Skip to main content
Neural Regeneration Research logoLink to Neural Regeneration Research
. 2025 May 6;21(3):908–922. doi: 10.4103/NRR.NRR-D-24-01638

Neuroprotection provided by polyphenols and flavonoids in photoreceptor degenerative diseases

Théo Henrique de Lima-Vasconcellos 1, Gabrieli Bovi dos Santos 1, Marília Inês Móvio 1, Giovanna Klemenc Donnici 1, Gabriela Maria Badin 1, Daniele Ribeiro de Araujo 2,3, Alexandre Hiroaki Kihara 1,4,*
PMCID: PMC12296498  PMID: 40364630

Abstract

The intricate landscape of neurodegenerative diseases complicates the search for effective therapeutic approaches. Photoreceptor degeneration, the common endpoint in various retinal diseases, including retinitis pigmentosa and age-related macular degeneration, leads to vision loss or blindness. While primary cell death is driven by genetic mutations, oxidative stress, and neuroinflammation, additional mechanisms contribute to disease progression. In retinitis pigmentosa, a multitude of genetic alterations can trigger the degeneration of photoreceptors, while other retinopathies, such as age-related macular degeneration, are initiated by combinations of environmental factors, such as diet, smoking, and hypertension, with genetic predispositions. Nutraceutical therapies, which blend the principles of nutrition and pharmaceuticals, aim to harness the health benefits of bioactive compounds for therapeutic applications. These compounds generally possess multi-target effects. Polyphenols and flavonoids, secondary plant metabolites abundant in plant-based foods, are known for their antioxidant, neuroprotective, and anti-inflammatory properties. This review focuses on the potential of polyphenols and flavonoids as nutraceuticals to treat neurodegenerative diseases such as retinitis pigmentosa. Furthermore, the importance of developing reliable delivery methods to enhance the bioavailability and therapeutic efficacy of these compounds will be discussed. By combining nutraceuticals with other emerging therapies, such as genetic and cell-based treatments, it is possible to offer a more comprehensive approach to treating retinal degenerative diseases. These advancements could lead to a viable and accessible option, improving the quality of life for patients with retinal diseases.

Keywords: flavonoids, neuroinflammation, nutraceutical therapy, oxidative stress, retinitis pigmentosa

Introduction

Nutraceutical therapies (NUT) represent a field at the intersection of nutrition and pharmaceuticals, aiming to harness the health-promoting properties of bioactive compounds for therapeutic purposes, emphasizing the dual role of these substances in promoting health and preventing or treating diseases. NUT incorporates diverse compounds, including bioactive compounds such as vitamins, minerals, amino acids, antioxidants, free fatty acids, and phytochemicals in general, which exhibit bioactive properties beyond their nutritional value. The recent literature points out polyphenols as a prominent bioactive group of molecules (Kaleem and Ahmad, 2018; Piccolella et al., 2019).

These compounds, which are abundant micronutrients in our diet, play a crucial role in preventing diseases, with health-promoting effects to combat chronic and degenerative processes. The health effects of polyphenols (flavonoids, phenolic acids, lignins, and stilbenes), especially flavonoids, are contingent upon both the quantity consumed and their bioavailability (Piccolella et al., 2019). Flavonoids stand out as the most prevalent polyphenols within the human diet, being known to date approximately 10,000 variants. This extensive diversity arises from three types of substituents positioned differently within their molecular structures (Naróg and Sobkowiak, 2023). These compounds exhibit a C6–C3–C6 backbone structure and are categorized into several major classes: flavones, flavonols, flavanones, flavanonols, isoflavones, flavan-3-ols, anthocyanins, and chalcones. Widely distributed across nearly all plant-based foods, flavonoids are particularly abundant in vegetables, fruits, teas, flowers, and seeds (Shen et al., 2022).

These molecules are associated with numerous bioactive properties, such as anticancer, anti-diabetic, antibacterial, antiparasitic, antiviral, anti-aging, immunomodulatory, anti-inflammatory, immunobalance, neuroprotective, and antioxidant properties. Notably, the primary activity of flavonoids is their antioxidant capability, which helps prevent damage caused by free radicals by scavenging free reactive oxygen species (ROS). This property stands out as one of most extensively studied aspects of flavonoid activity (Dias et al., 2021; Naróg and Sobkowiak, 2023; Ferreira et al., 2024). The primary challenge in exploring the therapeutic potential of flavonoids lies in their poor solubility in water and low bioavailability, often hindered by a slow absorption rate and rapid metabolism – as addressed later in this review. This limitation is attributed to the structural characteristics of flavonoids, which can be countered by chemical modifications, nanoparticles, and other mechanisms discussed in this review (Koirala et al., 2016; Carullo et al., 2020).

Based on meta-analysis tracking trends from 2003 to 2023, we investigated several terms (Figure 1A–D). The correlation between health-related terms shows an increasing number of links and higher weighting, indicating a growing number of articles emphasizing two terms (Figure 1C). This is evidenced by the increase in the number of triangles in the graphs over time, which means a greater interaction between the terms, as well as the density of the graph (the ratio between the edges and the maximum number of edges that the graph can contain), which jumped from 0.68 in 2003 to 0.97 in 2023 (Figure 1B). Still, it is possible to note a gradual decrease in the average path length over the years, indicating that the terms are increasingly closer to each other, reflecting their more significant mutual influence on each other (Figure 1C).

Figure 1.

Figure 1

Trend over time and meta-analysis combining several terms.

(A) Graphs representing the correlation between terms by their weighted degree, showing a higher correlation as time goes by. (B) Regression line of the total number of triangles and the density per year (2003–2023). (C) Regression line of the average path length and the weighted average grade per year (2003–2023). (D) Line graphs show the timeline of publications (number per year), from PubMed.com, using the keyword “flavonoids” (updated to 2023) - with coefficient R2 indicated in the graphs. Created with BioRender.com. AD: Alzheimer’s disease; ALS: amyotrophic lateral sclerosis; AMD: age-related macular degeneration; HD: Huntington’s disease; NUT: nutraceutical therapy; PD: Parkinson’s disease; ROS: reactive species of oxygen; RP: retinitis pigmentosa.

Notably, there has been growing interest in flavonoids, NUTs, and polyphenols from 2003 to 2023, evidenced by the increased connections with terms such as neuroprotective, diseases, and anti-inflammatory (Figure 1A). These terms increase their weighted degree in the 10 years studied, indicating their growing influence in disease treatment. Over the past two decades, there has been an increased interest in flavonoids, supported by a rising number of published papers (Figure 1D). Economically, NUTs have also expanded significantly, with a projected increase in market size (Ashfaq et al., 2023).

Polyphenols and flavonoids have recently demonstrated remarkable outcomes in the treatment of developmental and age-related diseases, including chronic diseases, Alzheimer’s disease (AD), Parkinson’s disease (Abdul-Latif et al., 2021; Prajapati et al., 2021; Calderaro et al., 2022; Minocha et al., 2022; Bovi Dos Santos et al., 2024; Ferreira et al., 2024; Wu et al., 2024), and enhancing cognition (Gómez-Pinilla, 2008; Brickman et al., 2014). Surprisingly, studies in humans demonstrated that the dietary intake of flavonoids contributes to a delay in the biological aging process, particularly in organs such as the heart and liver (Xing et al., 2023). Also, dietary intake of flavonoids was positively associated with lower mortality risk, AD, and age-related macular degeneration (AMD) development (Kim et al., 2017; Gopinath et al., 2018; Zong et al., 2024). In mouse models, the increased lifespan was demonstrated to be associated with the mechanism of ameliorating cellular senescence (Xu et al., 2021; Wang et al., 2022c). Besides these important effects, polyphenols also regulate epigenetic factors, like histone modifications such as methylation (Wu et al., 2022).

