in the retina of db/db diabetic mice. Treatment with FA significantly increased cell viability and suppressed cell apoptosis in ARPE-19 cells compared with the untreated group. In addition, treatment with FA (0.05 g/kg for two months) alleviated degenerative changes in the retinal tissues of db/db mice. It sharply decreased the expression of apoptosis-related markers in the retinal tissues compared to the control group. The effect shown in the FA-treated groups was related to P53 and BAX inactivation and Bcl2 activation.
Adenosine monophosphate-activated protein kinase (AMPK) is a potent regulator of metabolism and an attractive target for antidiabetic drugs. Hyperglycemia induces oxidative stress and decreases AMPK phosphorylation. Sigh and Patil [128] showed that trans-FA increased cellular phosphorylation of AMPK in cultured HepG2 and L6 cells exposed to high glucose in a time-dependent manner, besides causing upregulation of GLUT2 and GLUT4 and inhibition of JNK1/2 activity.
In diabetic patients, the formation and accumulation of AGEs are related to the onset and development of atherosclerosis by inducing oxidative damage, vascular contraction, and blood coagulation state, stimulating the release of cytokines, and increasing the inflammatory process in blood vessels. Reducing or inhibiting AGE formation may be a prospective strategy to prevent or delay the development of vascular complications in DR.
Liu et al. [129] have demonstrated an inhibitory effect of FA on AGE formation in vitro. Pretreatment with FA prevented AGE-induced p38 MAPK signaling and NF-κB pathway activation in human vascular endothelial cells (HUVEC). Because of the many possibilities, researchers have worked on strategies for the ocular delivery of FA by microencapsulation. This drug delivery system eliminates losses during the digestive process and increases in situ bioavailability [130]. Table 1 summarizes the findings for the use of FA in DR.

5.2. Curcumin

Curcumin (1,7-bis[4-hydroxy-3-methoxyphenyl]-1,6-heptadiene-3,5-dione), also known as diferuloylmethane, Natural Yellow 3, or E100, according to the European coding of food additives, belongs to the group of polyphenols. It was first isolated from the rhizome of Curcuma longa L. (turmeric) in 1815, the main component of this plant species. It is mainly grown in India and China, and it was first known for its role as a food additive or spice in cooking (e.g., curry) [131,132]. Clinical studies have reported that curcumin exerts anticancer, anti-inflammatory, and antioxidant effects. Furthermore, curcumin has a potential neuroprotective effect [133]. Studies in diabetic mouse models have shown that oral administration of various doses of curcumin reduces blood glucose levels [134] and HbA1C levels [135] and improve insulin sensitivity [136,137]. Dietary curcumin also effectively reduced fasting blood glucose, urine glucose, and urine volume in STZ-induced diabetic rats [138].
Early evidence has shown that using curcumin as a therapeutic agent can help to improve the condition of patients with retinal diseases [139,140,141,142]. Many of these effects have been demonstrated using RPE cells and the retina of diabetic rats as models. Curcumin may exert its pharmacological properties by inhibiting cell proliferation and angiogenesis by targeting molecular and cellular pathways involved in the pathogenesis of retinal diseases [143].
Under hypoxic conditions, HIF-1 plays a significant role in the expression of VEGF [144,145,146], and studies have described how curcumin can decrease VEGF production through the control of HIF-1 [147,148]. The progression of DM is associated with increased ROS levels in the neural retina and vascular endothelium [149,150]. These events may decrease the antioxidant defense system and enhance oxidative damage in diabetic retinopathy [143,151,152].
Curcumin has a potent anti-inflammatory effect by downregulating the expression of TNF-α, IL-1, IL-6, and IL-8. In addition, curcumin can downregulate the cellular action of angiogenic proteins, such as VEGF, by inhibiting transcription factors, including NF-κB, HIF-1α, AP-1, and early growth response-1 (EGR-1) [153,154,155]. Curcumin treatment reduced NFκ-B and phosphorylated NFκ-B, IKB-α, IL-1β, and Bax levels, while increasing Bcl-2 expression in rat retinal vascular endothelial cells (RRVECs) exposed to high glucose [156].
In addition, Ran et al. [157] observed that curcumin downregulated high glucose-induced inflammatory injury in ARPE-19 by modulating the ROS/PI3K/AKT/mTOR signaling pathway and decreasing the levels of pro-inflammatory cytokines TNF-α, IL-1β, and IL-6. These findings show that curcumin exerts anti-inflammatory effects by modulating multiple signaling pathways, making it a promising molecule against inflammation and oxidative stress in DR.
Under physiological conditions, the nuclear erythroid 2 p45-related factor (Nrf2) is an essential protein located primarily in the cytoplasm. In response to oxidative stress, Nrf2 primarily modulates endogenous defense. Nrf2 translocates to the nucleus and binds to specific DNA sites, such as electrophile response elements (EpRE) or antioxidant response elements (ARE), to initiate the transcription of cytoprotective genes [158,159]. Then, 1,5-bis(2-trifluoromethylphenyl)-1,4-pentadien-3-one(C3), a curcumin analog, inhibited the cytotoxic effects of acrolein in the ARPE-19 cells by directly activating the Nrf2 signaling cascade. More effectively than curcumin, it increased GSH and mitochondrial reductive enzymes while protecting ARPE-19 cells from oxidative stress [160].
Curcumin may help treat DR since it increases Nrf2/HO-1 and ERK1/2 expression and reduces ROS-induced apoptosis in RPE cells [161]. Xie et al. [162] investigated the effect of curcumin on retinal damage in streptozotocin (STZ)-induced diabetic rats and found that four weeks of oral curcumin, subcutaneous insulin, or combination therapy improved histopathologic parameters, oxidative stress markers, and reduced plasma glucose in all groups studied. In addition, curcumin had a better anti-photoreceptor apoptosis effect compared to insulin treatment. Curcumin has a regulatory impact on oxidative stress-induced activation of the Nfr2 pathway and inhibition of the AGE–RAGE pathway and extracellular matrix (ECM)-receptor interaction in the diabetic retina. Table 2 summarizes the findings for the use of curcumin in DR.

5.3. Flavonoids

Polyphenols are classified into flavonoids (e.g., catechins, isoflavones, apigenin, quercetin, lutein, and anthocyanins) and non-flavonoids (e.g., chlorogenic acids in coffee) [163]. Flavonoids represent one of the main groups of antioxidants found in plants and are divided into seven subclasses, flavonols, flavones, isoflavones, anthocyanidins, flavanones, flavanols, and chalcones (these molecules are precursors of flavonoids and isoflavonoids) [164]. In addition to antioxidant activity, flavonoids have important biological activities, such as anti-inflammatory, antimicrobial, anticarcinogenic, and immunostimulatory effects [165]. The consumption of flavonoids is inversely associated with cancer, cardiovascular disease, and neurodegenerative disorders [166].

5.3.1. Epigallocatechin Gallate

Flavanols (flavan-3-ols) differ from other flavonoids by having a hydroxyl in position 3 of the C ring and no double bond between positions 2 and 3 of the same ring. These compounds are found as single molecules (glycated or aglycone) or organized in polymers. Among the flavanols, the catechin groups (catechin, catechin gallate, epicatechin, epicatechin gallate, epigallocatechin, and epigallocatechin gallate), proanthocyanidins, theaflavins, and thearubigins stand out. Food sources of flavanols include tea (Camellia sinensis), grape seeds, cherries, berries, and the skins of various fruits [164].
Epidemiological studies associated C. sinensis consumption, mainly green tea, with a reduced risk of several types of cancer [167,168], and its polyphenols are effective chemoprevention agents [169,170]. Other properties include antiallergic, antimicrobial, immunostimulatory, and anti-inflammatory effects [171], as well as increased insulin activity [172] and protection against cardiovascular and brain disease [173,174].
Retinal GSH levels and activity of the antioxidant enzymes SOD and CAT are reduced in diabetic rats. The administration of green tea extract at 200 mg/kg daily for 16 weeks to diabetic rats restored GSH levels. It almost increased the SOD and CAT enzyme activities to the same levels as the non-diabetic group. In addition, green tea treatment led to a reduction in the expression of pro-inflammatory markers TNF and VEGF in the retina and a decrease in the thickness of the retinal capillary basement membrane [175].
According to Silva et al. [176], daily consumption of green tea (5.7 g/kg) for 12 weeks reduced retinal oxidative stress, prevented the upregulation of GFAP and the downregulation of occludin and restored the expression of activated neuronal nitric oxide synthase (nNOS) in diabetic mice. Green tea regulated the AKT-dependent activation of the endothelial enzyme nitric oxide synthase (eNOS). NO produced by eNOS activation and inflammatory markers are required for VEGF-induced increased vascular permeability, which contributes significantly to BRB breakdown. In addition, green tea restored glutamate transporter, glutamate receptor, and glutamine synthetase.
Many of the beneficial effects promoted by the consumption of green tea may be due to several antioxidant compounds in its composition, most notably EGCG, as evidenced by in vitro studies that evaluated the effect of incubating retinal cells with EGCG in a hyperglycemic medium. Chan et al. [177] observed that incubation of RPE with 10 µM EGCG inhibited the migration of adult ARPE19 cells and decreased the adhesion of epithelial cells to fibronectin. In addition, EGCG (3 µM) effectively inhibited the migration of RPE cells by preventing the activation of PI3K/Akt, ERK1/2, and p38 phosphorylation and blocking the MAPKs pathway.
Wang et al. [178] demonstrated that EGCG at 10 µM promoted autophagy in rat retinal Müller cells incubated in a hyperglycemic medium. The authors found that EGCG treatment decreased P62 protein levels, maintained LC3-B and Beclin-1 protein levels, and stimulated autophagosome and autolysosome formation. The EGCG-induced decrease in the mTOR pathway can explain these effects on autophagy.

5.3.2. Quercetin

Compared to other flavonoids, flavonols (3-hydroxyflavones) have a higher concentration of hydroxyls in the A and B rings. Quercetin, myricetin, kaempferol, and galanin are some of the primary representatives of this class, which are present in different foods, such as tea (Carmellia sinensis), onions, apples, and berries [179,180].
Quercetin can exert several biological effects, including anti-inflammatory, antioxidant, antidiabetic, anti-hypertensive, anti-obesity, anti-hypercholesterolemic, anti-atherosclerotic, anti-cancer, and anti-tumor [181,182,183]. In addition, quercetin consumption has been associated with protection against retinal ischemia-reperfusion injury, inflammation, and age-related ophthalmopathy [184,185].
Cheng et al. [186] showed that ARPE-19 cells incubated with quercetin (2.5–20 µM) could dose-dependently reduce IL-1β-induced expression of inflammatory markers, such as ICAM-1, sICAM-1, IL-6, IL-8, and MCP-1. This phenomenon was associated with decreased phosphorylation of MAPKs (ERK1/2, p38, and JNK1/2) and cAMP response element-binding protein (CREB).
Chai et al. [187] found that intraperitoneal administration of quercetin (150 mg/kg) for 16 weeks reduced the number of apoptotic cells and improved the thickness of both the inner and outer nuclear layers. Such retinal histopathologic changes were accompanied by a decrease in the pro-inflammatory cytokines IL-1, IL-18, IL-6, and TNF-α, as well as in the production of the pro-angiogenic factors and soluble adhesion proteins VEGF and sICAM-1. In addition, quercetin treatment increased the expression of neurotrophic factors, including brain-derived neurotrophic factor (BDNF) and nerve growth factor (NGF), suggesting that quercetin may have a neuroprotective effect in DR. The results also showed that quercetin supplementation increased the expression of heme oxygenase-1 (HO-1), decreased the expression of both HMGB-1 and TLR4, and reduced the hyperactivation of NLRP3 and NF-kB inflammasomes. The activation of HO-1 by quercetin may contribute to some of its effects. Treatment with zinc protoporphyrin (ZnPP), an HO-1 inhibitor, prevented the retinal protective effects of quercetin. The results suggest that quercetin may have therapeutic effects in DR by increasing the expression of HO-1.
The formation of retinal neovascularization (RNV) is a hallmark of PDR. In this context, understanding the mechanisms involved in retinal angiopathy and inflammation associated with DR has become essential in developing therapeutic strategies. High glucose conditions promote the activation of angiogenic mechanisms and increase the expression of NLRP3, ASC, caspase-1, IL-1, and IL-18, as well as the autophagic proteins LC3-B and Beclin-1. Quercetin treatment effectively inhibits the activation of angiogenic, inflammatory, and autophagic mechanisms induced by hyperglycemia in a dose-dependent manner (20 to 80 µmol/L) [188]. The results may suggest that the therapeutic potential of quercetin for retinal neovascularization in DR involves the inhibition of NLRP3 inflammation and autophagy signaling.
Chen et al. [189] demonstrated that STZ-induced diabetic rats receiving quercetin supplementation had decreased expression of matrix metalloproteinase-9 (MMP-9) and VEGF proteins. However, quercetin did not affect the expression of the inflammatory mediator MCP-1.
Daily five-week quercetin supplementation (50 mg/kg) increased retinal BDNF, NGF, and tropomyosin-related kinase B (TrkB) in diabetic rats. The treatment also prevented apoptosis by reducing cytochrome c and caspase-3 activity while increasing levels of the anti-apoptotic protein Bcl-2. In addition, quercetin treatment significantly increased retinal GSH levels compared to untreated diabetic rats. The results suggest that quercetin may protect neurons in the diabetic retina by increasing neurotrophic factors and preventing neuronal death through potential anti-apoptotic effects mediated by the BDNF-TrkB/Akt-synaptophysin pathway [190].
Like EGCG, quercetin inhibits photoreceptor cell apoptosis by inhibiting the PI3K/Akt pathway. By stimulating Bcl-2, which prevents Bax-mediated apoptosis, quercetin most likely suppresses the PI3K/Akt signaling pathway [191]. Another pathway of quercetin is the activation of sirtuins, a protein involved in DNA repair and regulation of metabolic processes. Sirtuin 1 protein is found in the outer nuclear layer and is secreted in the early stages of retinal degeneration, aiding the repair process [192]. Table 3 summarizes the findings for the use of quercetin in DR.