Connected to these findings, the modern human diet has shifted towards increased consumption of ultra-processed foods and additives, potentially resulting in a diminished supply of nutrients and micronutrients. This, coupled with a reduced intake of fruits and vegetables, may contribute to a higher susceptibility to diseases and neurodegeneration, once a healthy diet is crucial for the prevention of several diseases (Aidoo et al., 2023; Nagata et al., 2024; Zhang et al., 2023c; Dong et al., 2024; Whelan et al., 2024). Additionally, a cross-sectional study indicates that the elderly tend to have lower food intake, resulting in a reduced consumption of flavonoids (Zujko et al., 2015). This could be associated with the increasing prevalence of neurodegenerative diseases, considering both improved diagnosis and lifespan due to advancements in science (Hepsomali and Groeger, 2021). These collective findings underscore the practical significance for public health, as flavonoids can be supplemented through simple daily dietary modifications and improvements in eating habits. These substances may be employed as therapeutic agents or used as prophylactic strategies for neurodegenerative diseases.

Nevertheless, the potential applications of flavonoids in addressing retinal degeneration, particularly concerning photoreceptor cells (PR), are still undervalued. The demise of PR in blinding diseases is a complex and multifaceted process, varying significantly across different conditions. While the underlying causes differ, common mechanisms contribute to PR loss in many retinopathies and blinding diseases, including retinitis pigmentosa (RP), AMD, and certain forms of inherited retinal dystrophies (committing 1 to 3000 worldwide) (Wright et al., 2010; Broadgate et al., 2017). Genetic mutations, inflammatory responses, vascular dysfunctions, oxidative stress, and several other pathophysiological processes contribute to the intricate landscape of retinal degeneration (Olivares-González et al., 2021b; Ortega et al., 2021).

Retinal dystrophies present a progressive and severe loss of vision that can be widespread to the whole visual field or concentrated in its center (Provis et al., 2005; Wright et al., 2010), leading to legal blindness. The blinding diseases are considered a significant public health problem (Broadgate et al., 2017; DeAngelis et al., 2017; Retina International, 2025), and the lack of efficient treatments, especially for RP, leads to a sense of hopelessness in those patients. Therefore, treatments that focus on these processes or aim at stabilizing PR during disease progression hold promise for positively impacting the visual lifespan of patients. Fortunately, polyphenols emerge as potential strategies for therapies aiming at PR degeneration, especially cursing with the oxidative stress present in retinal dystrophies (Tao et al., 2016a; Jing et al., 2023; Yan et al., 2023), in which the use of vitamins and oxidants has been proposed as treatments (DeAngelis et al., 2017).

RP is the most prevalent subtype of retinal dystrophy (Chiang and Trzupek, 2015; Bravo-Gil et al., 2017; Costa et al., 2017). It is characterized by a diverse range of mutations exhibiting significant heterogeneity, stemming from a variety of genetic alterations linked to both syndromic and non-syndromic (autosomal recessive, dominant, and X-linked) forms of the condition. In this disease, the patient initially loses peripheral vision due to rod cell loss and gradually affects the central vision by secondary cone cell death. The clinical symptoms are related to photoreceptor and/or retinal pigment epithelium (RPE) misfunction (Daiger et al., 2013; Xu et al., 2014; Costa et al., 2017).

With estimated incidence in 1 to 3500–5000 individuals (Xu et al., 2014; Bravo-Gil et al., 2017; Costa et al., 2017) varying according to the racial group (Okonkwo et al., 2023), RP progression varies depending on the associated mutation. Although the prevalence of the disease may differ depending on the study, it is estimated that non-syndromic RP cases are linked to autosomal dominant (30%–40%), followed by X-linked (5%–15%) and autosomal recessive (50%–60%) mutation. Syndromic RP represents 20%–30% of the cases (Verbakel et al., 2018; Cortinhal et al., 2024). More rarely, RP can be exhibited as spontaneous/simplex RP cases (Bravo-Gil et al., 2017). Approximately 60 genes are involved in the RP (Broadgate et al., 2017), and the majority are associated with phototransduction, visual processes, or related pathways (Xu et al., 2014).

The RP progression is directly influenced by the type of gene and mutation involved in the clinical presentation. Among the mutations associated with RP, some result in loss-of-function (e.g., RHO, PDE6A, PDE6B, among others), leading to impaired protein activity and progressive degeneration, while others create a toxic cellular environment (e.g., RHO [autosomal dominant mutation], PRPF31, RP1, among others), accelerating photoreceptor death. Furthermore, most cases of autosomal dominant RP tend to progress more slowly, as cone photoreceptors may remain functional for a longer period. In contrast, autosomal recessive forms are typically more aggressive, often leading to earlier and more severe retinal degeneration (Jauregui et al., 2019; O’Neal et al., 2024). Regardless of the type of mutation, the first insult of cell death due to the rods creates an imbalance in retinal homeostasis, which could lead to the death of the secondary photoreceptor. Although the secondary deaths are mainly related to cones, some rods die due to the secondary death.

However, cone cell death may occur via distinct mechanisms from those affecting rods, due in part to differences in genetic factors and the unique functions of phototransduction (Campochiaro and Mir, 2018). In a very characteristic way, cone cells usually die after most rods have already degenerated. This may occur once the rod death leads to an increase of toxic substances and activates microglia and Müller cells, which in turn increases the phagocytosis of other photoreceptor cells. On the other hand, the relationship between oxidative stress and cone cell death is robust, with several reports linking the decrease in oxidant species with cone survival (Campochiaro and Mir, 2018; Brunet et al., 2022; Cantó et al., 2022; Vingolo et al., 2022; Kanan et al., 2023).

In contrast, AMD also leads to photoreceptor loss but differs significantly in its etiology and clinical presentation. Unlike RP, whose onset is predominantly driven by inherited genetic mutations affecting phototransduction, AMD is largely influenced by environmental factors, such as body fat, smoking, sedentary lifestyle, diet, and hypertension (Lambert et al., 2016; Pennington and DeAngelis, 2016a). Moreover, CFH and CETP mutations were related to AMD risk (Buschini et al., 2015; DeAngelis et al., 2017). AMD is a known cause of the major age-blinding disease, affecting approximately 1 in 30 individuals as they age (Pennington and DeAngelis, 2016a; Hanna et al., 2022). For example, it is estimated that AMD affects 6.5% of people older than 45 years (Klein et al., 2011), and 1 in 3 people over 85 years old, as cited in (Broadgate et al., 2017). As age is the major risk factor, and considering that life expectancy has been increasing and is projected to rise even further, the 170 million people diagnosed nowadays with IRDs tend to increase to nearly 300 million by 2040 (Wong et al., 2014; Pennington and DeAngelis, 2016a).

AMD typically presents with central vision loss due to drusen formation in the macula, reduced visual acuity, and abnormal color vision (Buschini et al., 2015; Broadgate et al., 2017). Although AMD affects the RPE and often follows a different pathological cascade compared to RP, oxidative stress remains a critical component in both diseases. In AMD, oxidative damage to the RPE and photoreceptors is a key driver of pathology, and therapeutic strategies have focused on antioxidants, anti-vascular endothelial growth factor (VEGF) treatments (for wet AMD), and lifestyle modifications to mitigate these effects (Buschini et al., 2015; Pennington and DeAngelis, 2016b). Besides these environmental and genetic factors, Buschini et al. (2015) extensively reviewed the strategies for AMD management, focusing on reducing inflammation, drusen formation, oxidative stress, and other pathological processes.

Focusing on RP, it is clear that while both RP and AMD share aspects of oxidative stress and inflammation, the primary pathology in RP is rooted in inherited mutations that disrupt photoreceptor function and survival. This leads to a characteristic pattern where rod loss precedes and likely precipitates cone degeneration through toxic byproduct accumulation and glial activation. In contrast, the progression of AMD, driven largely by environmental and metabolic factors, culminates in the loss of central vision through distinct mechanisms involving RPE dysfunction. By emphasizing the unique genetic underpinnings and sequential photoreceptor loss in RP, our work aims to deepen the understanding of the mechanisms underlying PR cell loss. At the same time, insights from AMD research, particularly regarding oxidative stress and inflammation, offer valuable comparative perspectives that could help refine intervention strategies for both conditions.