5.4. Resveratrol

Resveratrol (3,5,4 trihydroxystilbene) is a non-flavonoid polyphenol found in grapes, peanuts, blackberries, and blueberries. Its high concentration in red wine has been associated with a lower risk of cardiovascular disease in European populations who consume the beverage frequently [193,194]. Resveratrol consists of two aromatic rings connected by a methylene bridge. The molecule naturally has two isomers, cis and trans-resveratrol, the latter being the biologically active form [195,196].
Early studies have reported several beneficial health properties of resveratrol, including anti-tumor, anti-aging, anti-inflammatory, antioxidant, cardioprotective, and neuroprotective [195,197,198]. In addition, resveratrol has been shown to modulate angiogenesis, proliferation, and cell death [194].
Streptozotocin-induced diabetic rats have increased blood glucose, glycosylated hemoglobin, fructosamine, advanced glycation end products (AGEs), and decreased body weight. At different time points (1–7 months), these parameters were partially inhibited by the administration of resveratrol diluted in saline (5, 10, and 20 mg/kg per day) [199,200,201,202,203] or coated with gold nanoparticles (300 mg/kg per day) [204]. Furthermore, after months of diabetes, the retina of these animals showed intense cell death, concentrated in the INL and the GCL, as demonstrated by various techniques [199,204]. The death induced by high glucose is probably due to increased production of pro-inflammatory cytokines (TNF-α, VEGF, INFγ, MCP-1, IL-6, IL1-β, and COX-2) and increased expression and activity of NF-kB [200,201,203,204].
An imbalance between ROS production and antioxidant defenses has been reported in the blood and retina of diabetic rats, with a decrease in SOD activity and an increase in the ratio of GSSH/GSH and 8-isoprostane levels, an indicator of oxidative stress [199,200,204]. Hyperglycemia also contributed to the rise in vessels in the ganglion cell layer, probably due to the increase in VEGF [204]. Resveratrol has been shown to reverse functional, morphological, and molecular effects in the retina of diabetic rats [199,200,201,202,204]. It appears that inhibition of NF-kB activity contributes to many of the neuroprotective effects of resveratrol in diabetes by reducing the levels of proteins associated with inflammation and cell death [200,201,204].
Data support the anti-inflammatory action of resveratrol and suggest that this is an essential mechanism for preventing high glucose-induced cell death. On the other hand, the neuroprotective activity of resveratrol may reflect a joint effort of different effects. In this sense, it has already been shown that resveratrol inhibits apoptosis in the retina of diabetic rats and Müller cell cultures by increasing microRNA 29b, which decreases pro-apoptotic proteins (SP1, activated caspase-3, and Bax) and increases anti-apoptotic (Bcl-2) proteins [205].
In vitro studies with endothelial cell lines showed that the protective effect of resveratrol also depends on the activation of sirtuins (SIRT), a classical target of this polyphenol. Bovine retinal capillary endothelial cells (BRECs) exposed to high glucose (30 mM) died by apoptosis induced by increased production of ROS. High glucose also reduces the expression of p-AMPK, SIRT-1, and PGC-1α, leading to cell death [206]. It has been demonstrated that the treatment with resveratrol (20 µM) prevented the reduction in protein levels of p-AMPK, SIRT-1, and PGC-1α, and blocked ROS-induced cell death [206]. A decrease in SIRT-1 levels was also observed in peripheral blood mononuclear cell cultures from patients with proliferative diabetic retinopathy and an increase in interleukin-17, a pro-inflammatory cytokine. The effects were reversed when these cells were exposed to resveratrol (10 µM) [207]. In human retinal microvascular endothelial cells (HRECs), exposure to high glucose (25 mM and 30 mM) resulted in decreased mRNA expression for mitochondrial SIRT-3, 4, and 5, which was accompanied by early endothelial senescence [208]. There was also oxidative damage and increased transition from endothelial to mesenchymal cells, contributing to tissue fibrosis resulting from increased glucose [208,209]. The phenotypic change from an endothelial to a mesenchymal cell depends on ROS production through NADPH oxidase activity via PKC. In this case, resveratrol treatment inhibited this phenotypic transition by inhibiting PKC activation [209]. However, whether the activation of AMPK/SIRT is also involved in this phenomenon is unknown. Therefore, more evidence is needed to answer this question.
Resveratrol has excellent therapeutic potential against abnormalities induced by high glucose in experimental models of diabetes. Its protective effects include neuronal, vascular endothelial, and pigmented epithelial cells by activating different pathways that may or may not be complementary. What would be the primary molecular target of resveratrol, although SIRT is a strong candidate? In any case, resveratrol has an anti-inflammatory effect combined with its antioxidant effect under high glucose conditions. The sum of these effects probably contributes to its overall protective effect. Table 4 summarizes the findings for the use of resveratrol in DR.
Lastly, Figure 3 shows a schematic diagram illustrating the cellular and molecular pathways in diabetic retinopathy leading to oxidative stress, inflammation, and apoptosis and how polyphenols act to downregulate these effects.

5.5. Lutein and Zeaxanthin

Lutein and zeaxanthin are carotenoids that have essential functions in eye health and are available as dietary supplements or foods [210]. In addition to potential antioxidant action, these compounds are found in the human retina and absorb wavelengths emitted by light, contributing to eye protection [211]. Individuals with type 2 DM have lower macular pigments, such as lutein and zeaxanthin, than healthy subjects [212].
Studies have demonstrated the benefits of lutein and zeaxanthin supplementation in diabetic retinopathy. Both short- and long-term supplementation with lutein and zeaxanthin in rodents reduced inflammatory biomarkers and vascular damage in the retina [213,214]. In addition, lutein and zeaxanthin supplementation reduced oxidative stress and inflammation, increased neuroprotection, decreased production of reactive oxygen species, increased GSH, peroxidase, and SOD levels in the retina of diabetic rats [215,216,217,218,219,220,221], as well as inhibited retinal histologic changes and increased retinal thickness [222]. The protective mechanisms of lutein and zeaxanthin in preventing eye diseases, such as cataracts, age-related macular degeneration, and diabetic retinopathy, have been reported [223].

6. Potential Interactions among Polyphenols, Probiotics, and Gut Microbiota in DR

The gastrointestinal tract is a primary site of action of polyphenols [224]. In this environment, polyphenols can interact beneficially with the gut microbiota and exert health benefits in the gut and beyond. However, the bidirectional relationship between gut microbiota and polyphenols is not straightforward. The beneficial health effects of polyphenols on the gut microbiota depend on numerous factors, such as their chemical structure, glycation pattern, and number of hydroxyl groups [224]. Similarly, gut dysbiosis, a condition characterized by an impairment of gut microbial composition and altered functional capacity that exceeds the resilience and resistance of the gut, can impair the metabolism and bioavailability of polyphenols.
Early evidence has suggested the role of gut dysbiosis in developing and maintaining DR [225,226,227]. Although previous studies have reported antioxidant and anti-inflammatory benefits with polyphenol therapy in DR, to our knowledge, there are no studies evaluating the role of microbiota in the reported effects of polyphenol therapy in DR. This gap should be explored in further clinical and animal studies.
Probiotics, prebiotics, and symbiotic consumption can alleviate intestinal dysbiosis, inflammation, and oxidative stress. In addition, selected probiotic strains can improve the metabolism of polyphenols. In contrast, polyphenols can positively modulate the composition of the gut microbiota, with effects like those of known prebiotics [228]. In addition, polyphenols can act as prebiotics and have been shown to regulate intestinal metabolism and immunity, besides influencing inflammatory pathways by altering T-cell functions, inhibiting mast cell degranulation, and reducing inflammatory cytokine responses [229,230,231,232].
In this way, the combined intake of probiotic strains and polyphenols has been considered an approach for a bi-directional improvement of their health-promoting effects [233] (Figure 2). Although an early randomized, controlled, crossover study found an attenuated early-phase insulin response after ingestion of probiotic fruit drinks with different polyphenol profiles in healthy young adults [234], whether combined therapy with pro-pre-symbiotic and polyphenols reduces oxidative stress, inflammation, and diabetic complications in DR remains to be elucidated.
Considering the early clinical evidence that probiotics, prebiotics, symbiotics, and polyphenols may alleviate glycemic dysfunction in DM conditions [235], while preclinical and clinical studies evaluating the effects of combined probiotics and polyphenols [236,237,238] in DR are not conducted, it is reasonable to suggest that DR patients may benefit from daily consumption of probiotics and dietary polyphenols for better glycemic control (Figure 4).

7. Conclusions

The potential of polyphenols as an efficient, non-invasive, and inexpensive treatment approach in the face of ophthalmologic pathologies is evident based on the results from the available literature discussed in the present review. It appears that dietary and/or nutritional supplementation with polyphenols, alone or in combination with probiotics, for example, may slow the progression of retinal diseases. The beneficial effects of the antioxidants studied seem to act indirectly rather than directly in controlling diabetic retinopathy through the activation of metabolic pathways responsible for increasing the antioxidant capacity of cells. Specifically in the retina, these antioxidant compounds stimulate several retinal anti-inflammatory pathways leading to inhibition/reduction in cell death caused by inflammation, AGEs, and hyperglycemia, in addition to improving membrane permeability and retinal cell structure. In addition, understanding the molecular mechanisms and how polyphenols affect the signaling pathways involved in neuroinflammation and oxidative stress may help develop effective therapeutic options for various retinopathies, including diabetic retinopathy. The importance of performing well-defined clinical trials to evaluate the efficacy, safety, dose range, and potential side effects, as well as chemical and pharmacokinetic analyses to improve the stability and bioavailability of these compounds, is emphasized.
Polyphenols are extensively metabolized in the gastrointestinal tract and liver, resulting in low systemic levels of active compounds reaching target organs, such as the retina. Strategies to increase the bioavailability of polyphenols, such as nanoformulations or combination with absorption enhancers, appear promising for this challenge. Nevertheless, the beneficial effects of polyphenols in various models of DR are evident and suggest that, despite the complexity of their molecular structure, they can cross the blood–retinal barrier. The literature lacks information on the bioavailability and metabolism of these polyphenols in the retina, so studies to address this issue are essential. One strategy would be to use high-performance liquid chromatography, which has been used to assess the levels of other drugs in the retina, to assess polyphenol levels in animal studies and specifically in retinal tissue. Furthermore, it is possible that the product of the metabolism of these polyphenols by the cytochrome P450 system in the retinal tissue has beneficial biological effects and that the protective effect is a synergistic response of these produced compounds. Another relevant point is correlating the doses administered in animal models with the dose and human consumption from different food sources.
On the other hand, the beneficial effects of polyphenols and other drugs of natural origin on the retina are much more associated with their chronic consumption than with punctual administration. In this sense, additional preclinical and clinical research is needed to discover the ideal dosage and treatment duration for optimal results, which is challenging to establish a uniform methodology. Furthermore, due to the multifactorial nature of DR, relying on a single polyphenol may not sufficiently inhibit the various risk factors involved. Therefore, a dietary intervention using a combination of polyphenols may offer advantages in preventing sight-threatening retinal diseases. By simultaneously suppressing multiple risk variables, this approach has the potential to be more effective in mitigating the complexities associated with DR. Another issue to consider is the interaction of polyphenols with medications. DR often requires multiple medications, such as insulin, oral hypoglycemic agents, or blood pressure medications. The interaction of polyphenols with various medications must be studied to ensure their safety and efficacy. It is also important to investigate the compatibility of polyphenols and their potential synergistic or antagonistic effects with other drugs.
Finally, genetic and environmental factors may cause individuals to respond differently to polyphenol treatment. Several factors, such as gut microbiota composition, genetic polymorphisms related to polyphenol metabolism, and individual dietary patterns, may influence the therapeutic benefits of polyphenols. Individual differences may require personalized approaches or targeted interventions to optimize outcomes. This information will help clarify whether polyphenol interventions could be an adjuvant and effective strategy for improving glycemic parameters and the quality of life of people with diabetes.

Author Contributions

Conceptualization, M.P.C.N.; investigation, G.B.F. and M.P.C.N.; writing—original draft preparation, G.B.F. and M.P.C.N.; writing—review and editing, M.R.M., K.d.C.C., R.B., A.M.P., H.R.M., D.E.d.A.L., J.L.d.B.A. and E.L.d.S.; project administration, M.P.C.N.; funding acquisition, E.L.d.S. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. American Diabetes Association. 5. Lifestyle Management: Standards of Medical Care in Diabetes—2019. Diabetes Care 2019, 42, 46–60. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Klein, R.; Knudtson, M.D.; Lee, K.E.; Gangnon, R.; Klein, B.E.K. The Wisconsin Epidemiologic Study of Diabetic Retinopathy XXII. The Twenty-Five-Year Progression of Retinopathy in Persons with Type 1 Diabetes. Ophthalmology 2008, 115, 1859–1868. [Google Scholar] [CrossRef] [Green Version]
  3. Buysschaert, M.; Hermans, M.P. Non-pharmacological management of type 2 diabetes. Acta Clin Belg. 2014, 59, 14–19. [Google Scholar] [CrossRef]
  4. IDF Diabetes Atlas. Available online: www.diabetesatlas.org (accessed on 2 March 2023).
  5. Klein, R.; Klein, B.E.K.; Moss, S.E.; Cruickshanks, K.J. The Wisconsin Epidemiologic Study of Diabetic Retinopathy: XVII. The 14-year incidence and progression of diabetic retinopathy and associated risk factors in type 1 diabetes. Ophthalmology 1998, 105, 1801–1815. [Google Scholar] [CrossRef]
  6. Olsen, B.S.; Sjølie, A.-K.; Hougaard, P.; Johannesen, J.; Borch-Johnsen, K.; Marinelli, K.; Thorsteinsson, B.; Pramming, S.; Mortensen, H.B. A 6-year nationwide cohort study of glycaemic control in young people with Type 1 diabetes: Risk markers for the development of retinopathy, nephropathy and neuropathy. J. Diabetes Its Complicat. 2000, 14, 295–300. [Google Scholar] [CrossRef] [PubMed]
  7. Van Leiden, H.A.; Dekker, J.M.; Moll, A.C.; Nijpels, G.; Heine, R.J.; Bouter, L.M.; Stehouwer, C.D.A.; Polak, B.C.P. Risk factors for incident retinopathy in a diabetic and nondiabetic population: The Hoorn study. Arch. Ophthalmol. 2003, 121, 245–251. [Google Scholar] [CrossRef] [PubMed]
  8. Klein, R.; Klein, B.E.K.; Moss, S.E.; Tasman, W. The relation of systemic hypertension to changes in the retinal vasculature: The Beaver Dam Eye Study. Trans. Am. Ophthalmol. Soc. 1997, 95, 329–350. [Google Scholar] [PubMed]
  9. Klein, R.; Klein, B.E.K.; Moss, S.E.; Cruickshanks, K.J. The Wisconsin Epidemiologic Study of Diabetic Retinopathy XV. The long-term incidence of macular edema. Ophthalmology 1995, 102, 7–16. [Google Scholar] [CrossRef]
  10. Surburg, H.; Panten, J. Common Fragrance and Flavor Materials: Preparation, Properties and Uses, 5th ed.; Wiley-Vch Verlag GmbH & Co. KGaA: Weinheim, Germany, 2006; pp. 1–305. [Google Scholar]
  11. Keenan, H.A.; Costacou, T.; Sun, J.K.; Doria, A.; Cavellerano, J.; Coney, J.; Orchard, T.J.; Aiello, L.P.; King, G.L. Clinical factors associated with resistance to microvascular complications in diabetic patients of extreme disease duration: The 50-year medalist study. Diabetes Care 2007, 30, 1995–1997. [Google Scholar] [CrossRef] [Green Version]
  12. Zhang, L.; Krzentowski, G.; Albert, A.; Lefebvre, P.J. Risk of developing retinopathy in Diabetes Control and Complications Trial type 1 diabetic patients with good or poor metabolic control. Diabetes Care 2001, 24, 1275–1279. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Donaghue, K.C.; Fairchild, J.M.; Craig, M.E.; Chan, A.K.; Hing, S.; Cutler, L.R.; Howard, N.J.; Silink, M. Do All Prepubertal Years of Diabetes Duration Contribute Equally to Diabetes Complications? Diabetes Care 2003, 26, 1224–1229. [Google Scholar] [CrossRef] [Green Version]
  14. Best, R.M.; Chakravarthy, U. Diabetic retinopathy in pregnancy. Br. J. Ophthalmol. 1997, 81, 249–251. [Google Scholar] [CrossRef] [Green Version]
  15. Klein, B.E.K.; Moss, S.E.; Klein, R. Effect of pregnancy on progression of diabetic retinopathy. Diabetes Care 1990, 13, 34–40. [Google Scholar] [CrossRef]
  16. Aiello, L.P.; Cahill, M.T.; Wong, J.S. Systemic considerations in the management of diabetic retinopathy. Am. J. Ophthalmol. 2001, 132, 760–776. [Google Scholar] [CrossRef]
  17. Chou, Y.; Ma, J.; Su, X.; Zhong, Y. Emerging insights into the relationship between hyperlipidemia and the risk of diabetic retinopathy. Lipids Health Dis. 2020, 19, 241. [Google Scholar] [CrossRef] [PubMed]
  18. Cruickshanks, K.J.; Ritter, L.L.; Klein, R.; Moss, S.E. The association of microalbuminuria with diabetic retinopathy. The Wisconsin Epidemiologic Study of Diabetic Retinopathy. Ophthalmology 1993, 100, 862–867. [Google Scholar] [CrossRef] [PubMed]
  19. Klein, R.; Moss, S.E.; Klein, B.E.K. Is gross proteinuria a risk factor for the incidence of proliferative diabetic retinopathy? Ophthalmology 1993, 100, 1140–1406. [Google Scholar] [CrossRef]
  20. Kusuhara, S.; Fukushima, Y.; Ogura, S.; Inoue, N.; Uemura, A. Pathophysiology of Diabetic Retinopathy: The Old and the New. Diabetes Metab. J. 2018, 42, 364–376. [Google Scholar] [CrossRef]
  21. Sen, S.; Ramasamy, K.; Sivaprasad, S. Indicators of Visual Prognosis in Diabetic Macular Oedema. J. Pers. Med. 2021, 11, 449. [Google Scholar] [CrossRef] [PubMed]
  22. Yau, J.W.Y.; 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] [Green Version]
  23. Shah, J.; Cheong, Z.Y.; Tan, B.; Wong, D.; Liu, X.; Chua, J. Dietary Intake and Diabetic Retinopathy: A Systematic Review of the Literature. Nutrients 2022, 14, 5021. [Google Scholar] [CrossRef]
  24. Millen, A.E.; Sahli, M.W.; Nie, J.; LaMonte, M.J.; Lutsey, P.L.; Klein, B.E.K.; Mares, J.A.; Meyers, K.J.; Andrews, C.A.; Klein, R. Adequate vitamin D status is associated with the reduced odds of prevalent diabetic retinopathy in African Americans and Caucasians. Cardiovasc. Diabetol. 2016, 15, 128. [Google Scholar] [CrossRef] [Green Version]
  25. Roy, M.S.; Janal, M.N. High caloric and sodium intakes as risk factors for progression of retinopathy in type 1 diabetes mellitus. Arch. Ophthalmol. 2010, 128, 33–39. [Google Scholar] [CrossRef] [Green Version]
  26. Millen, A.E.; Klein, R.; Folsom, A.R.; Stevens, J.; Palta, M.; Mares, J.A. Relation between intake of vitamins C and E and risk of diabetic retinopathy in the Atherosclerosis Risk in Communities Study. Am. J. Clin. Nutr. 2004, 79, 865–873. [Google Scholar] [CrossRef] [Green Version]
  27. Sasaki, M.; Kawasaki, R.; Rogers, S.; Man, R.E.K.; Itakura, K.; Xie, J.; Flood, V.; Tsubota, K.; Lamoureux, E.; Wang, J.J. The Associations of Dietary Intake of Polyunsaturated Fatty Acids with Diabetic Retinopathy in Well-Controlled Diabetes. Investig. Opthalmol. Vis. Sci. 2015, 56, 7473–7479. [Google Scholar] [CrossRef] [PubMed]
  28. Cundiff, D.K.; Nigg, C.R. Diet and Diabetic Retinopathy: Insights from the Diabetes Control and Complications Trial (DCCT). Medscape Gen. Med. 2005, 7, 3. [Google Scholar]
  29. Díaz-López, A.; Babio, N.; Martínez-González, M.A.; Corella, D.; Amor, A.J.; Fitó, M.; Estruch, R.; Arós, F.; Gómez-Gracia, E.; Fiol, M.; et al. Mediterranean Diet, Retinopathy, Nephropathy, and Microvascular Diabetes Complications: A Post Hoc Analysis of a Randomized Trial. Diabetes Care 2015, 38, 2134–2141. [Google Scholar] [CrossRef] [Green Version]
  30. American Diabetes Association. Standards of Medical Care in Diabetes—2013. Diabetes Care 2012, 36, S11–S66. [Google Scholar] [CrossRef] [Green Version]
  31. Franz, M.J.; Wylie-Rosett, J. The 2006 American Diabetes Association Nutrition Recommendations and Interventions for the Prevention and Treatment of Diabetes. Diabetes Spectr. 2007, 20, 49–52. [Google Scholar] [CrossRef] [Green Version]
  32. Bryl, A.; Mrugacz, M.; Falkowski, M.; Zorena, K. The Effect of Diet and Lifestyle on the Course of Diabetic Retinopathy-A Review of the Literature. Nutrients 2022, 14, 1252. [Google Scholar] [CrossRef]
  33. Guilbert, E.; Perry, R.; Whitmarsh, A.; Sauchelli, S. Short-term effectiveness of nutrition therapy to treat type 2 diabetes in low-income and middle-income countries: Systematic review and meta-analysis of randomised controlled trials. BMJ Open 2022, 12, e056108. [Google Scholar] [CrossRef]
  34. Tomita, Y.; Lee, D.; Tsubota, K.; Negishi, K.; Kurihara, T. Updates on the Current Treatments for Diabetic Retinopathy and Possibility of Future Oral Therapy. J. Clin. Med. 2021, 10, 4666. [Google Scholar] [CrossRef]
  35. Whitehead, M.; Wickremasinghe, S.; Osborne, A.; Van Wijngaarden, P.; Martin, K.R. Diabetic retinopathy: A complex pathophysiology requiring novel therapeutic strategies. Expert Opin. Biol. Ther. 2018, 18, 1257–1270. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Simó, R.; Hernández, C. New Insights into Treating Early and Advanced Stage Diabetic Retinopathy. Int. J. Mol. Sci. 2022, 23, 8513. [Google Scholar] [CrossRef]
  37. Aiello, L.P.; Avery, R.L.; Arrigg, P.G.; Keyt, B.A.; Jampel, H.D.; Shah, S.T.; Pasquale, L.R.; Thieme, H.; Iwamoto, M.A.; Park, J.E.; et al. Vascular endothelial growth factor in ocular fluid of patients with diabetic retinopathy and other retinal disorders. N. Engl. J. Med. 1994, 331, 1480–1487. [Google Scholar] [CrossRef] [PubMed]
  38. Vujosevic, S.; Simó, R. Local and Systemic Inflammatory Biomarkers of Diabetic Retinopathy: An Integrative Approach. Investig. Ophthalmol. Vis. Sci. 2017, 58, 68–75. [Google Scholar] [CrossRef] [PubMed]
  39. Rudrapal, M.; Khairnar, S.J.; Khan, J.; Bin Dukhyil, A.; Ansari, M.A.; Alomary, M.N.; Alshabrmi, F.M.; Palai, S.; Deb, P.K.; Devi, R. Dietary Polyphenols and Their Role in Oxidative Stress-Induced Human Diseases: Insights into Protective Effects, Antioxidant Potentials and Mechanism(s) of Action. Front. Pharmacol. 2022, 13, 806470. [Google Scholar] [CrossRef]
  40. Rana, A.; Samtiya, M.; Dhewa, T.; Mishra, V.; Aluko, R.E. Health benefits of polyphenols: A concise review. J. Food Biochem. 2022, 46, e14264. [Google Scholar] [CrossRef]
  41. Rasouli, H.; Farzaei, M.H.; Khodarahmi, R. Polyphenols and their benefits: A review. Int. J. Food Prop. 2017, 20, 1700–1741. [Google Scholar] [CrossRef] [Green Version]
  42. Alves, J.L.D.B.; De Sousa, V.P.; Neto, M.P.C.; Magnani, M.; Braga, V.D.A.; Da Costa-Silva, J.H.; Leandro, C.G.; Vidal, H.; Pirola, L. New Insights on the Use of Dietary Polyphenols or Probiotics for the Management of Arterial Hypertension. Front. Physiol. 2016, 7, 448. [Google Scholar] [CrossRef] [Green Version]
  43. Heavner, W.; Pevny, L. Eye Development and Retinogenesis. Cold Spring Harb. Perspect. Biol. 2012, 4, a008391. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Wässle, H. Parallel processing in the mammalian retina. Nat. Rev. Neurosci. 2004, 5, 747–757. [Google Scholar] [CrossRef]
  45. Hoon, M.; Okawa, H.; Della Santina, L.; Wong, R.O.L. Functional architecture of the retina: Development and disease. Prog. Retin. Eye Res. 2014, 42, 44–84. [Google Scholar] [CrossRef] [Green Version]
  46. Campos, V.S.; Calaza, K.C.; Adesse, D. Implications of TORCH Diseases in Retinal Development-Special Focus on Congenital Toxoplasmosis. Front. Cell. Infect. Microbiol. 2020, 10, 585727. [Google Scholar] [CrossRef] [PubMed]
  47. Newman, E.A. Glial cell regulation of neuronal activity and blood flow in the retina by release of gliotransmitters. Phil. Trans. R. Soc. B 2015, 370, 20140195. [Google Scholar] [CrossRef] [PubMed]
  48. Reichenbach, A.; Bringmann, A. Glia of the human retina. Glia 2020, 68, 768–796. [Google Scholar] [CrossRef] [PubMed]