In contrast to AMD, RP still does not have a consistent therapy for all the patients, only for a small group of people, being Luxturna® the sole FDA-approved treatment, employing genetic therapy focused on the RPE65 mutation. This reinforces the need for treatments that are economically accessible and capable of benefiting a broader group of patients, thereby enhancing life quality and extending vision lifespan across mutations. One such therapy that might be able to accomplish this goal could be NUT based on polyphenols. Several clinical trials have been engaged in NUT, but primarily used for retinopathies like glaucoma, diabetic retinopathy, and AMD (Additional Tables 14). Still, there are few NUT clinical trials in progress or evaluating the efficacy of this therapy for RP, none of these employ polyphenols as NUT strategy.

Additional Table 1.

Clinical trials based on dietary supplements or nutraceutical therapy for AMD

Clinical data ID Compounds Methods Results Years
NCT00060749 DHA Daily oral supplementation for three months (2000 mg/day). Group 1: 20 mg lutein, 2 mg zeaxanthin, 510 mg omega-3-FA; No perceived effect of DHA supplementation on macular function. 2003
NCT00763659 Combination of lutein/zeaxanthin and omega-3- fatty acids Group 2: 10 mg lutein, 1mg zeaxanthin, 255 mg Omega-3-FA; both daily oral supplementation about one year. Increase in of the macular pigment optical density, improvement and stabilization of best-corrected visual acuity test. 2008
NCT00951288 Saffron (Crocus Sativus extract) Daily oral supplementation for three months 20 mg/day. Improved focal electroretinograms amplitude and threshold, improving retinal flicker sensitivity in early AMD. 2009
NCT01042860 Lutein Daily oral supplementation for twelve months 10 mg/day. Increase of macular pigment optical density levels in early-stage AMD patients. 2010
NCT01258335 Omega-3-fatty acids Omega-3 fatty acids 4 g orally daily. No measurable changes in visual acuity or retinal function by multifocal electroretinographic testing. 2010
NCT01404845 Lutein and zeaxanthin 2 capsules per day, for 8 months (5 mg lutein + 1 mg zeaxanthin / capsule). Finished, no results published. 2011
NCT01316198 Lutein and zeaxanthin 4 weeks of supplementation with a beverage containing 10 mg lutein and 3 mg zeaxanthin per day. The concentrations of the xanthophylls in plasma and the MPOD increased significantly in the kale group after 4 weeks of intervention. Nevertheless, the improvements did not persist over 4 weeks of washout. 2011
NCT04756310 Retilut and theavit Two capsules/day before breakfast each day for 2 years. Reduction of inflammatory cytokines and improved the fatty acid profile and serum lutein concentration. 2014
NCT02264938 Lutein (12 mg), zeaxanthin (2 mg), astaxanthin (8 mg), omega-3 fatty acids (DHA 540 mg + EPA 360mg), vitamin C (40 mg), vitamin E (20 mg), zinc (16 mg), copper (2 mg) Daily oral supplementation with antioxidants for 1 year. Antioxidant vitamin and mineral supplementation (AREDS: vitamin C, E, beta-carotene, and zinc) probably slows down progression to late AMD 2014
NCT02625376 Trans-resveratrol Group 1: Dietary supplementation with resveratrol 250 mg BD 2 capsules daily. Group 2 (active comparator): Dietary supplementation with Resvega BD, two capsules daily: one in the morning and evening for 24 months. Results not published. 2015
NCT03919019 Macuprev (Ganoderma lucidum (Reishi), calendula officinalis, lutepure-marigold (5% lutein and 1% zeaxanthin), blueberry, rutin, alpha-lipoic acid, bromelain, NAC, vitamin C, B9, B12, D3, selenium, zinc and copper) Oral supplementation (Macuprev) 2 capsules per day for 6 months. Intermediate AMD, Macuprev® supplementation increases the function of the macular pre-ganglionic elements. 2018
NCT03946085 Lumega-Z carotenoid liquid-supplement and AREDS2 Experimental group (LM): carotenoid supplementation Lumega-Z for 6 months Active Comparator (PV): AREDS2 for 6 months Benefits in visual performance in the LM group. 2019
NCT05062486 Resveratrol, quercetin, curcumin Daily oral intake of 100 mg resveratrol, 120 mg quercetin, 1000 mg curcumin, for 24 months. Unknown status, no results published. 2021
NCT05005884 Oral phenolics Oral phenolics 250 mg two times daily for 1 month. Unknown status, no results published. 2021
NCT06391411 Mix containing lutein (10 mg), zeaxanthin (2 mg), saffron (20 mg), vitamin C (80 mg), vitamin E (12 mg) and zinc (10 mg). Intravitreal injections of anti-VEGF (aflibercept 2 mg, 0.05 mL) at a fixed regimen and daily supplementation with a micronutrient mix containing lutein (10 mg), zeaxanthin (2 mg), saffron (20 mg), vitamin C (80 mg), vitamin E (12 mg) and zinc (10 mg) for 6 months. In progress, no results. 2024

Additional Table 4.

Clinical trials based on dietary supplements or nutraceutical therapy for RP

Clinical data ID Compounds Methods Results Years
NCT00000116 Docosahexaenoic acid + vitamin A. Daily oral administration of 1200 mg/d docosahexaenoic acid and 15000 IU/d vitamin A as retinyl palmitate, for 5 years. No effect in the course of disease in patients with retinitis pigmentosa. 1999
NCT00346333 Lutein Daily oral administration of 12 mg of Lutein plus 15,000 IU/d of Vitamin A palmitate, over a 4-year interval. Slower loss of midperipheral visual field on average among nonsmoking adults with retinitis pigmentosa taking vitamin A. 2006
NCT01256697 9-cis rich powder of the alga dunaliella bardawil. Daily oral administration of capsules containing 300 mg of 9-cis β-carotene-ich alga dunaliella bardawil or placebo (starch) for 90 days - 90 day washout period - Other capsule administration for more 90 days. Increased retinal function in patients with RP. 2010
NCT01680510 Alga dunaliella bardawil Daily oral administration of capsules containing alga dunaliella bardawil 9-cis beta-carotene rich powder for 24 weeks followed by a washout period with placebo capsules. Active, not recruiting 2012
NCT02018692 9-cis-beta caroten rich D. Brdawiil extract (food supplement made from alga dunaliella bardawil). Daily oral supplementation for 24 weeks (5 mg/kg). Not yet recruiting, last updated post in 2024. 2013
NCT02244996 Lycium barbarum (also known as "Goji berry"). Daily oral supplementation for twelve months (10 g of granules). Reduced thinning of the macular layer, no significant differences in the sensitivity of the visual field or amplitude of the full- field electroretinogram. 2014

This review aims to comprehensively explore the potential therapeutic benefits of nutraceutical interventions, specifically focusing on polyphenols and flavonoids in the management of PR deterioration, focusing on RP and AMD intricate pathophysiology.

Search Strategy

A comprehensive literature search was conducted using PubMed and Google Scholar to identify relevant studies on nutraceutical approaches in retinal diseases. The keywords included: “retina,” “polyphenols,” “retinitis pigmentosa,” “nutraceutical,” “dietary compounds,” “retinal degeneration,” “flavonoids,” “neuroprotection,” and “natural compounds.” Additionally, the reference lists of selected studies were screened for relevant literature on research investigating the effects of flavonoids and other nutraceutical compounds on health and diseases of the central nervous system. The cited articles were all in English, with a preference for the most recent publications. However, key older references were included when necessary to trace the discovery and development of nutraceutical compounds. For the meta-analysis, the search covered publications ranging from 2003 to 2023, using PubMed exclusively for studies where the keywords appeared in the title/abstract. All terms used in the graphs were searched in combination to ensure a comprehensive analysis of the available data. Data on clinical trials were collected from ClinicalTrials.gov, using keyword combinations such as “dietary compounds,” “nutraceutical,” “supplementation,” “flavonoids,” and “polyphenols” and several retinopathies as seem in Additional Tables 14.