  49. Ye, X.; Wang, Y.; Nathans, J. The Norrin/Frizzled4 signaling pathway in retinal vascular development and disease. Trends Mol. Med. 2010, 16, 417–425. [Google Scholar] [CrossRef] [Green Version]
  50. Coughlin, B.A.; Feenstra, D.J.; Mohr, S. Müller cells and diabetic retinopathy. Vis. Res. 2017, 139, 93–100. [Google Scholar] [CrossRef]
  51. Carpi-Santos, R.; Melo Reis, R.A.; Gomes, F.C.A.; Calaza, K.C. Contribution of Müller Cells in the Diabetic Retinopathy Development: Focus on Oxidative Stress and Inflammation. Antioxidants 2022, 11, 617. [Google Scholar] [CrossRef]
  52. Bandello, F.; Lattanzio, R.; Zucchiatti, I.; Del Turco, C. Pathophysiology and treatment of diabetic retinopathy. Acta Diabetol. 2013, 50, 1–20. [Google Scholar] [CrossRef] [PubMed]
  53. Mendonca, H.; Carpi-Santos, R.; Costa Calaza, K.; Blanco Martinez, A. Neuroinflammation and oxidative stress act in concert to promote neurodegeneration in the diabetic retina and optic nerve: Galectin-3 participation. Neural Regen. Res. 2020, 15, 625–635. [Google Scholar] [CrossRef] [PubMed]
  54. Honasoge, A.; Nudleman, E.; Smith, M.; Rajagopal, R. Emerging Insights and Interventions for Diabetic Retinopathy. Curr. Diabetes Rep. 2019, 19, 100. [Google Scholar] [CrossRef] [PubMed]
  55. Wang, W.; Lo, A.C.Y. Diabetic Retinopathy: Pathophysiology and Treatments. Int. J. Mol. Sci. 2018, 19, 1816. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Frank, R.N. Diabetic retinopathy. N. Engl. J. Med. 2004, 350, 48–58. [Google Scholar] [CrossRef]
  57. Wong, T.Y.; Cheung, C.M.G.; Larsen, M.; Sharma, S.; Simó, R. Diabetic retinopathy. Nat. Rev. Dis. Prim. 2016, 2, 16012. [Google Scholar] [CrossRef]
  58. Santiago, A.R.; Boia, R.; Aires, I.D.; Ambrósio, A.F.; Fernandes, R. Sweet Stress: Coping with Vascular Dysfunction in Diabetic Retinopathy. Front. Physiol. 2018, 9, 820. [Google Scholar] [CrossRef]
  59. Aiello, L.P.; Gardner, T.W.; King, G.L.; Blankenship, G.; Cavallerano, J.D.; Ferris, F.L.; Klein, R. Diabetic retinopathy. Diabetes Care 1998, 21, 143–156. [Google Scholar] [CrossRef]
  60. Lechner, J.; O’Leary, O.E.; Stitt, A.W. The pathology associated with diabetic retinopathy. Vis. Res. 2017, 139, 7–14. [Google Scholar] [CrossRef] [PubMed]
  61. Duh, E.J.; Sun, J.K.; Stitt, A.W. Diabetic retinopathy: Current understanding, mechanisms, and treatment strategies. JCI Insight 2017, 2, e93751. [Google Scholar] [CrossRef]
  62. Rosenblatt, B.; Benson, W. Vascular Disorders: Diabetic Retinopathy, 2nd ed.; Mosby: St. Louis, MO, USA, 2004. [Google Scholar]
  63. Romero-Aroca, P.; Baget-Bernaldiz, M.; Pareja-Rios, A.; Lopez-Galvez, M.; Navarro-Gil, R.; Verges, R. Diabetic Macular Edema Pathophysiology: Vasogenic versus Inflammatory. J. Diabetes Res. 2016, 2016, 2156273. [Google Scholar] [CrossRef] [Green Version]
  64. Sohn, E.H.; van Dijk, H.W.; Jiao, C.; Kok, P.H.B.; Jeong, W.; Demirkaya, N.; Garmager, A.; Wit, F.; Kucukevcilioglu, M.; van Velthoven, M.E.J.; et al. Retinal neurodegeneration may precede microvascular changes characteristic of diabetic retinopathy in diabetes mellitus. Proc. Natl. Acad. Sci. USA 2016, 113, E2655–E2664. [Google Scholar] [CrossRef] [Green Version]
  65. Lieth, E.; Gardner, T.W.; Barber, A.J.; Antonetti, D.A. Retinal neurodegeneration: Early pathology in diabetes. Clin. Exp. Ophthalmol. 2000, 28, 3–8. [Google Scholar] [CrossRef] [PubMed]
  66. Carrasco, E.; Hernández, C.; Miralles, A.; Huguet, P.; Farrés, J.; Simó, R. Lower somatostatin expression is an early event in diabetic retinopathy and is associated with retinal neurodegeneration. Diabetes Care 2007, 30, 2902–2908. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  67. Ali, T.K.; Matragoon, S.; Pillai, B.A.; Liou, G.I.; El-Remessy, A.B. Peroxynitrite mediates retinal neurodegeneration by inhibiting nerve growth factor survival signaling in experimental and human diabetes. Diabetes 2008, 57, 889–898. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  68. Barber, A.J. A new view of diabetic retinopathy: A neurodegenerative disease of the eye. Prog. Neuro-Psychopharmacol. Biol. Psychiatry 2003, 27, 283–290. [Google Scholar] [CrossRef]
  69. Carpineto, P.; Toto, L.; Aloia, R.; Ciciarelli, V.; Borrelli, E.; Vitacolonna, E.; Di Nicola, M.; Di Antonio, L.; Mastropasqua, R. Neuroretinal alterations in the early stages of diabetic retinopathy in patients with type 2 diabetes mellitus. Eye 2016, 30, 673–679. [Google Scholar] [CrossRef] [Green Version]
  70. Ng, D.S.; Chiang, P.P.; Tan, G.; Cheung, C.G.; Cheng, C.-Y.; Cheung, C.Y.; Wong, T.Y.; Lamoureux, E.L.; Ikram, M.K. Retinal ganglion cell neuronal damage in diabetes and diabetic retinopathy. Clin. Exp. Ophthalmol. 2016, 44, 243–250. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  71. Pinilla, I.; Idoipe, M.; Perdices, L.; Sanchez-Cano, A.; Acha, J.; Lopez-Galvez, M.I.; Cuenca, N.; Abecia, E.; Orduna-Hospital, E. Changes in total and inner retinal thicknesses in type 1 diabetes with no retinopathy after 8 years of follow-up. Retina 2020, 40, 1379–1386. [Google Scholar] [CrossRef]
  72. Zhu, T.P.; Ma, J.; Li, Y.H.; Zhang, Z. Association between retinal neuronal degeneration and visual function impairment in type 2 diabetic patients without diabetic retinopathy. Sci. China Life Sci. 2015, 58, 550–555. [Google Scholar] [CrossRef] [PubMed]
  73. Sinclair, S.H.; Schwartz, S.S. Diabetic Retinopathy—An Underdiagnosed and Undertreated Inflammatory, Neuro-Vascular Complication of Diabetes. Front. Endocrinol. 2019, 10, 843. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Kowluru, R.A.; Chan, P.S. Oxidative stress and diabetic retinopathy. Exp. Diabesity Res. 2007, 2007, 043603. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Brownlee, M. The pathobiology of diabetic complications: A unifying mechanism. Diabetes 2005, 54, 1615–1625. [Google Scholar] [CrossRef] [Green Version]
  76. Sullivan, L.B.; Chandel, N.S. Mitochondrial reactive oxygen species and cancer. Cancer Metab. 2014, 2, 99–116. [Google Scholar] [CrossRef] [Green Version]
  77. Klebanoff, S.J. Oxygen Metabolites from Phagocytes; Lippincott Williams & Wilkins: Philadelphia, PA, USA, 1999. [Google Scholar]
  78. Paul, S.; Ali, A.; Katare, R. Molecular complexities underlying the vascular complications of diabetes mellitus—A comprehensive review. J. Diabetes Its Complicat. 2020, 34, 107613. [Google Scholar] [CrossRef]
  79. Fukai, T.; Ushio-Fukai, M. Cross-Talk between NADPH Oxidase and Mitochondria: Role in ROS Signaling and Angiogenesis. Cells 2020, 9, 1849. [Google Scholar] [CrossRef]
  80. Peng, J.J.; Xiong, S.Q.; Ding, L.X.; Peng, J.; Xia, X.B. Diabetic retinopathy: Focus on NADPH oxidase and its potential as therapeutic target. Eur. J. Pharmacol. 2019, 853, 381–387. [Google Scholar] [CrossRef] [PubMed]
  81. Laddha, A.P.; Kulkarni, Y.A. NADPH oxidase: A membrane-bound enzyme and its inhibitors in diabetic complications. Eur. J. Pharmacol. 2020, 881, 173206. [Google Scholar] [CrossRef]
  82. Al-Mesallamy, H.O.; Hammad, L.N.; El-Mamoun, T.A.; Khalil, B.M. Role of advanced glycation end product receptors in the pathogenesis of diabetic retinopathy. J. Diabetes Complicat. 2011, 25, 168–174. [Google Scholar] [CrossRef] [PubMed]
  83. Milne, R.; Brownstein, S. Advanced glycation end products and diabetic retinopathy. Amino Acids 2013, 44, 1397–1407. [Google Scholar] [CrossRef] [PubMed]
  84. Chrysanthou, M.; Estruch, I.M.; Rietjens, I.M.C.M.; Wichers, H.J.; Hoppenbrouwers, T. In Vitro Methodologies to Study the Role of Advanced Glycation End Products (AGEs) in Neurodegeneration. Nutrients 2022, 14, 363. [Google Scholar] [CrossRef] [PubMed]
  85. Zhang, J.; Chen, C.; Wu, L.; Wang, Q.; Chen, J.; Zhang, S.; Chen, Z. Synoviolin inhibits the inflammatory cytokine secretion of Müller cells by reducing NLRP3. J. Mol. Endocrinol. 2022, 68, 125–136. [Google Scholar] [CrossRef]
  86. Kinuthia, U.M.; Wolf, A.; Langmann, T. Microglia and Inflammatory Responses in Diabetic Retinopathy. Front. Immunol. 2020, 11, 564077. [Google Scholar] [CrossRef]
  87. Subedi, L.; Lee, J.H.; Gaire, B.P.; Kim, S.Y. Sulforaphane Inhibits MGO-AGE-Mediated Neuroinflammation by Suppressing NF-κB, MAPK, and AGE–RAGE Signaling Pathways in Microglial Cells. Antioxidants 2020, 9, 792. [Google Scholar] [CrossRef]
  88. Li, W.; Liu, X.; Tu, Y.; Ding, D.; Yi, Q.; Sun, X.; Wang, Y.; Wang, K.; Zhu, M.; Mao, J. Dysfunctional Nurr1 promotes high glucose-induced Müller cell activation by up-regulating the NF-κB/NLRP3 inflammasome axis. Neuropeptides 2020, 82, 102057. [Google Scholar] [CrossRef]
  89. Chen, J.; Sun, Z.; Jin, M.; Tu, Y.; Wang, S.; Yang, X.; Chen, Q.; Zhang, X.; Han, Y.; Pi, R. Inhibition of AGEs/RAGE/Rho/ROCK pathway suppresses non-specific neuroinflammation by regulating BV2 microglial M1/M2 polarization through the NF-κB pathway. J. Neuroimmunol. 2017, 305, 108–114. [Google Scholar] [CrossRef] [PubMed]
  90. Li, X.; Liu, J.; Hoh, J.; Liu, J. Müller cells in pathological retinal angiogenesis. Transl. Res. 2019, 207, 96–106. [Google Scholar] [CrossRef]
  91. Mohammad, G.; AlSharif, H.M.; Siddiquei, M.M.; Ahmad, A.; Alam, K.; El-Asrar, A.M.A. Rho-associated protein kinase-1 mediates the regulation of inflammatory markers in diabetic retina and in retinal Müller cells. Ann. Clin. Lab. Sci. 2018, 48, 137–145. [Google Scholar]
  92. Mendonça, H.R.; Carvalho, J.N.A.; Abreu, C.A.; Mariano de Souza Aguiar dos Santos, D.; Carvalho, J.R.; Marques, S.A.; da Costa Calaza, K.; Martinez, A.M.B. Lack of Galectin-3 attenuates neuroinflammation and protects the retina and optic nerve of diabetic mice. Brain Res. 2018, 1700, 126–137. [Google Scholar] [CrossRef]
  93. Nahirnyj, A.; Livne-Bar, I.; Guo, X.; Sivak, J.M. ROS Detoxification and Proinflammatory Cytokines Are Linked by p38 MAPK Signaling in a Model of Mature Astrocyte Activation. PLoS ONE 2013, 8, e83049. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Zhao, Y.; Luo, C.; Chen, J.; Sun, Y.; Pu, D.; Lv, A.; Zhu, S.; Wu, J.; Wang, M.; Zhou, J.; et al. High glucose-induced complement component 3 up-regulation via RAGE-p38MAPK-NF-κB signalling in astrocytes: In vivo and in vitro studies. J. Cell. Mol. Med. 2018, 22, 6087–6098. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Costa, G.N.; Vindeirinho, J.; Cavadas, C.; Ambrósio, A.F.; Santos, P.F. Contribution of TNF receptor 1 to retinal neural cell death induced by elevated glucose. Mol. Cell. Neurosci. 2012, 50, 113–123. [Google Scholar] [CrossRef] [PubMed]
  96. McLeod, D.S.; Lefer, D.J.; Merges, C.; Lutty, G.A. Enhanced expression of intracellular adhesion molecule-1 and P-selectin in the diabetic human retina and choroid. Am. J. Pathol. 1995, 147, 642. [Google Scholar] [PubMed]
  97. Cambien, B.; Pomeranz, M.; Millet, M.A.; Rossi, B.; Schmid-Alliana, A. Signal transduction involved in MCP-1-mediated monocytic transendothelial migration. Blood 2001, 97, 359–366. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  98. Henkels, K.M.; Frondorf, K.; Gonzalez-Mejia, M.E.; Doseff, A.L.; Gomez-Cambronero, J. IL-8-induced neutrophil chemotaxis is mediated by Janus kinase 3 (JAK3). FEBS Lett. 2011, 585, 159–166. [Google Scholar] [CrossRef] [Green Version]
  99. Wang, L.; Zhou, X.; Yin, Y.; Mai, Y.; Wang, D.; Zhang, X. Hyperglycemia Induces Neutrophil Extracellular Traps Formation Through an NADPH Oxidase-Dependent Pathway in Diabetic Retinopathy. Front. Immunol. 2019, 9, 3076. [Google Scholar] [CrossRef] [Green Version]
  100. Phillipson, M.; Heit, B.; Colarusso, P.; Liu, L.; Ballantyne, C.M.; Kubes, P. Intraluminal crawling of neutrophils to emigration sites: A molecularly distinct process from adhesion in the recruitment cascade. J. Exp. Med. 2006, 203, 2569–2575. [Google Scholar] [CrossRef] [Green Version]
  101. Rossino, M.G.; Lulli, M.; Amato, R.; Cammalleri, M.; Dal Monte, M.; Casini, G. Oxidative Stress Induces a VEGF Autocrine Loop in the Retina: Relevance for Diabetic Retinopathy. Cells 2020, 9, 1452. [Google Scholar] [CrossRef]
  102. Antonetti, D.A.; Barber, A.J.; Hollinger, L.A.; Wolpert, E.B.; Gardner, T.W. Vascular endothelial growth factor induces rapid phosphorylation of tight junction proteins occludin and zonula occluden 1. A potential mechanism for vascular permeability in diabetic retinopathy and tumors. J. Biol. Chem. 1999, 274, 23463–23467. [Google Scholar] [CrossRef] [Green Version]
  103. Titchenell, P.M.; Lin, C.M.; Keil, J.M.; Sundstrom, J.M.; Smith, C.D.; Antonetti, D.A. Novel atypical PKC inhibitors prevent vascular endothelial growth factor-induced blood-retinal barrier dysfunction. Biochem. J. 2012, 446, 455–467. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Yan-Hwa, C.; Chang, C.L.; Hsu, H.F. Flavonoid content of several vegetables and their antioxidant activity. J. Sci. Food Agric. 2000, 80, 561–566. [Google Scholar] [CrossRef]
  105. Liyana-Pathirana, C.M.; Shahidi, F. Antioxidant properties of commercial soft and hard winter wheats (Triticum aestivum L.) and their milling fractions. J. Sci. Food Agric. 2006, 86, 477–485. [Google Scholar] [CrossRef]
  106. Luximon-Ramma, A.; Bahorun, T.; Crozier, A. Antioxidant actions and phenolic and vitamin C contents of common Mauritian exotic fruits. J. Sci. Food Agric. 2003, 83, 496–502. [Google Scholar] [CrossRef]
  107. de Araújo, D.S.M.; Brito, R.; Pereira-Figueiredo, D.; dos Santos-Rodrigues, A.; De Logu, F.; Nassini, R.; Zin, A.; Calaza, K.C. Retinal Toxicity Induced by Chemical Agents. Int. J. Mol. Sci. 2022, 23, 8182. [Google Scholar] [CrossRef] [PubMed]
  108. Sim, R.H.; Sirasanagandla, S.R.; Das, S.; Teoh, S.L. Treatment of Glaucoma with Natural Products and Their Mechanism of Action: An Update. Nutrients 2022, 14, 534. [Google Scholar] [CrossRef] [PubMed]
  109. Wang, M.; Sheng, K.-J.; Fang, J.-C.; Zhao, H.; Lu, S.-M.; Liu, Z.-Y.; Chen, B.-T. Redox signaling in diabetic retinopathy and opportunity for therapeutic intervention through natural products. Eur. J. Med. Chem. 2022, 244, 114829. [Google Scholar] [CrossRef] [PubMed]
  110. Fernandez-Gonzalez, P.; Mas-Sanchez, A.; Garriga, P. Polyphenols and Visual Health: Potential Effects on Degenerative Retinal Diseases. Molecules 2021, 26, 3407. [Google Scholar] [CrossRef] [PubMed]
  111. Shi, C.; Wang, P.; Airen, S.; Brown, C.; Liu, Z.; Townsend, J.H.; Wang, J.; Jiang, H. Nutritional and medical food therapies for diabetic retinopathy. Eye Vis. 2020, 7, 33. [Google Scholar] [CrossRef]
  112. Arulselvan, P.; Fard, M.T.; Tan, W.S.; Gothai, S.; Fakurazi, S.; Norhaizan, M.E.; Kumar, S.S. Role of Antioxidants and Natural Products in Inflammation. Oxid. Med. Cell. Longev. 2016, 2016, 5276130. [Google Scholar] [CrossRef] [Green Version]
  113. Kalt, W.; Hanneken, A.; Milbury, P.; Tremblay, F. Recent Research on Polyphenolics in Vision and Eye Health. J. Agric. Food Chem. 2010, 58, 4001–4007. [Google Scholar] [CrossRef]
  114. Brglez Mojzer, E.; Knez Hrnčič, M.; Škerget, M.; Knez, Ž.; Bren, U. Polyphenols: Extraction Methods, Antioxidative Action, Bioavailability and Anticarcinogenic Effects. Molecules 2016, 21, 901. [Google Scholar] [CrossRef]
  115. Aqil, F.; Munagala, R.; Jeyabalan, J.; Vadhanam, M.V. Bioavailability of phytochemicals and its enhancement by drug delivery systems. Cancer Lett. 2013, 334, 133–141. [Google Scholar] [CrossRef] [Green Version]
  116. Xiao, J. Stability of dietary polyphenols: It’s never too late to mend? Food Chem. Toxicol. 2018, 119, 3–5. [Google Scholar] [CrossRef] [PubMed]
  117. Sies, H. Polyphenols and health: Update and perspectives. Arch. Biochem. Biophys. 2010, 501, 2–5. [Google Scholar] [CrossRef] [PubMed]
  118. Coman, V.; Vodnar, D.C. Hydroxycinnamic acids and human health: Recent advances. J. Sci. Food Agric. 2020, 100, 483–499. [Google Scholar] [CrossRef]
  119. Marcia, M.M. Feruloylation in grasses: Current and future perspectives. Mol. Plant. 2009, 2, 861–872. [Google Scholar] [CrossRef]
  120. Zhao, Z.; Moghadasian, M.H. Chemistry, natural sources, dietary intake and pharmacokinetic properties of ferulic acid: A review. Food Chem. 2008, 109, 691–702. [Google Scholar] [CrossRef]
  121. Thapliyal, S.; Singh, T.; Handu, S.; Bisht, M.; Kumari, P.; Arya, P.; Srivastava, P.; Gandham, R. A Review on Potential Footprints of Ferulic Acid for Treatment of Neurological Disorders. Neurochem. Res. 2021, 46, 1043–1057. [Google Scholar] [CrossRef]
  122. Mancuso, C.; Santangelo, R. Ferulic acid: Pharmacological and toxicological aspects. Food Chem. Toxicol. 2014, 65, 185–195. [Google Scholar] [CrossRef]
  123. Taofiq, O.; González-Paramás, A.M.; Barreiro, M.F.; Ferreira, I.C.F.R.; McPhee, D.J. Hydroxycinnamic Acids and Their Derivatives: Cosmeceutical Significance, Challenges and Future Perspectives, a Review. Molecules 2017, 22, 281. [Google Scholar] [CrossRef] [Green Version]
  124. Kohno, M.; Musashi, K.; Ikeda, H.O.; Horibe, T.; Matsumoto, A.; Kawakami, K. Oral administration of ferulic acid or ethyl ferulate attenuates retinal damage in sodium iodate-induced retinal degeneration mice. Sci. Rep. 2020, 10, 8688. [Google Scholar] [CrossRef] [PubMed]
  125. Sun, X.; Sun, P.; Liu, L.; Jiang, P.; Li, Y. Ferulic acid attenuates microglia-mediated neuroinflammation in retinal degeneration. BMC Ophthalmol. 2021, 21, 13. [Google Scholar] [CrossRef]
  126. Xie, K.; Jin, B.; Zhu, H.; Zhou, P.; Du, L.; Jin, X. Ferulic acid (FA) protects human retinal pigment epithelial cells from H2O2-induced oxidative injuries. J. Cell. Mol. Med. 2020, 24, 13454–13462. [Google Scholar] [CrossRef]
  127. Zhu, D.; Zou, W.; Cao, X.; Xu, W.; Lu, Z.; Zhu, Y.; Hu, X.; Hu, J.; Zhu, Q. Ferulic acid attenuates high glucose-induced apoptosis in retinal pigment epithelium cells and protects retina in db/db mice. PeerJ 2022, 10, e13375. [Google Scholar] [CrossRef]
  128. Singh, S.S.B.; Patil, K.N. Trans-ferulic acid attenuates hyperglycemia-induced oxidative stress and modulates glucose metabolism by activating AMPK signaling pathway in vitro. J. Food Biochem. 2022, 46, e14038. [Google Scholar] [CrossRef]
  129. Liu, J.-L.; He, Y.-L.; Wang, S.; He, Y.; Wang, W.-Y.; Li, Q.-J.; Cao, X.-Y. Ferulic acid inhibits advanced glycation end products (AGEs) formation and mitigates the AGEs-induced inflammatory response in HUVEC cells. J Funct Foods 2018, 48, 19–26. [Google Scholar] [CrossRef]
  130. Romeo, A.; Musumeci, T.; Carbone, C.; Bonaccorso, A.; Corvo, S.; Lupo, G.; Anfuso, C.D.; Puglisi, G.; Pignatello, R. Ferulic Acid-Loaded Polymeric Nanoparticles for Potential Ocular Delivery. Pharmaceutics 2021, 13, 687. [Google Scholar] [CrossRef]
  131. Joe, B.; Vijaykumar, M.; Lokesh, B.R. Biological properties of curcumin-cellular and molecular mechanisms of action. Crit. Rev. Food Sci. Nutr. 2004, 44, 97–111. [Google Scholar] [CrossRef] [PubMed]
  132. Miłobȩdzka, J.; Kostanecki, S.V.; Lampe, V. Zur Kenntnis des Curcumins. Berichte Der Dtsch Chem Gesellschaft. 1910, 43, 2163–2170. [Google Scholar] [CrossRef] [Green Version]
  133. Xu, X.Y.; Meng, X.; Li, S.; Gan, R.Y.; Li, Y.; Li, H.B. Bioactivity, Health Benefits, and Related Molecular Mechanisms of Curcumin: Current Progress, Challenges, and Perspectives. Nutrients 2018, 10, 1553. [Google Scholar] [CrossRef] [Green Version]
  134. Mahesh, T.; Sri Balasubashini, M.M.; Menon, V.P. Photo-irradiated curcumin supplementation in streptozotocin-induced diabetic rats: Effect on lipid peroxidation. Therapie 2004, 59, 639–644. [Google Scholar] [CrossRef] [PubMed]
  135. Arun, N.; Nalini, N. Efficacy of turmeric on blood sugar and polyol pathway in diabetic albino rats. Plant Foods Hum. Nutr. 2002, 57, 41–52. [Google Scholar] [CrossRef]
  136. Gutierres, V.O.; Campos, M.L.; Arcaro, C.A.; Assis, R.P.; Baldan-Cimatti, H.M.; Peccinini, R.G.; Paula-Gomes, S.; Kettelhut, I.C.; Baviera, A.M.; Brunetti, I.L. Curcumin Pharmacokinetic and Pharmacodynamic Evidences in Streptozotocin-Diabetic Rats Support the Antidiabetic Activity to Be via Metabolite(s). Evid.-Based Complement. Altern. Med. 2015, 2015, 678218. [Google Scholar] [CrossRef] [Green Version]
  137. El-Moselhy, M.A.; Taye, A.; Sharkawi, S.S.; El-Sisi, S.F.I.; Ahmed, A.F. The antihyperglycemic effect of curcumin in high fat diet fed rats. Role of TNF-α and free fatty acids. Food Chem. Toxicol. 2011, 49, 1129–1140. [Google Scholar] [CrossRef]
  138. Chougala, M.B.; Bhaskar, J.J.; Rajan, M.G.R.; Salimath, P.V. Effect of curcumin and quercetin on lysosomal enzyme activities in streptozotocin-induced diabetic rats. Clin. Nutr. 2012, 31, 749–755. [Google Scholar] [CrossRef]
  139. Allegrini, D.; Raimondi, R.; Borgia, A.; Sorrentino, T.; Montesano, G.; Tsoutsanis, P.; Cancian, G.; Verma, Y.; De Rosa, F.P.; Romano, M.R. Curcumin in Retinal Diseases: A Comprehensive Review from Bench to Bedside. Int. J. Mol. Sci. 2022, 23, 3557. [Google Scholar] [CrossRef]
  140. Chandrasekaran, P.R.; Madanagopalan, V.G. Role of Curcumin in Retinal Diseases—A review. Graefe’s Arch. Clin. Exp. Ophthalmol. 2022, 260, 1457–1473. [Google Scholar] [CrossRef]
  141. Yang, J.; Miao, X.; Yang, F.-J.; Cao, J.-F.; Liu, X.; Fu, J.-L.; Su, G.-F. Therapeutic potential of curcumin in diabetic retinopathy (Review). Int. J. Mol. Med. 2021, 47, 75. [Google Scholar] [CrossRef]
  142. López-Malo, D.; Villarón-Casares, C.A.; Alarcón-Jiménez, J.; Miranda, M.; Díaz-Llopis, M.; Romero, F.J.; Villar, V.M. Curcumin as a Therapeutic Option in Retinal Diseases. Antioxidants 2020, 9, 48. [Google Scholar] [CrossRef] [Green Version]
  143. Farajipour, H.; Rahimian, S.; Taghizadeh, M. Curcumin: A new candidate for retinal disease therapy? J. Cell. Biochem. 2019, 120, 6886–6893. [Google Scholar] [CrossRef]
  144. Zhang, J.; Qin, Y.; Martinez, M.; Flores-Bellver, M.; Rodrigues, M.; Dinabandhu, A.; Cao, X.; Deshpande, M.; Qin, Y.; Aparicio-Domingo, S.; et al. HIF-1α and HIF-2α redundantly promote retinal neovascularization in patients with ischemic retinal disease. J. Clin. Investig. 2021, 131, e139202. [Google Scholar] [CrossRef]
  145. Rattner, A.; Williams, J.; Nathans, J. Roles of HIFs and VEGF in angiogenesis in the retina and brain. J. Clin. Investig. 2019, 129, 3807–3820. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  146. Zhang, D.; Lv, F.L.; Wang, G.H. Effects of HIF-1α on diabetic retinopathy angiogenesis and VEGF expression. Eur. Rev. Med. Pharmacol. Sci. 2018, 22, 5071–5076. [Google Scholar] [CrossRef] [PubMed]
  147. Bae, M.-K.; Kim, S.-H.; Jeong, J.-W.; Lee, Y.M.; Kim, H.-S.; Kim, S.-R.; Yun, I.; Bae, S.-K.; Kim, K.-W. Curcumin inhibits hypoxia-induced angiogenesis via down-regulation of HIF-1. Oncol. Rep. 2006, 15, 1557–1562. [Google Scholar] [CrossRef] [PubMed]
  148. Choi, H.; Chun, Y.S.; Kim, S.W.; Kim, M.S.; Park, J.W. Curcumin inhibits hypoxia-inducible factor-1 by degrading aryl hydrocarbon receptor nuclear translocator: A mechanism of tumor growth inhibition. Mol. Pharmacol. 2006, 70, 1664–1671. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  149. Carpi-Santos, R.; Ferreira, M.J.; Netto, A.D.P.; Giestal-De-Araujo, E.; Ventura, A.L.M.; Cossenza, M.; Calaza, K.C. Early changes in system xc− and glutathione in the retina of diabetic rats. Exp. Eye Res. 2016, 146, 35–42. [Google Scholar] [CrossRef]