Pathophysiology of Photoreceptor Degeneration

Understanding the intricate pathophysiological mechanisms underlying PR death is crucial for identifying targeted therapeutic approaches. This holds particularly true for conditions such as RP and AMD, complex retinal disorders that majorly impact photoreceptors. Although each condition may have distinct causes, they exhibit common issues such as cellular stress, metabolic stress, oxidative stress, and extensive neuroinflammatory response. Regardless of the specific biological dysfunction associated with the mutated gene, the most common outcome is PR death by apoptosis – and triggering oxidative stress and proinflammatory responses. As discussed ahead, NUT based on polyphenols and flavonoids might be a valuable therapy for RP.

Pathways of photoreceptor cell death in retinitis pigmentosa

Focusing on the RP group of mutations and considering its multigenic factor, the pathogenesis may vary between patients; however, certain aspects of the pathophysiology remain consistent (Martin-Merida et al., 2019; Gallenga et al., 2021; Zhao et al., 2022). In RP with rod mutations in the PDE6B (Koyanagi et al., 2019; Kuehlewein et al., 2021) or rhodopsin genes (Ploier et al., 2016), the mechanisms involve dysregulated visual cycles, like phototransduction, and the altered catalytic function of the mutated PDE6B protein results in the accumulation of cyclic GMP. Consequently, the cyclic nucleotide-gated (CNG) channels are overactivated, causing a significant increase in intracellular calcium ions (Ca2+). Elevated calcium levels lead to two notable consequences: the initiation of apoptosis and the disruption of chromatin. These effects ultimately culminate in the loss of rod cells (see more in Gao et al., 2020; Li et al., 2023). Increased levels of cellular cGMP and the consequent upregulation of protein kinase G (PKG) signaling disrupt various cellular functions, affecting endoplasmic reticulum (ER) stress (Paquet-Durand et al., 2009; Xu et al., 2013), and ER Ca2+ homeostasis (Butler et al., 2017; Yang et al., 2021). This dysregulation also activates histone deacetylase (HDAC)/poly-ADP-ribose polymerase (PARP) signaling pathways, leading to DNA condensation/damage and eventual cell death (Sancho-Pelluz et al., 2010). Furthermore, heightened cGMP signaling and subsequent cellular Ca2+ overload initiate the calpain-associated cell death pathway. Additionally, ER stress, PARP signaling activation, and calpain signaling induce mitochondrial damage, triggering the release of the apoptosis-inducing factor and exacerbating the cell death process (Paquet-Durand et al., 2007; Comitato et al., 2020).

The ER is a crucial organelle responsible for protein biosynthesis, folding, and secreted proteins, as well as serving as a reservoir for free calcium ions. Maintaining ER homeostasis is vital for proper cellular function, and disruptions can trigger unfolded protein response and ER stress, leading to impaired protein processing, trafficking, and degradation (Chen et al., 2023). ER plays a pivotal role in cellular and ER Ca2+ homeostasis, regulated by IP3Rs, RyRs, and SERCA (Butler et al., 2017; Zhang et al., 2023a). In CNG channel-deficient mice, IP3Rs can change cGMP-dependent PKG activation, and disrupt ER Ca2+ homeostasis, resulting in increased expression/activity of IP3R1 and RyR2, exacerbating ER stress and photoreceptor death (Ma et al., 2015; Butler et al., 2017). Inhibition of PKG or depletion of cGMP restores ER Ca2+ balance and ameliorates ER stress-associated photoreceptor degeneration. Elevated PKG signaling, as observed in conditions like CNG channel deficiency in a model of ACHM (Cnga3−/−), exacerbates ER stress and opsin mistrafficking (Sundaram et al., 2014; Ma et al., 2015; Butler et al., 2017).

Additionally, ER stress contributes to aberrant protein trafficking and activation of calpain-associated cell death pathways leading to cell death (Nakagawa et al., 2000; Haidara et al., 2008). Chemical or genetic inhibition of ER marks or ER Ca2+ efflux mitigates ER stress and improves protein trafficking (Hu et al., 2012; Butler et al., 2017; Chen et al., 2022c). High intracellular calcium levels prompt the activation of calpains, enzymes that cleave proenzymes into their active forms, targeting various cytoplasmic and nuclear substrates, such as caspase-3 and PARP. This activation initiates a cascade of events, including the release of apoptosis-inducing factor from mitochondria, and promoting apoptosis (Sanges et al., 2006; Vosler et al., 2009; Vu et al., 2022; Yan et al., 2022). Several polyphenols have been reported to impact ER/mitochondrial stress and cell death pathways (Ha et al., 2014; Bardak et al., 2017; Liu et al., 2018; Pawlowska et al., 2019). For instance, morin has been shown to ameliorate neuron cell loss and improve function in a Huntington’s disease rat model by affecting these pathways, also modulating IP3R (El-Emam et al., 2024). Additionally, the flavonoids baicalin and baicalein have been identified as inhibitors of guanylate cyclase, thereby reducing cGMP levels in rat aortic rings (Huang et al., 2004).

ER stress is highly observed in RHO mutations, the first mutated gene identified in RP (Dryja et al., 1990), and mutations in this gene are also related to congenital stationary night blindness (Zhou et al., 2012). Rhodopsin is a light-sensitive protein present in rod photoreceptors that plays a crucial role in phototransduction (Zhou et al., 2012). Under normal physiology, rhodopsin is synthesized in the photoreceptor’s ER and translocated to the outer segments of the photoreceptor to participate in the phototransduction process (Athanasiou et al., 2018). Seven distinct classes of RHO mutations have been observed, and the related cell death subtype might vary according to the mutation (Azam and Jastrzebska, 2025), which impacts both clinical presentation and rhodopsin cellular distribution (Lewin et al., 2014). The alterations observed in RHO lead to cellular defects related to incorrect protein folding, changes in posttranslational modifications, and mutations that prevent RHO dimerization – these alterations might affect RHO’s intracellular traffic, stability, and function (Zhou et al., 2012; Athanasiou et al., 2018; Azam and Jastrzebska, 2025). RHO alterations may lead to either autosomal dominant or autosomal recessive RP, associated with Rho gain-of-function and Rho loss-of-function mutations, respectively (Athanasiou et al., 2018). One of the most common RHO mutations is P23H, a prevalent form of adRP that results in a non-functional Rho monomer. The P23H mutation is characterized by a misfolded Rho protein that affects its function in phototransduction, contributing to rod photoreceptor dysfunction. P23H mutation leads to RHO retention in the ER, unfolded protein response activation, and subsequent rhodopsin degradation, activating several intracellular pathways such as oxidative stress, release of intracellular calcium and neuroinflammation (Athanasiou et al., 2018; Zhen et al., 2023).

In addition to gene alteration present in rod photoreceptors, RP can also be caused by alterations in genes within RPE cells, such as the RPE65, a retinoid isomerase. This is due to the RPE’s role in the visual cycle, as it is in these cells that retinal regeneration occurs after photoisomerization, which is crucial for photon capture and the regeneration of rhodopsin after phototransduction (Cai et al., 2009). RPE65 is essential in this process, and mutations in this protein can lead to both RP and Leber congenital amaurosis (Jauregui et al., 2018). Due to RPE65 function in retinal regeneration, any kinds of dysfunction interferes with retinal regeneration, affecting photoreceptor function, particularly rods (Pierrache et al., 2020). Interestingly, some studies suggest that the type of RPE65 mutation might affect the age of disease onset, with truncating mutations leading to earlier onset, while missense mutations are associated with later onset (Lopez-Rodriguez et al., 2021). However, others reported no relation between RPE65 genotype and phenotype (Chung et al., 2019; Aoun et al., 2021).