  150. Hammes, H.P. Diabetic retinopathy: Hyperglycaemia, oxidative stress and beyond. Diabetologia 2018, 61, 29–38. [Google Scholar] [CrossRef] [Green Version]
  151. Mokgalaboni, K.; Ntamo, Y.; Ziqubu, K.; Nyambuya, T.M.; Nkambule, B.B.; Mazibuko-Mbeje, S.E.; Gabuza, K.B.; Chellan, N.; Tiano, L.; Dludla, P.V. Curcumin supplementation improves biomarkers of oxidative stress and inflammation in conditions of obesity, type 2 diabetes and NAFLD: Updating the status of clinical evidence. Food Funct. 2021, 12, 12235–12249. [Google Scholar] [CrossRef] [PubMed]
  152. Tabrizi, R.; Vakili, S.; Akbari, M.; Mirhosseini, N.; Lankarani, K.B.; Rahimi, M.; Mobini, M.; Jafarnejad, S.; Vahedpoor, Z.; Asemi, Z. The effects of curcumin-containing supplements on biomarkers of inflammation and oxidative stress: A systematic review and meta-analysis of randomized controlled trials. Phytother. Res. 2019, 33, 253–262. [Google Scholar] [CrossRef]
  153. Nawaz, M.I.; Alhomida, A.S.; Ola, M.S. The Potential Beneficial Effects of Curcumin in Diabetic Retinopathy. Curcumin Neurol. Psychiatr. Disord. Neurochem. Pharm. Prop. 2019, 22, 401–417. [Google Scholar] [CrossRef]
  154. Pradhan, D.; Dasmohapatra, T.; Tripathy, G. Pharmacognostic Evaluation of Curcumin on Diabetic Retinopathy in Alloxan-induced Diabetes through NF-KB and Brn3a Related Mechanism. Pharmacogn. J. 2018, 10, 324–332. [Google Scholar] [CrossRef] [Green Version]
  155. Chen, A.; Xu, J.; Johnson, A.C. Curcumin inhibits human colon cancer cell growth by suppressing gene expression of epidermal growth factor receptor through reducing the activity of the transcription factor Egr-1. Oncogene 2006, 25, 278–287. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  156. Huang, J.; Yi, Q.; You, Y.; Chen, Y.; Niu, T.; Li, Y.; Zhang, J.; Ji, X.; Xu, G.; Zou, W.; et al. Curcumin suppresses oxidative stress via regulation of ROS/NF-κB signaling pathway to protect retinal vascular endothelial cell in diabetic retinopathy. Mol. Cell. Toxicol. 2021, 17, 367–376. [Google Scholar] [CrossRef]
  157. Ran, Z.; Zhang, Y.; Wen, X.; Ma, J. Curcumin inhibits high glucose-induced inflammatory injury in human retinal pigment epithelial cells through the ROS-PI3K/AKT/mTOR signaling pathway. Mol. Med. Rep. 2019, 19, 1024–1031. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  158. Venugopal, R.; Jaiswal, A.K. Nrf2 and Nrf1 in association with Jun proteins regulate antioxidant response element-mediated expression and coordinated induction of genes encoding detoxifying enzymes. Oncogene 1998, 17, 3145–3156. [Google Scholar] [CrossRef] [Green Version]
  159. Itoh, K.; Chiba, T.; Takahashi, S.; Ishii, T.; Igarashi, K.; Katoh, Y.; Oyake, T.; Hayashi, N.; Satoh, K.; Hatayama, I.; et al. An Nrf2/small Maf heterodimer mediates the induction of phase II detoxifying enzyme genes through antioxidant response elements. Biochem. Biophys. Res. Commun. 1997, 236, 313–322. [Google Scholar] [CrossRef]
  160. 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]
  161. Bucolo, C.; Drago, F.; Maisto, R.; Romano, G.L.; D’Agata, V.; Maugeri, G.; Giunta, S. Curcumin prevents high glucose damage in retinal pigment epithelial cells through ERK1/2-mediated activation of the Nrf2/HO-1 pathway. J. Cell. Physiol. 2019, 234, 17295–17304. [Google Scholar] [CrossRef] [PubMed]
  162. Xie, T.; Chen, X.; Chen, W.; Huang, S.; Peng, X.; Tian, L.; Wu, X.; Huang, Y. Curcumin is a Potential Adjuvant to Alleviates Diabetic Retinal Injury via Reducing Oxidative Stress and Maintaining Nrf2 Pathway Homeostasis. Front. Pharmacol. 2021, 12, 796565. [Google Scholar] [CrossRef]
  163. Ota, N.; Soga, S.; Murase, T.; Shimotoyodome, A.; Hase, T. Consumption of Coffee Polyphenols Increases Fat Utilization in Humans. J. Health Sci. 2010, 56, 745–751. [Google Scholar] [CrossRef] [Green Version]
  164. Shen, N.; Wang, T.; Gan, Q.; Liu, S.; Wang, L.; Jin, B. Plant flavonoids: Classification, distribution, biosynthesis, and antioxidant activity. Food Chem. 2022, 383, 132531. [Google Scholar] [CrossRef]
  165. Nijveldt, R. Flavonoids: A review of probable mechanism of action and potential applications. Am. J. Clin. Nutr. 2001, 74, 418–425. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  166. Zhou, D.; Bai, Z.; Guo, T.; Li, J.; Li, Y.; Hou, Y.; Chen, G.; Li, N. Dietary flavonoids and human top-ranked diseases: The perspective of in vivo bioactivity and bioavailability. Trends Food Sci. Technol. 2022, 120, 374–386. [Google Scholar] [CrossRef]
  167. Du Toit, R.; Volsteedt, Y.; Apostolides, Z. Comparison of the antioxidant content of fruits, vegetables and teas measured as vitamin C equivalents. Toxicology 2001, 166, 63–69. [Google Scholar] [CrossRef]
  168. Sato, T.; Miyata, G. The nutraceutical benefit, part I: Green tea. Nutrition 2000, 16, 315–317. [Google Scholar] [CrossRef] [PubMed]
  169. Gomez-Cabrera, M.C.; Ferrando, B.; Brioche, T.; Sanchis-Gomar, F.; Viña, J. Exercise and antioxidant supplements in the elderly. J. Sport Health Sci. 2013, 2, 94–100. [Google Scholar] [CrossRef] [Green Version]
  170. Gosslau, A.; Chen, K.Y. Nutraceuticals, apoptosis, and disease prevention. Nutrition 2004, 20, 95–102. [Google Scholar] [CrossRef]
  171. Saito, S.T.; Welzel, A.; Suyenaga, E.S.; Bueno, F. A method for fast determination of epigallocatechin gallate (EGCG), epicatechin (EC), catechin (C) and caffeine (CAF) in green tea using HPLC. Food Sci. Technol. 2006, 26, 394–400. [Google Scholar] [CrossRef] [Green Version]
  172. Anderson, R.A.; Polansky, M.M. Tea Enhances Insulin Activity. J. Agric. Food Chem. 2002, 50, 7182–7186. [Google Scholar] [CrossRef]
  173. Sano, J.; Inami, S.; Seimiya, K.; Ohba, T. Effects of Green Tea Intake on the Development. Circ. J. 2004, 68, 665–670. [Google Scholar] [CrossRef] [Green Version]
  174. Suzuki, M.; Tabuchi, M.; Ikeda, M.; Umegaki, K.; Tomita, T. Protective effects of green tea catechins on cerebral ischemic damage. Med. Sci. Monit. 2004, 10, 166–174. [Google Scholar]
  175. Kumar, B.; Gupta, S.K.; Nag, T.C.; Srivastava, S.; Saxena, R. Green tea prevents hyperglycemia-induced retinal oxidative stress and inflammation in streptozotocin-induced diabetic rats. Ophthalmic. Res. 2012, 47, 103–108. [Google Scholar] [CrossRef]
  176. Silva, K.C.; Rosales, M.A.B.; Hamassaki, D.E.; Saito, K.C.; Faria, A.M.; Ribeiro, P.A.O.; de Faria, J.B.L.; De Faria, J.M.L. Green tea is neuroprotective in diabetic retinopathy. Investig. Ophthalmol. Vis. Sci. 2013, 54, 1325–1336. [Google Scholar] [CrossRef] [Green Version]
  177. Chan, C.M.; Huang, J.H.; Chiang, H.S.; Wu, B.; Lin, H.H.; Hong, J.Y.; Hung, C.F. Effects of (−)-epigallocatechin gallate on rpe cell migration and adhesion. Mol. Vis. 2010, 16, 586–595. [Google Scholar] [PubMed]
  178. Wang, L.; Sun, X.; Zhu, M.; Du, J.; Xu, J.; Qin, X.; Xu, X.; Song, E. Epigallocatechin-3-gallate stimulates autophagy and reduces apoptosis levels in retinal Müller cells under high-glucose conditions. Exp. Cell Res. 2019, 380, 149–158. [Google Scholar] [CrossRef] [PubMed]
  179. Duan, Y.; Santiago, F.E.M.; dos Reis, A.R.; de Figueiredo, M.A.; Zhou, S.; Thannhauser, T.W.; Li, L. Genotypic variation of flavonols and antioxidant capacity in broccoli. Food Chem. 2021, 15, 127997. [Google Scholar] [CrossRef] [PubMed]
  180. Martínez-Lüscher, J.; Brillante, L.; Kurtural, S.K. Flavonol profile is a reliable indicator to assess canopy architecture and the exposure of red wine grapes to solar radiation. Front. Plant Sci. 2019, 10, 10. [Google Scholar] [CrossRef] [Green Version]
  181. da Costa, P.C.T.; de Souza, E.L.; Lacerda, D.C.; Neto, J.P.R.C.; de Sales, L.C.S.; Luis, C.C.S.; Pontes, P.B.; Neto, M.P.C.; Alves, J.L.d.B. Evidence for Quercetin as a Dietary Supplement for the Treatment of Cardio-Metabolic Diseases in Pregnancy: A Review in Rodent Models. Foods 2022, 11, 2772. [Google Scholar] [CrossRef]
  182. Hosseini, A.; Razavi, B.M.; Banach, M.; Hosseinzadeh, H. Quercetin and metabolic syndrome: A review. Phytother. Res. 2021, 35, 5352–5364. [Google Scholar] [CrossRef] [PubMed]
  183. Tang, S.M.; Deng, X.T.; Zhou, J.; Li, Q.P.; Ge, X.X.; Miao, L. Pharmacological basis and new insights of quercetin action in respect to its anti-cancer effects. Biomed. Pharmacother. 2020, 121, 109604. [Google Scholar] [CrossRef]
  184. Bungau, S.; Abdel-Daim, M.M.; Tit, D.M.; Ghanem, E.; Sato, S.; Maruyama-Inoue, M.; Yamane, S.; Kadonosono, K. Health Benefits of Polyphenols and Carotenoids in Age-Related Eye Diseases. Oxid. Med. Cell. Longev. 2019, 2019, 9783429. [Google Scholar] [CrossRef] [PubMed]
  185. Arikan, S.; Erşan, I.; Karaca, T.; Kara, S.; Gencer, B.; Karaboga, I.; Hasan Ali, T. Quercetin protects the retina by reducing apoptosis due to ischemia-reperfusion injury in a rat model. Arq. Bras. Oftalmol. 2015, 78, 100–104. [Google Scholar] [CrossRef] [Green Version]
  186. Cheng, S.C.; Huang, W.C.; Pang, J.H.S.; Wu, Y.H.; Cheng, C.Y. Quercetin inhibits the production of il-1β-induced inflammatory cytokines and chemokines in arpe-19 cells via the mapk and nf-κb signaling pathways. Int. J. Mol. Sci. 2019, 20, 2957. [Google Scholar] [CrossRef] [Green Version]
  187. Chai, G.R.; Liu, S.; Yang, H.W.; Chen, X.L. Quercetin protects against diabetic retinopathy in rats by inducing heme oxygenase-1 expression. Neural Regen. Res. 2021, 16, 1344–1350. [Google Scholar] [CrossRef] [PubMed]
  188. Li, R.; Chen, L.; Yao, G.M.; Yan, H.L.; Wang, L. Effects of quercetin on diabetic retinopathy and its association with NLRP3 inflammasome and autophagy. Int. J. Ophthalmol. 2021, 14, 42–49. [Google Scholar] [CrossRef] [PubMed]
  189. Chen, B.; He, T.; Xing, Y.; Cao, T. Effects of quercetin on the expression of MCP-1, MMP-9 and VEGF in rats with diabetic retinopathy. Exp. Ther. Med. 2017, 14, 6022–6026. [Google Scholar] [CrossRef] [Green Version]
  190. Ola, M.S.; Ahmed, M.M.; Shams, S.; Al-Rejaie, S.S. Neuroprotective effects of quercetin in diabetic rat retina. Saudi J. Biol. Sci. 2017, 24, 1186–1194. [Google Scholar] [CrossRef] [PubMed]
  191. Chang, Y.-Y.; Lee, Y.-J.; Hsu, M.-Y.; Wang, M.; Tsou, S.-C.; Chen, C.-C.; Lin, J.-A.; Hsiao, Y.-P.; Lin, H.-W. Protective Effect of Quercetin on Sodium Iodate-Induced Retinal Apoptosis through the Reactive Oxygen Species-Mediated Mitochondrion-Dependent Pathway. Int. J. Mol. Sci. 2021, 22, 4056. [Google Scholar] [CrossRef] [PubMed]
  192. Ortega, J.T.; Jastrzebska, B. Neuroinflammation as a therapeutic target in retinitis pigmentosa and quercetin as its potential modulator. Pharmaceutics 2021, 13, 1935. [Google Scholar] [CrossRef]
  193. Tian, B.; Liu, J. Resveratrol: A review of plant sources, synthesis, stability, modification and food application. J. Sci. Food Agric. 2020, 100, 1392–1404. [Google Scholar] [CrossRef]
  194. Zhang, L.-X.; Li, C.-X.; Kakar, M.U.; Khan, M.S.; Wu, P.-F.; Amir, R.M.; Dai, D.-F.; Naveed, M.; Li, Q.-Y.; Saeed, M.; et al. Resveratrol (RV): A pharmacological review and call for further research. Biomed. Pharmacother. 2021, 143, 112164. [Google Scholar] [CrossRef]
  195. Ahmad, I.; Hoda, M. Attenuation of diabetic retinopathy and neuropathy by resveratrol: Review on its molecular mechanisms of action. Life Sci. 2020, 245, 117350. [Google Scholar] [CrossRef] [PubMed]