Pathways of photoreceptor cell death in age-related macular degeneration

AMD, encompassing early and intermediate stages, is characterized by the presence of medium or large drusen (protein, lipid, and minerals aggregates), respectively, and/or RPE anomalies (Belmouhand et al., 2022; Oncel et al., 2023). Patients in early or intermediate stages may develop either of the two late-stage forms of the disease: dry AMD or wet AMD, both resulting in photoreceptor death and vision loss (Davis et al., 2005; Ferris et al., 2013; Ruan, Jiang and Gericke, 2021).

The dry AMD is characterized by sharply demarcated atrophic lesions of photoreceptors and RPE cells (Sadda et al., 2016; Holz et al., 2017), and abnormal choriocapillaris perfusion contributes to disease progression (Shi et al., 2021). These lesions result in a region of RPE hypopigmentation and/or hyperpigmentation, typically originating around the fovea perimeter (see more in Fleckenstein et al., 2021). This condition leads to disorganization and eventual loss of photoreceptor inner and outer segments, culminating in photoreceptor death. Additionally, age-related toxic accumulation of autofluorescent lipofuscin within the RPE is evident (Ibbett et al., 2019; Zhang et al., 2019; Edwards et al., 2023), and RPE mitochondrial dysfunction may be involved in disease progression (Senabouth et al., 2022). Recently, the first therapy for dry AMD was approved by the FDA with pegcetacoplan (Heier et al., 2023).

In wet AMD, leaking choroidal neovascular vessels lead to RPE or photoreceptor detachment, hemorrhage, and subsequent fibrovascular scarring, which is responsible for most cases of blindness (Gass et al., 2003; Brandli et al., 2022; Liu et al., 2023). This AMD form is characterized by the pathological proliferation of new blood vessels into the macula, leading to macular edema, subretinal hemorrhage, and fibrous scarring. These abnormal vessels can be leaky, causing fluid and blood accumulation in the subretinal space and neural retina, resulting in fibrotic scar, rapid and severe vision impairment (Bakri et al., 2019; Liu et al., 2023; Pawloff et al., 2023). Fibrotic changes in the late stages of neovascular AMD can further contribute to photoreceptor dysfunction and degeneration (Bloch et al., 2013; Bakri et al., 2019; Fan et al., 2023). While anti-VEGF therapies targeting VEGF have shown visual improvement in many patients by reducing abnormal blood vessel growth and leakage, a portion of patients does not experience visual improvement (Finger et al., 2020). Interestingly, polyphenols such as curcumin and resveratrol demonstrated anti-angiogenic effects on retinal and choroidal neovascular on administration (Nagai et al., 2014; Sulaiman et al., 2014; Ivanescu et al., 2015). In the AMD animal model, a prodrug of the flavonoid epigallocatechin-3-gallate shows interesting results in alleviating the choroidal neovascular and neuroinflammation via downregulation of the hypoxia-inducible factor-1α/VEGF/VEGF receptor 2 pathways (Xu et al., 2020). The RPE plays a critical role in supporting the function of PR, such as glycolysis (Sinha et al., 2020) and lipid metabolism (Farnoodian et al., 2023), phagocytosis of PR outer segments (Reyes-Reveles et al., 2017; Iker Etchegaray et al., 2023), secretion of neurotrophic factors to stabilize the neural retina (Ahluwalia et al., 2023), and cleaning for damaged ROS (Atienzar-Aroca et al., 2016; Sinha, Naash and Al-Ubaidi, 2020; Chen et al., 2022a). Furthermore, the RPE actively participates in the visual cycle by recycling all-trans retinal back to 11-cis retinal, a process vital for phototransduction (Jin et al., 2005). Because each RPE cell interacts with multiple PRs, it becomes a highly metabolically active tissue, ensuring the continuous regeneration and recycling of lipid-rich photoreceptor outer segments to maintain optimal visual function (Bazan et al., 1992; Storti et al., 2017; Kocherlakota et al., 2023).

However, in AMD, dysfunction of the RPE has a significant impact on photoreceptor function. Impaired phagocytosis by RPE cells leads to the accumulation of undigested waste within the RPE, contributing to oxidative stress and, ultimately, photoreceptor damage (Inana et al., 2018; Scrivo et al., 2018). Dysregulated autophagy further exacerbates intracellular waste accumulation and inflammation, while disturbances in lipid metabolism disrupt energy metabolism and nutrient supply to the retina, impairing photoreceptor function and survival (Mitter et al., 2014; Somasundaran et al., 2020). Extracellular deposits such as drusen and reticular pseudodrusen create a hostile microenvironment, obstructing nutrient diffusion and promoting local inflammation, further compromising PR health by structural disruption of the segment’s junction (Doyle et al., 2012; Hartmann et al., 2012; Sadigh et al., 2015; Marsh-Armstrong et al., 2022). In summary, AMD-induced changes in RPE function can contribute to photoreceptor degeneration. Understanding the mechanisms of photoreceptor cell death is crucial for developing new treatment strategies and identifying biomarkers for these complex retinal diseases.

Polyphenols such as curcumin and quercetin have been described as having several regulatory mechanisms in RPE cells. They promote pro-survival responses to injurious stimuli and also downregulate VEGF (Bardak et al., 2017; Parmar et al., 2020). In vitro observations of Sheu et al. (2010) have demonstrated that natural products like polyphenols and carotenoids, such as resveratrol and lutein, exert a protective effect on RPE cells when exposed to acrolein and H2O2. Importantly, these compounds also alleviate the inhibition of phagocytosis by acrolein. Additionally, quercetin exhibits a property to enhance RPE phagocytosis of degraded photoreceptors’ outer segments by inhibiting mTOR pathways (Huang et al., 2018). It is noteworthy that a clinical trial examined patients with AMD following anti-VEGF therapy and their dietary intake of polyphenols. Higher intakes of polyphenols were found to potentially correlate with improved long-term treatment outcomes in AMD (Detaram et al., 2021).

Oxidative stress & vascular dysfunction

Oxidative stress arises from an excessive production of ROS and/or a decrease in antioxidant levels. ROS, primarily comprising superoxide anions, hydrogen peroxide, and hydroxyl radicals, are generated during cellular energy production, where molecular oxygen is reduced to water. The maintenance between ROS generation and antioxidant defense balance is crucial for several disease prevention, including those that curse with photoreceptor degeneration. Elevated ROS levels are recognized for their damaging effects on major cellular macromolecules, such as lipids, proteins, and nucleic acids, ultimately leading to neuronal dysfunction and neurodegeneration (Böhm et al., 2023).

The retinal tissue displays a high metabolic rate, leading to increased oxygen consumption. Additionally, considering its exposure to direct light, the retina becomes susceptible to oxidative stress (Böhm et al., 2023). Studies using animal models of RP, such as rd1 and rd10 mice, have demonstrated elevated ROS production in degenerative retinal loci, indicating oxidative stress as a key pathology in retinal degeneration. RP patients present several clinical alterations associated with oxidative stress: blood samples show higher levels of ROS and H2O2, with an imbalance of the oxidative-antioxidant ratio. The aqueous humor and vitreous presented reduced antioxidant capacity and increased oxidative stress markers, with DNA and protein oxidative damage (Martínez-Fernández de la Cámara et al., 2013; Wang et al., 2022b; Vingolo et al., 2022). Therapeutic interventions targeting oxidative stress in animal models, such as treatment with antioxidants such as vitamin E, superoxide dismutase (SOD) mimetics, vitamin C, and N-acetylcysteine, have shown promising neuroprotective effects in preserving rod and cone PRs. Polyphenols, as previously mentioned, are especially renowned for their role as scavengers of ROS. They not only sequester and neutralize these molecules but also induce the upregulation of important endogenous antioxidants such as glutathione, SODs, and catalase. This dual action helps to maintain a more balanced oxidative-antioxidant ratio (Song et al., 2015; Shen et al., 2022; Naróg and Sobkowiak, 2023).