  196. Toro, M.D.; Nowomiejska, K.; Avitabile, T.; Rejdak, R.; Tripodi, S.; Porta, A.; Reibaldi, M.; Figus, M.; Posarelli, C.; Fiedorowicz, M. Effect of resveratrol on in vitro and in vivo models of diabetic retinophathy: A systematic review. Int. J. Mol. Sci. 2019, 20, 3503. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  197. Galiniak, S.; Aebisher, D.; Bartusik-Aebisher, D. Health benefits of resveratrol administration. Acta Biochim. Pol. 2019, 66, 13–21. [Google Scholar] [CrossRef] [Green Version]
  198. Huang, D.D.; Shi, G.; Jiang, Y.; Yao, C.; Zhu, C. A review on the potential of Resveratrol in prevention and therapy of diabetes and diabetic complications. Biomed. Pharmacother. 2020, 125, 109767. [Google Scholar] [CrossRef] [PubMed]
  199. Kim, Y.H.; Kim, Y.S.; Kang, S.S.; Cho, G.J.; Choi, W.S. Resveratrol inhibits neuronal apoptosis and elevated Ca2+/calmodulin-dependent protein kinase II activity in diabetic mouse retina. Diabetes 2010, 59, 1825–1835. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  200. Soufi, F.G.; Mohammad-nejad, D.; Ahmadieh, H. Resveratrol improves diabetic retinopathy possibly through oxidative stress—Nuclear factor κB—Apoptosis pathway. Pharmacol. Rep. 2012, 64, 1505–1514. [Google Scholar] [CrossRef]
  201. Ghadiri Soufi, F.; Arbabi-Aval, E.; Rezaei Kanavi, M.; Ahmadieh, H. Anti-inflammatory properties of resveratrol in the retinas of type 2 diabetic rats. Clin. Exp. Pharmacol. Physiol. 2015, 42, 63–68. [Google Scholar] [CrossRef]
  202. Zeng, K.; Yang, N.; Wang, D.; Li, S.; Ming, J.; Wang, J.; Yu, X.; Song, Y.; Zhou, X.; Yang, Y. Resveratrol Prevents Retinal Dysfunction by Regulating Glutamate Transporters, Glutamine Synthetase Expression and Activity in Diabetic Retina. Neurochem. Res. 2016, 41, 1050–1064. [Google Scholar] [CrossRef] [PubMed]
  203. Chen, Y.; Meng, J.; Li, H.; Wei, H.; Bi, F.; Liu, S.; Tang, K.; Guo, H.; Liu, W. Resveratrol exhibits an effect on attenuating retina inflammatory condition and damage of diabetic retinopathy via PON1. Exp. Eye Res. 2019, 181, 356–366. [Google Scholar] [CrossRef]
  204. Dong, Y.; Wan, G.; Yan, P.; Qian, C.; Li, F.; Peng, G. Fabrication of resveratrol coated gold nanoparticles and investigation of their effect on diabetic retinopathy in streptozotocin induced diabetic rats. J. Photochem. Photobiol. B 2019, 195, 51–57. [Google Scholar] [CrossRef]
  205. Zeng, K.; Wang, Y.; Yang, N.; Wang, D.; Li, S.; Ming, J.; Wang, J.; Yu, X.; Song, Y.; Zhou, X.; et al. Resveratrol Inhibits Diabetic-Induced Müller Cells Apoptosis through MicroRNA-29b/Specificity Protein 1 Pathway. Mol. Neurobiol. 2017, 54, 4000–4014. [Google Scholar] [CrossRef] [PubMed]
  206. Li, J.; Yu, S.; Ying, J.; Shi, T.; Wang, P. Resveratrol Prevents ROS-Induced Apoptosis in High Glucose-Treated Retinal Capillary Endothelial Cells via the Activation of AMPK/Sirt1/PGC-1 α Pathway. Oxid. Med. Cell. Longev. 2017, 2017, 7584691. [Google Scholar] [CrossRef] [Green Version]
  207. Liu, S.; Lin, Y.; Liu, X. Protective effects of SIRT1 in patients with proliferative diabetic retinopathy via the inhibition of IL-17 expression. Exp. Ther. Med. 2016, 11, 257–262. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  208. Liu, J.; Chen, S.; Biswas, S.; Nagrani, N.; Chu, Y.; Chakrabarti, S.; Feng, B. Glucose-induced oxidative stress and accelerated aging in endothelial cells are mediated by the depletion of mitochondrial SIRTs. Physiol. Rep. 2020, 8, e14331. [Google Scholar] [CrossRef] [PubMed]
  209. Giordo, R.; Nasrallah, G.K.; Posadino, A.M.; Galimi, F.; Capobianco, G.; Eid, A.H.; Pintus, G. Resveratrol-Elicited PKC Inhibition Counteracts NOX-Mediated Endothelial to Mesenchymal Transition in Human Retinal Endothelial Cells Exposed to High Glucose. Antioxidants 2021, 10, 224. [Google Scholar] [CrossRef]
  210. Eggersdorfer, M.; Wyss, A. Carotenoids in human nutrition and health. Arch. Biochem. Biophys. 2018, 652, 18–26. [Google Scholar] [CrossRef]
  211. Barker, F.M.; Snodderly, D.M.; Johnson, E.J.; Schalch, W.; Koepcke, W.; Gerss, J.; Neuringer, M. Nutritional Manipulation of Primate Retinas, V: Effects of lutein, zeaxanthin, and n-3 fatty acids on retinal sensitivity to blue-light-induced damage. Investig. Ophthalmol. Vis. Sci. 2011, 52, 3934–3942. [Google Scholar] [CrossRef] [Green Version]
  212. Scanlon, G.; Loughman, J.; Farrell, D.; Mccartney, D. A review of the putative causal mechanisms associated with lower macular pigment in diabetes mellitus. Nutr. Res. Rev. 2019, 32, 247–264. [Google Scholar] [CrossRef] [Green Version]
  213. Kamoshita, M.; Toda, E.; Osada, H.; Narimatsu, T.; Kobayashi, S.; Tsubota, K.; Ozawa, Y. Lutein acts via multiple antioxidant pathways in the photo-stressed retina. Sci. Rep. 2016, 6, 30226. [Google Scholar] [CrossRef] [PubMed]
  214. Wang, W.; Tam, K.C.; NG, T.C.; Goit, R.K.; Chan, K.L.S.; Lo, A.C.Y. Long-term lutein administration attenuates retinal inflammation and functional deficits in early diabetic retinopathy using the Ins2Akita/+ mice. BMJ Open Diabetes Res. Care 2020, 8, e001519. [Google Scholar] [CrossRef]
  215. Sasaki, M.; Ozawa, Y.; Kurihara, T.; Kubota, S.; Yuki, K.; Noda, K.; Kobayashi, S.; Ishida, S.; Tsubota, K. Neurodegenerative influence of oxidative stress in the retina of a murine model of diabetes. Diabetologia 2010, 53, 971–979. [Google Scholar] [CrossRef] [Green Version]
  216. Kowluru, R.A.; Zhong, Q.; Santos, J.M.; Thandampallayam, M.; Putt, D.; Gierhart, D.L. Beneficial effects of the nutritional supplements on the development of diabetic retinopathy. Nutr. Metab. 2014, 11, 8. [Google Scholar] [CrossRef] [Green Version]
  217. Arnal, E.; Miranda, M.; Johnsen-Soriano, S.; Alvarez-Nölting, R.; Díaz-Llopis, M.; Araiz, J.; Cervera, E.; Bosch-Morell, F.; Romero, F.J. Beneficial Effect of Docosahexanoic Acid and Lutein on Retinal Structural, Metabolic, and Functional Abnormalities in Diabetic Rats. Curr. Eye Res. 2009, 34, 928–938. [Google Scholar] [CrossRef]
  218. Kowluru, R.A.; Menon, B.; Gierhart, D.L. Beneficial Effect of Zeaxanthin on Retinal Metabolic Abnormalities in Diabetic Rats. Investig. Ophthalmol. Vis. Sci. 2008, 49, 1645–1651. [Google Scholar] [CrossRef] [PubMed]
  219. Tang, L.; Zhang, Y.; Jiang, Y.; Willard, L.; Ortiz, E.; Wark, L.; Medeiros, D.; Lin, D. Dietary wolfberry ameliorates retinal structure abnormalities in db/db mice at the early stage of diabetes. Exp. Biol. Med. 2011, 236, 1051–1063. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  220. Muriach, M.; Bosch-Morell, F.; Alexander, G.; Blomhoff, R.; Barcia, J.; Arnal, E.; Almansa, I.; Romero, F.J.; Miranda, M. Lutein effect on retina and hippocampus of diabetic mice. Free Radic. Bio. Med. 2006, 41, 979–984. [Google Scholar] [CrossRef] [PubMed]
  221. Yu, H.; Wark, L.; Ji, H.; Willard, L.; Jaing, Y.; Han, J.; He, H.; Ortiz, E.; Zhang, Y.; Medeiros, D.M. Dietary wolfberry upregulates carotenoid metabolic genes and enhances mitochondrial biogenesis in the retina of db/db diabetic mice. Mol. Nutr. Food Res. 2013, 57, 1158–1169. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  222. Kern, T.S.; Engerman, R.L. Pharmacological Inhibition of Diabetic Retinopathy. Diabetes 2001, 50, 1636–1642. [Google Scholar] [CrossRef] [Green Version]
  223. Johra, F.T.; Bepari, A.K.; Bristy, A.T.; Reza, H.M. A Mechanistic Review of β-Carotene, Lutein, and Zeaxanthin in Eye Health and Disease. Antioxidantes 2020, 9, 1046. [Google Scholar] [CrossRef] [PubMed]
  224. Domínguez-Avila, J.A.; Villa-Rodriguez, J.A.; Montiel-Herrera, M.; Pacheco-Ordaz, R.; Roopchand, D.E.; Venema, K.; González-Aguilar, G.A. Phenolic Compounds Promote Diversity of Gut Microbiota and Maintain Colonic Health. Dig. Dis. Sci. 2021, 66, 3270–3289. [Google Scholar] [CrossRef]
  225. Thakur, P.S.; Aggarwal, D.; Takkar, B.; Shivaji, S.; Das, T. Evidence Suggesting the Role of Gut Dysbiosis in Diabetic Retinopathy. Investig. Opthalmol. Vis. Sci. 2022, 63, 21. [Google Scholar] [CrossRef]
  226. Yempén, R.E.A.; Venzel, R.; Campos, M.C.P.; de Oliveira, L.P.; Lins, R.V.D.; Pessoni, A.M.; Fanaro, G.B.; Souza, A.D.O.; Calaza, K.D.C.; de Brito Alves, J.L.; et al. Gut microbiota: A potential therapeutic target for management of diabetic retinopathy? Life Sci. 2021, 286, 120060. [Google Scholar] [CrossRef]
  227. Tanase, D.M.; Gosav, E.M.; Neculae, E.; Costea, C.F.; Ciocoiu, M.; Hurjui, L.L.; Tarniceriu, C.C.; Maranduca, M.A.; Lacatusu, C.M.; Floria, M.; et al. Role of Gut Microbiota on Onset and Progression of Microvascular Complications of Type 2 Diabetes (T2DM). Nutrients 2020, 12, 3719. [Google Scholar] [CrossRef] [PubMed]
  228. Souza, E.L.; Albuquerque, T.M.R.; Santos, A.S.; Massa, N.M.L.; de Brito Alves, J.L. Potential interactions among phenolic compounds and probiotics for mutual boosting of their health-promoting properties and food functionalities—A review. Crit. Rev. Food Sci. Nutr. 2019, 59, 1645–1659. [Google Scholar] [CrossRef] [PubMed]
  229. Mileo, A.M.; Nisticò, P.; Miccadei, S. Polyphenols: Immunomodulatory and Therapeutic Implication in Colorectal Cancer. Front. Immunol. 2019, 10, 729. [Google Scholar] [CrossRef] [PubMed]
  230. Hachimura, S.; Totsuka, M.; Hosono, A. Immunomodulation by food: Impact on gut immunity and immune cell function. Biosci. Biotechnol. Biochem. 2018, 82, 584–599. [Google Scholar] [CrossRef] [PubMed]
  231. Vitetta, L.; Vitetta, G.; Hall, S. Immunological Tolerance and Function: Associations Between Intestinal Bacteria, Probiotics, Prebiotics, and Phages. Front. Immunol. 2018, 9, 2240. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  232. Stan, M.S.; Voicu, S.N.; Caruntu, S.; Nica, I.C.; Olah, N.-K.; Burtescu, R.; Balta, C.; Rosu, M.; Herman, H.; Hermenean, A.; et al. Antioxidant and Anti-Inflammatory Properties of a Thuja occidentalis Mother Tincture for the Treatment of Ulcerative Colitis. Antioxidants 2019, 8, 416. [Google Scholar] [CrossRef] [Green Version]
  233. Pereira-Caro, G.; Oliver, C.M.; Weerakkody, R.; Singh, T.; Conlon, M.; Borges, G.; Sanguansri, L.; Lockett, T.; Roberts, S.A.; Crozier, A.; et al. Chronic administration of a microencapsulated probiotic enhances the bioavailability of orange juice flavanones in humans. Free Radic. Biol. Med. 2015, 84, 206–214. [Google Scholar] [CrossRef] [PubMed]
  234. Xu, J.; Jönsson, T.; Plaza, M.; Håkansson, Å.; Antonsson, M.; Ahrén, I.L.; Turner, C.; Spégel, P.; Granfeldt, Y. Probiotic fruit beverages with different polyphenol profiles attenuated early insulin response. Nutr. J. 2018, 17, 34. [Google Scholar] [CrossRef]
  235. Lacerda, D.C.; da Costa, P.C.T.; Pontes, P.B.; dos Santos, L.A.C.; Neto, J.P.R.C.; Luis, C.C.S.; Brito, V.P.d.S.; de Brito Alves, J.L. Potential role of Limosilactobacillus fermentum as a probiotic with anti-diabetic properties: A review. World J. Diabetes 2022, 13, 717–728. [Google Scholar] [CrossRef] [PubMed]
  236. Sampaio, K.B.; de Brito Alves, J.L.; Nascimento, Y.M.D.; Tavares, J.F.; da Silva, M.S.; Nascimento, D.d.S.; Rodrigues, N.P.d.A.; Monteiro, M.C.; Garcia, E.F.; de Souza, E.L. Effects of Simulated Gastrointestinal Conditions on Combined Potentially Probiotic Limosilactobacillus fermentum 296, Quercetin, and/or Resveratrol as Bioactive Components of Novel Nutraceuticals. Probiotics Antimicrob. Proteins 2023, 1–12. [Google Scholar] [CrossRef] [PubMed]
  237. Sampaio, K.B.; de Brito Alves, J.L.; Nascimento, Y.M.D.; Tavares, J.F.; da Silva, M.S.; Nascimento, D.D.S.; Lima, M.D.S.; Rodrigues, N.P.d.A.; Garcia, E.F.; de Souza, E.L. Nutraceutical formulations combining Limosilactobacillus fermentum, quercetin, and or resveratrol with beneficial impacts on the abundance of intestinal bacterial populations, metabolite production, and antioxidant capacity during colonic fermentation. Food Res. Int. 2022, 161, 111800. [Google Scholar] [CrossRef] [PubMed]