Similarly to the neuroinflammatory response, the extent of vascular dysfunction is directly correlated with the progression of RP in terms of PR cell loss, manifesting as a secondary event prior to rod PR degeneration. Oxidative stress can be directly correlated to the retinal vascular; this correlation is likely attributed to alterations in the metabolic demand of the retina, particularly in the outer retina (Lang et al., 2019). The oxygen in the outer retina is supplied by retinal blood vessels located at the blood-retinal barrier (BRB) and choroidal vasculature, supported by the RPE (Selvam et al., 2018). The rod PR loss leads to increased oxygen levels in the extracellular matrix of the outer retina, resulting in increased oxygen influx towards the inner retina, leading to a state of hyperoxia. It is noteworthy that individuals with RP exhibit reduced oxygen consumption in the retina and higher oxygen saturation compared to non-RP controls. This phenomenon may be attributed to the absence of regulation of retinal circulation, resulting in a spontaneous increase in oxygen supply (Lang et al., 2019; Wang et al., 2022b). The PR degeneration also impacts reducing blood flow in the choroid by elevated expression of endothelin-1, a vasoconstriction modulator; this might be associated with reactive gliosis and RPE cells in response to oxidative stress. Ultimately, deterioration of the vasculature may lead to vascular remodeling, retinal atrophy, and BRB disruption (McMurtrey and Tso, 2018; Lang et al., 2019; Peddada et al., 2019; Kajtna et al., 2022). The combination of oxidative stress exacerbated inflammation and disruption of the vasculature is also associated with secondary cell loss of PR cone cells (Piano et al., 2019). Studies in retinopathies like diabetic retinopathy and AMD highlights the positive usage of flavonoids and polyphenols, impacting the vasculature and ROS contention, providing a solid base for these compounds as therapeutic potential (Allegrini et al., 2022; Choo et al., 2022; Fanaro et al., 2023; Wang et al., 2024).

Neuroinflammation

The inflammatory response in RP and AMD is associated with several important cells – these being the macro – and microglia cells. Microglia, the resident immune cells of the retina, present a ramified morphology in healthy retinas, carrying out the function of immune surveillance of the microenvironment. However, upon stimulation by changes in the microenvironment, whether they are signs of injury, infection, or cellular stress, they undergo activation, initiating an immune response, and their morphology is altered into an ameboid-like. This alteration enhances their mobility within the tissue and their phagocytic capability, facilitating the engulfing and removal of cellular debris, and pathogens (Murenu et al., 2022; Leinonen et al., 2023).

This activation is a hallmark of neuroinflammation and has been observed in several RP models and human patients (Ma et al., 2012; Roche et al., 2016; Zhou et al., 2017; Yu and Saban, 2023). The degeneration of PR or formation of the druses in early stages triggers microglia activation and migration to the injury site in response to the stress signals (i.e., metabolic and oxidative stress) emitted and the remnants of dying cells. The release of inflammatory mediators further contributes to the neuroinflammatory milieu in the retina. Although this response is intended to clear cellular debris and facilitate tissue repair, excessive or prolonged inflammation may intensify the degenerative process (Peng et al., 2014; Blank et al., 2018; Lew et al., 2020; Murenu et al., 2022; Augustin et al., 2023). Notably, the phagocytosis of viable photoreceptors has been demonstrated to contribute to the progression of the degeneration (Zhao et al., 2015). The activated state can also be classified based on the polarization/phenotype expressed, traditionally categorized as M1 pro-inflammatory and M2 anti-inflammatory (Jurga et al., 2020). During the degeneration, microglial cells express the M1 phenotype releasing cytokines that lead to increased PR cell death, such as tumor necrosis factor-α (Zhou et al., 2017). In this state, several pathways exhibit increased expression and regulation of the response, mainly nuclear factor-κB, Toll-like receptor 4, mitogen-activated protein kinase, and others. Studies focused on AD and retina showed potent regulation of flavonoids in several important aspects of the microglia, such as regulation of these pathways, ultimately leading to phenotype change, generally to an anti-inflammatory state or reduced activation state (Jang and Johnson, 2010; Carullo et al., 2020; Medrano-Jiménez et al., 2022).

The neuroinflammation response in the retina is also mediated by macroglial cells such as Müller cells. Müller cells are the most numerous macroglial types in the retina, characterized by their radial morphology. Functionally, Müller cells perform the maintenance of the BRB, besides being responsible for helping in the cytoarchitecture, retinal homeostasis/functional stability, and playing an active role in the modulation of the phototransduction process and photoreceptor metabolism (de Hoz et al., 2016). Like the microglial response to the PR degeneration, the Müller cells undergo reactive gliosis, mediating the release of cytokines, chemokines, and molecules of a neuroprotective nature. This response may exacerbate the degeneration of neurons and vasculature (de Hoz et al., 2016; Tan et al., 2020; Leinonen et al., 2023). This process is characterized by morphological changes and alterations in gene expression, increased proliferation, and expression of cellular markers characteristic of inflammation (Uren et al., 2014; Fernández-Sánchez et al., 2015; Hippert et al., 2015; Roche et al., 2016; Tomita et al., 2021; Navneet et al., 2024; Xu et al., 2024).

Finally, at the late stage of the retinal degeneration progression, Müller cells are responsible for the retinal remodeling (Jones et al., 2016; Navneet et al., 2024). Conversely, some studies suggest that Müller cells may also play a neuroprotective role by releasing factors that promote cell survival. Some studies focused on other retinopathies such as diabetic retinopathy show increasing evidence of the effects of flavonoids in macroglial cells, reducing the GFAP expression, which contributes to a lower reactive gliosis state (Nones et al., 2010; Matos et al., 2020; Fanaro et al., 2023). Understanding the balance between neurotoxic and neuroprotective functions of the crosstalk of microglial cells and Müller cells is essential for devising targeted therapeutic strategies (Roche et al., 2018; Díaz-Lezama et al., 2023).

In addition to the glial response, the immune system is also affected, with infiltrates of white blood cells observed in RP and AMD retinas due to BRB dysfunction and systemic inflammation (Murakami et al., 2020; Tan et al., 2020; He et al., 2022; Mohan et al., 2022; Ascunce et al., 2023). Furthermore, samples of blood of patients show higher numbers of activated T cells, immune cells fundamental to humoral response, which in this case appears to express the Th1 phenotype response (McMurtrey and Tso, 2018). Related to this systemic inflammation, it is also an aspect that nutraceutical strategies utilizing flavonoids modulate, as seen in rheumatic diseases (Yi, 2021), reducing systemic inflammation in human trials of coronary artery disease patients (Serafini et al., 2010). Ferreira et al. (2024) extensively reviewed polyphenols as immunonutrients, pointing out their effects in chronic diseases and inflammation. Noteworthy, as described above, patients with retinal degeneration showed several systemic alterations, further supporting the NUT with flavonoids as a promising therapeutic strategy (Zhao et al., 2022).

Pre-Clinical Data of Polyphenols & Flavonoids

The pre-clinical data presented here is majorly based on RP animal models (Additional Table 5). As discussed before, several mutations are recognized and classified as RP-link mutations, the same ones found in human RP patients can be studied in murine models. Animal models have been developed to facilitate the study of disease progression, aiming at discoveries to improve the quality of life of patients with RP. One of the most used models for laboratory studies is the mice with alteration in the gene PDE6B (Ali et al., 2017; Xu et al., 2017). This gene, essential for cGMP stability in phototransduction (Ali et al., 2017; Xu et al., 2017), is altered in 50%–60% of patients with non-syndromic RP of the autosomal recessive type (Ali et al., 2017; Xu et al., 2017). With the specific RD1 and RD10 mutation in rods, the Pde6Brd1 and Pde6Brd10 mouse, respectively, are the most well-characterized animal models, RD10 being the one that most closely matches disease progression in humans (Punzo et al., 2009; Stasheff et al., 2011; He et al., 2017; Iraha et al., 2018; Trachsel-Moncho et al., 2018). The degeneration timeline of these mice has been widely described over the years by various scholars. In both models, the timeline that determines the onset of rod photoreceptor cell death, its peak, and the onset of secondary cone photoreceptor death is well known (Barhoum et al., 2008; He et al., 2017). One of the main differences besides different time progressions is that RD10 retinas are more mature by the time the degeneration starts (Stasheff et al., 2011; Olivares-González et al., 2021a).

Additional Table 5.

Pre-clinical data utilizing in vivo experimentation with retinal degeneration models and polyphenols as treatment

Years of publication Compounds Animal models Methods Results References
2017 Curcumin RD1 mice Intravitreal injection, at a dose of 10 μM/L dissolved in DMSO, in mice at P7 (before the start of rod cell death). Reduced number of TUNEL-positive cells, and visual function improved. Regulation of microglial activation. Wang et al., 2017
2021 Quercetin and myricetin P23H rhodopsin knock-in mice Intraperitoneal injection, at a dose of 20 mg/kg body weight, starting from P14 to P21. Reduction of pro-inflammatory markers. Ortega and Jastrzebska, 2021
2021 Quercetin and myricetin Abca4-/-Rdh8-/ - or BALB/c mice The Abca4-/-Rdh8-/- or BALB/c mice. The flavonoid compounds at a concentration of 20 mg/kg or DMSO vehicle were delivered to mice through intraperitoneal administration 30 minutes before exposure to bright light. Preservation of photoreceptor cells and visual function. Reduced number of TUNEL-positive cells, GFAP expression and reactive oxygen species. Ortega etal., 2021
2022 Quercetin and myricetin RhoP23H/P23H was used to test the protective effects of flavonoids against retinal degeneration at 14-21 days of age for RhoP23H/P23H and at 21-33 days of age for RhoP23H/+. Flavonoids were dissolved in DMSO/phosphate-buffered saline vehicle and administered to mice by intraperitoneal injection every other day at the same time of the day, three injections total at 20 mg/kg. Preservation of photoreceptor cells and visual function. Downregulation of the UPR signaling and oxidative stress-related markers. Ortega etal., 2022
2022 Kaempferol Control (4% v/v dimethyl sulfoxide in normal saline) or kaempferol 20 mg/kg in vehicle solution was injected intraperitoneally once per day for 5 days after light injury. Preservation of photoreceptor cells and visual function. Reduced number of TUNEL-positive cells. Piano et al., 2022
2022 Quercetin and naringenin Tvrm4 mice is pre-treated with naringenin 100 mg/kg/die, quercetin 100 mg/kg/die, naringenin 50 + quercercetin 100 mg/kg/die or vehicle dimethyl sulfoxide (DMSO 0.025%) in the drinking water for 35 days. On the 30th day, retinal degeneration was induced by exposure for 1 min to the white light of 12,000 lux intensity, and the treatment was repeated for another 5 days. Preservation of photoreceptor cells and visual function. Upregulation of endogenous antioxidant enzymes, downregulation of apoptosis markers. Noguchi et al., 2023
2011 Curcumin P23H line 1 transgenic rat retina, 100 mg/ kg body weight of curcumin dissolved in alimentum was administered every day by oral gavage from P30 until P70 days. For evaluation of gene expression, curcumin was administered from postnatal day 7 (P7) to P30. Preservation of photoreceptor cells and visual function. Reduced number of TUNEL-positive cells. Downregulation of apoptosis markers. Vasireddy et al., 2011
2015 Curcumin A domestic sow was inseminated with semen from a Tg P23H miniswine founder. Her daily diet was supplemented with curcumin (100 mg/Kg body weight) from embryonic (E) day 80 to E112. The same diet without curcumin was fed to a second inseminated control sow. Preservation of photoreceptor cells and morphology. Scott et al., 2015
2019 Quercetin and naringenin rd10 mice were used in a range of ages from P18 (when they are able to drink on their own) to P45 (peak of death of cone photoreceptors). Stock solutions of naringenin and quercetin were prepared in DMSO and added to their drinking water, resulting in a dose of 100 mg/kg/die per animal Preservation of photoreceptor cells and visual function. Downregulation of SOD1 and SOD2, as well as a reduction in lipid peroxidation. Piano et al., 2019
2010 Resveratrol Mice were orally administered with vehicle or resveratrol at a dose of 50 mg/kg body weight (BW) by using a gastric intubation daily for 5 days until light exposure. Preservation of photoreceptor cells and visual function. Reduction of TUNEL-positive cells. Suppression of SIRT1 and AP-1. Kubota et al., 2010
2018 Resveratrol Mice were injected intraperitoneally with RSV at a dose of 22.4 mg/kg body wt per day for 7 consecutive days before the ERG. On the third day of RSV administration, the animals’ pupils were dilated with atropine sulfate solution. Then the mice were separated into individual boxes and exposed to bright continuous light for 12 h to induce retinal degeneration. Preservation of photoreceptor cells and visual function. Liu et al., 2018
2019 Resveratrol - Prodrug JC19 Rd10 mice postnatal day 13 (P13) were anesthetized with ketamine/xylazine (80/12 mg/kg BW) and subretinally injected with 1 of 5% DMSO (vehicle), 0.1 mMJC19 or 5.0 mMJC19. Preservation of photoreceptor cells and visual function. Valdés- Sanchez et al., 2019
2023 Resveratrol RSV was dissolved in dimethyl sulfoxide (DMSO) at 65 mg/ mL and then diluted with phosphate buffer saline to get a 6.67% concentration of DMSO. RSV was administered to the rats of the R group by gavage at two modes: (1) a dose of 200 mg/kg at 0 h (h), 2, 10 h after the MNU-administration, and then once a day for the following two days; (2) a daily dose of 50 mg/kg 3 days (d) prior to and 3 d after the MNU-administration Preservation of photoreceptor cells and visual function. Reduction of TUNEL-positive cells. Suppression of SIRT1. Yan et al., 2023
2024 Resveratrol - Prodrug Piceid Octanoate Postnatal day 14 (P14) rd10 mice were anesthetized with ketamine/xylazine (80/12 mg/kg body weight) and intravitreally injected with 1 μL of 10 mM PIC-OCT diluted in 5% DMSO Preservation of photoreceptor cells and visual function. Reduction of TUNEL-positive cells. Moshtaghion et al., 2024
2022 Epigallocatechin gallate EGCG was dissolved in normal saline injection (10 mg/mL). EGCG was freshly prepared, and the rats were given an IP injection at a dose of 25 mg/kg every wk from postnatal day (P) 100 to P160, with a total of 7 doses administered Preservation of visual function, improved antioxidant enzymes and reduced oxidative damage. Perdices et al., 2022
2015 Green tea extract - epigallocatechin gallate Female rats received a single intraperitoneal injection of 40 mg/kg MNU at 7 wk of age. A daily dose of 250 mg/kg GTE was orally administered by gastric intubation starting 3 days before MNU administration, and administration was continued once daily for a maximum 10 days Preservation of photoreceptor cells. Reduction of TUNEL-positive cells. Downregulation of H0-1. Emoto et al., 2015
2014 Green tea extract - epigallocatechin gallate The oral administration of 250 mg/kg/day GTE was initiated 3 days before MNU injection and continued once daily throughout the experiment. Rats were sacrificed at 12, 24, and 72 h and 7 days after MNU injection, and the eyes were examined morphologically and morphometrically. Preservation of photoreceptor cells and morphology. Emoto et al., 2014
2023 Epigallocatechin gallate and lutein RP model mice were administered with an MNU solution after two wk of intragastric administration. The gavage dose of LUT was set at 100 mg per kg (BW per day), 16 the gavage dose of EGCG was set at 500 mg per kg (BWper day), and the ratio between LUT and EGCG was referred from the previous study. 18 LUT was dissolved in soybean oil, and EGCG was dissolved in water to prepare the corresponding concentration solution. The quality of LUT in the LUT-EGCG group is equivalent to that in the LUT and EGCG groups alone. The dose of MNU was set at 50 mg kg. Preservation of photoreceptor cells and morphology. Upregulation of antiapoptotic genes. Downregulation of IL-6. Jing et al., 2023
2017 Epigallocatechin gallate EGCG was dissolved in 0.9% sodium chloride injection (10 mg/mL). EGCG was freshly prepared, and mice were given an IP injection at a dose of 50 mg/kg. An equal volume of 0.9% sodium chloride was used as a control. EGCG was given daily for 7 days to some mice before they were sacrificed. For some mice, daily IP injections of EGCG were given for 7 days before light-induced degeneration and also after until the mice were sacrificed. Preservation of photoreceptor cells and visual function. Downregulation of SOD1, SOD2 and Gpx4. Qi et al., 2017
2012 Anthocyanin extracts Anthocyanin extracts (50 mg/kg) were daily administered orally (p.o.). The effects of anthocyanin treatment were evaluated at 1,2, and 4 wk after administration in MNU-injected rats. Preservation of photoreceptor cells, morphology and visual function. Downregulation of GFAP. Paik et al., 2012
2016 Anthocyanin - peonidin Two hours after MNU administration, the mice of the experimental group received an intravenous injection of peonidin at a dose of 80 mg/kg body weight. Thereafter, they received an intravenous injection of peonidin once a day for four consecutive days. Meanwhile, the mice of the vehicle-treated group received intravenous injections of physiological saline containing 0.05% acetic acid. Preservation of photoreceptor cells, morphology and visual function. Tao et al., 2016a
2014 Cyanidin-3-O- glucoside (C3G) Experimental RD was induced in rats by the intraperitoneal injection of N-methyl-N-nitrosourea (MNU) at 50 mg/kg. C3G extracted from mulberry (Morus alba L.) fruit (50 mg/kg) was orally administered, daily for 1, 2 and 4 weeks after MNU injection. Preservation of photoreceptor cells and morphology. Downregulation of GFAP. Lee et al., 2014
2016 Cyanidin-3-O- glucoside (C3G) After a week-long adaptation period, the rabbits were randomly divided into five groups (n = 8 per group), as follows: normal group (no light exposure and vehicle administration; NG), light-induced retinal damage model group (18 000 lx light exposure and vehicle administration; MG), the C3G group (18 000 lx light exposure and administration of C3G, 50.00 mg/kg/day), the PCA group (18 000 lx light exposure and administration of PCA, 17.15 mg/kg/day), and the FA group (18 000 lx light exposure and administration of FA, 21.61 mg/kg/day). C3G, PCA, and FA were administered at the same molar concentration for each group (0.11 mmol/kg body weight). Preservation of photoreceptor cells and morphology. Reduced TUNEL-positive cells. Downregulation of inflammation. Modulated apoptotic-associated genes. Wang et al., 2016