  238. Nascimento, D.d.S.; Sampaio, K.B.; Nascimento, Y.M.D.; de Souza, T.A.; de Souza, F.S.; Júnior, J.V.C.; Tavares, J.F.; da Silva, M.S.; de Brito Alves, J.L.; de Souza, E.L. Evaluating the Stability of a Novel Nutraceutical Formulation Combining Probiotic Limosilactobacillus fermentum 296, Quercetin, and Resveratrol Under Different Storage Conditions. Probiotics Antimicrob. Proteins 2022, 1–13. [Google Scholar] [CrossRef]
Figure 1. Schematic diagram of the retina. The retina is organized into nuclear layers separated by plexiform layers. The photoreceptors (rods and cones) have nuclei in the outer nuclear layer and synapse with bipolar and horizontal cells in the outer plexiform layer. Bipolar cells are divided into rod bipolar (light green) and cone bipolar (light and dark purple). Cone bipolar cells form synapses directly with their respective ganglion cells in specific sublamina (a and b, for, respectively, OFF and ON pathways) of the inner plexiform layer (IPL). Rod bipolar indirectly transmits information to ganglion cells indirectly via an amacrine cell in the IPL. The ganglion cell layer and the nerve fiber layer are formed by the nuclei and axons of ganglion cells, respectively, which transmit the information to the visual encephalic centers. The retinal glial cells are Müller cells, astrocytes, and microglia. The retinal pigmented epithelium (RPE), composed of pigmented epithelial cells, plays several essential roles in maintaining the retina’s health.
Figure 1. Schematic diagram of the retina. The retina is organized into nuclear layers separated by plexiform layers. The photoreceptors (rods and cones) have nuclei in the outer nuclear layer and synapse with bipolar and horizontal cells in the outer plexiform layer. Bipolar cells are divided into rod bipolar (light green) and cone bipolar (light and dark purple). Cone bipolar cells form synapses directly with their respective ganglion cells in specific sublamina (a and b, for, respectively, OFF and ON pathways) of the inner plexiform layer (IPL). Rod bipolar indirectly transmits information to ganglion cells indirectly via an amacrine cell in the IPL. The ganglion cell layer and the nerve fiber layer are formed by the nuclei and axons of ganglion cells, respectively, which transmit the information to the visual encephalic centers. The retinal glial cells are Müller cells, astrocytes, and microglia. The retinal pigmented epithelium (RPE), composed of pigmented epithelial cells, plays several essential roles in maintaining the retina’s health.
Antioxidants 12 01237 g001
Figure 2. Chemical structures of polyphenolic compounds with potential therapeutic implications in diabetic retinopathy: ferulic acid, quercetin, resveratrol, curcumin, and epigallocatechin gallate.
Figure 2. Chemical structures of polyphenolic compounds with potential therapeutic implications in diabetic retinopathy: ferulic acid, quercetin, resveratrol, curcumin, and epigallocatechin gallate.
Antioxidants 12 01237 g002
Figure 3. Schematic illustration of cellular and molecular pathways in diabetic retinopathy leading to oxidative stress, inflammation, and apoptosis. Polyphenols, such as quercetin, curcumin, resveratrol, ferulic acid, and epicatechin gallate, contribute to the inhibition of positive feedback and thus reduce the occurrence of cellular signaling cascades involved in diabetic retinopathy.
Figure 3. Schematic illustration of cellular and molecular pathways in diabetic retinopathy leading to oxidative stress, inflammation, and apoptosis. Polyphenols, such as quercetin, curcumin, resveratrol, ferulic acid, and epicatechin gallate, contribute to the inhibition of positive feedback and thus reduce the occurrence of cellular signaling cascades involved in diabetic retinopathy.
Antioxidants 12 01237 g003
Figure 4. Food sources of polyphenols and the health benefits caused by the consumption of these substances are associated with probiotics, with the main results of reducing blood sugar levels, inflammation, oxidative stress, angiogenesis, intestinal dysbiosis, and increasing neuroprotective factors.
Figure 4. Food sources of polyphenols and the health benefits caused by the consumption of these substances are associated with probiotics, with the main results of reducing blood sugar levels, inflammation, oxidative stress, angiogenesis, intestinal dysbiosis, and increasing neuroprotective factors.
Antioxidants 12 01237 g004
Table 1. Primary outcomes of the studies testing the use of ferulic acid in diabetic retinopathy.
Table 1. Primary outcomes of the studies testing the use of ferulic acid in diabetic retinopathy.
CompoundFood SourceEffects on the Retina
Experimental ModelEffectsReference
Ferulic acidCereals (maize, rice endosperm, barley extract), grasses, oranges, tomatoes, carrots, sugar-beet pulp, and bamboo shootMiceAttenuated the NaIO3-induced retinal degeneration in the retina and choroid.[124]
Mice, intragastric administration.Suppression of microglia activation.[125]
Human retinal cells.Protection against oxidative stress-induced retinal damage.
Reduction in H2O2-induced apoptosis by upregulating Bcl-2, downregulating Bax, and cleaving caspase-3 protein expression.
[126]
RPE cells incubated in 30 mmol/L glucose medium. Decreased cell apoptosis and lowered P53, BAX, and Bcl2 expression levels.[127]
db/db diabetic mice.Decreased degenerative alterations in the retinal tissues.
Decreased expression of apoptosis-related markers in retinal tissues.
HepG2 and L6 cells incubated in a hyperglycemic medium.Increased phosphorylation of AMPK, upregulation of GLUT2 and GLUT4, and inhibition of JNK1/2 activity.[128]
HUVEC cells incubated in a hyperglycemic medium.Inhibitory effects on AGEs and p38 MAPK signaling pathway, activating NF-κB pathway.[129]
Table 2. Primary outcomes of the studies testing the use of curcumin in diabetic retinopathy.
Table 2. Primary outcomes of the studies testing the use of curcumin in diabetic retinopathy.
CompoundFood SourceEffects on the Retina
Experimental ModelEffectsReference
CurcuminRhizome of Curcuma longa L.RPE cells and the retina of diabetic ratsInhibited cell proliferation and angiogenesis by modulating cellular signaling pathways.[143]
RRVECs incubated in a hyperglycemic mediumReduced levels of NFκ-B and pNFκ-B, IKB-α, IL-1β, and Bax, and increased expression of Bcl-2,[156]
RRVECs incubated in a hyperglycemic medium
Diabetic rats
Improved basal membrane thickness.[154,156]
ARPE-19 cellsDownregulated ROS/PI3K/AKT/mTOR signaling pathway by decreasing the levels of pro-inflammatory cytokines and TNF-α, IL-1β, and IL-6.[157]
Human ARPE-19 cellsActivated Nrf2 signaling cascade.[160]
RPE cellsIncreased the expression of Nrf2/HO-1, ERK1/2, and reduced ROS-induced apoptosis.[161]
Streptozotocin (STZ)-induced diabetic ratsImproved histopathologic parameters, markers of oxidative stress, and plasma glucose.
Regulatory effect on Nfr2 pathway activation.
Inhibition of the AGE-RAGE pathway and extracellular matrix (ECM)-receptor interaction in the retina
[162]
Table 3. Primary outcomes of the studies testing the use of quercetin in diabetic retinopathy.
Table 3. Primary outcomes of the studies testing the use of quercetin in diabetic retinopathy.
CompoundFood SourceEffects on the Retina
Experimental ModelEffectsReference
QuercetinTeas (from C. sinensis), onions, apples, and berries.ARPE-19 cells hyperglycemic mediumDecreased inflammatory markers, phosphorylation of MAPKs (ERK1/2, p38, and JNK1/2), and cAMP response element-binding protein (CREB).
Activation of transcription factor 2 (ATF2), c-Jun, and nuclear factor-B kinase (IKK)/NFκ-B.
[186]
Intraperitoneal injection in diabetic ratsReduced the number of apoptotic cells and partially improved the retinal histopathology (decreased the thickness of the inner and outer nuclear layer, decreasing the distance between the inner and outer limiting membrane).
Increased the number of ganglion cells.
Decreased pro-inflammatory cytokines, production of pro-angiogenic factors and soluble adhesion proteins, VEGF, and sICAM-1.
[187]
HRMEC incubated in a hyperglycemic mediumInhibits the activation of angio-genic, inflammatory, and autophagic mechanisms.[188]
STZ diabetic ratsImproved histology in the retina (less edema and cell vacuolization).
Reduced expression of matrix metalloproteinase-9 and VEGF proteins.
[189]
Diabetic ratsIncreased retinal BDNF, NGF, TrkB, and phosphorylation of Akt.
Prevented apoptosis, decreased cytochrome c and caspase-3, and increased anti-apoptotic protein Bcl-2.
[190]
Human ARPE-19 cells incubated with NaIO3 (induce age-related macular degeneration-like retinal pathology)Inhibited the PI3K/Akt pathway and stimulated Bcl-2 production.[191]
Table 4. Primary outcomes of the studies testing the use of resveratrol in diabetic retinopathy.
Table 4. Primary outcomes of the studies testing the use of resveratrol in diabetic retinopathy.
CompoundHealth BenefitsEffects on the Retina
Experimental ModelEffectsReference
ResveratrolAnti-tumor, anti-aging, cardioprotective, neuroprotective, and antioxidant.Diabetic rat retinaInhibited NF-kB activity.
Reversed hyperglycemia
Promoted vascular enlargement in the ganglion cell layer and increased VEGF.
Inhibited the transcription factor levels of inflammatory and cell death proteins.
[200,201,204]
Diabetic rat retina and Muller cells cultureIncreased microRNA 29b (associated with inhibition of SP1, activated caspase 3, and Bax) and increased Bcl-2.[202]
BRECs incubated hyperglycemic mediumPrevents the degradation of AMPK, SIRT-1, and PGC-1α.[206]
Peripheral blood mononuclear cell cultures from patients with proliferative diabetic retinopathyIncreased SIRT-1 Levels and Decreased Proinflammatory Cytokine[207]
HRECs incubated in a hyperglycemic mediumInhibited PKC activation leads to the non-production of ROS, thus inhibiting the phenotypic change from endothelial to mesenchymal cells.[209]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Fanaro, G.B.; Marques, M.R.; Calaza, K.d.C.; Brito, R.; Pessoni, A.M.; Mendonça, H.R.; Lemos, D.E.d.A.; de Brito Alves, J.L.; de Souza, E.L.; Cavalcanti Neto, M.P. New Insights on Dietary Polyphenols for the Management of Oxidative Stress and Neuroinflammation in Diabetic Retinopathy. Antioxidants 2023, 12, 1237. https://doi.org/10.3390/antiox12061237

AMA Style

Fanaro GB, Marques MR, Calaza KdC, Brito R, Pessoni AM, Mendonça HR, Lemos DEdA, de Brito Alves JL, de Souza EL, Cavalcanti Neto MP. New Insights on Dietary Polyphenols for the Management of Oxidative Stress and Neuroinflammation in Diabetic Retinopathy. Antioxidants. 2023; 12(6):1237. https://doi.org/10.3390/antiox12061237

Chicago/Turabian Style

Fanaro, Gustavo Bernardes, Marcelo Rodrigues Marques, Karin da Costa Calaza, Rafael Brito, André Moreira Pessoni, Henrique Rocha Mendonça, Deborah Emanuelle de Albuquerque Lemos, José Luiz de Brito Alves, Evandro Leite de Souza, and Marinaldo Pacífico Cavalcanti Neto. 2023. "New Insights on Dietary Polyphenols for the Management of Oxidative Stress and Neuroinflammation in Diabetic Retinopathy" Antioxidants 12, no. 6: 1237. https://doi.org/10.3390/antiox12061237

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