C3G: Cyanidin-3-O-glucoside; DMSO: dimethyl sulfoxide; EGCG: epigallocatechin gallate; FA: ferulic acid; GFAP: glial fibrillary acidic protein; IP: intraperitoneal injection; MNU: N-methyl-N-nitrosourea; PCA: protocatechuic acid; RD: retinal degeneration; RD1: retinal degeneration 1; RSV: resveratrol; SOD1-2: superoxide dismutase 1 and 2; TUNEL: terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling; UPR: unfolded protein response.

In addition to the animal models mentioned above, which only cover autosomal recessive mutations, there are also important models for studying autosomal dominant mutations, such as the RPE65 mice models, known as RPE65rd12 and RPE65R91W knock-in. Both present a mutation in gene Rpe65, an important gene to RPE function and photoreceptor maintenance, being RD12 crucial in the development of Luxturna® (Liu et al., 2024). Still, there are also mutant-rhodopsin carrying models, like RhoP23H and Rho-/-, a knockout model) (Barwick and Smith, 2023; Vasudevan et al., 2024). Finally, and less utilized, the NR2E3-associated models (RD7 mice) (McNamee et al., 2024). This covers only partially the main used animal models, since there are induced models such as the N-methyl-N-nitrosourea-induced (MNU), which consists of systemic injection of MNU, a DNA-alkylating agent that targets rod photoreceptors, leading to apoptosis, very similar to the animal models described above (Tao et al., 2015). Another chemically induced model is the sodium iodate (NaIO₃)-induced retinal degeneration, an intravenous or intraperitoneal injection of sodium iodate that causes oxidative stress and direct toxicity to RPE, leading to secondary photoreceptor death (Kannan and Hinton, 2014). Still, there is non-chemically induced degeneration, such as light-induced retinal degeneration, that consists of prolonged exposure to intense light (Kubota et al., 2010; Grimm and Remé, 2013). Lastly, in vitro models are becoming more relevant and reliable, particularly with the advent of retinal organoids that allow a controlled and reproducible environment for studying retinal development and disease, offering a cleaner translation to humans, since the organoids are originated from induced pluripotent human cells (Schnichels et al., 2021; Móvio et al., 2023; Bovi Dos Santos et al., 2024).

In summary, whether using genetic, induced, or in vitro models, each approach provides unique insights into the complex mechanisms of retinal degeneration, offering precise temporal control over treatment protocols. These models enable a broad spectrum of studies, from preventive strategies to therapeutic interventions, spanning genetic, pharmacological, and nutraceutical therapies.

Quercetin, naringenin & myricetin

Quercetin is one of the most extensively studied flavonoids for the treatment of retinopathies, especially in RP (Ortega and Jastrzebska, 2021). Primarily present in food sources such as onions, buckwheat, citrus fruits, grapes, cherries, and others (Wang et al., 2022a), this compound has been associated with a range of biological activities, including anti-aging, anti-inflammatory, antioxidant, and several others (Alizadeh and Ebrahimzadeh, 2022).

In 2017, Herrera-Hernández et al. showed several in silico analyses that treatment of mutated Rho opsin associated with RP with quercetin shows an allosteric modulation of the opsin, having a positive effect on the stability and conformational properties, indicating a good target for a therapeutic approach. Later in 2019, Ortega et al. described the same effects for quercetin, myricetin, and their mono-glycosylated forms known as quercetin-3-rhamnoside and myricetrin (Figure 2A, B, F, and G). The same group also examined the relationship between these compounds and inflammatory response in P23H knock-in mice, administering 20 mg/kg every other day for 7 days resulted in a significant reduction in pro-inflammatory markers (Ortega and Jastrzebska, 2021).

Figure 2.

Figure 2

Chemical structure of the polyphenols cited in pre-clinical data.

Dietary flavonoids are predominantly present in their glycoside forms. However, in the plasma, glycosides are rarely found. Deglycosylation of flavonoids occurs in both the small and large intestines, depending on the type of sugar moiety attached. Interestingly, some flavonoid metabolites exhibit stronger or weaker physiological activities than their precursor compounds (Murota et al., 2018; Muñoz-Reyes et al., 2021).

The flavonoids further enhanced structural rigidity and promoted receptor self-association to biological membranes, making them even more intriguing by treating cells expressing the P23H opsin mutation — showing partial restoration to normal cellular trafficking, a feature observed in RP-linked rho mutations. In another report, authors also investigated these flavonoids in a P23H mice model with chronic treatment (20 mg/kg every other day for 7 and 14 days), showing improvement of this receptor stability and function, leading to improved retinal morphology and function electroretinography (ERG) by increased rod and cone opsins. This was achieved by reducing oxidative stress markers by the free radical scavenger property of these flavonoids and downregulation of the unfolded protein response signaling (Ortega et al., 2022).

Considering the potential therapy focused on quercetin potentials, in 2019 Piano et al. evaluated the nutraceutical therapy by treating Rd10 animal model with quercetin and naringenin, another flavonoid (Figure 2A and C). The chronic treatment with quercetin or naringenin (100 mg/kg/day) showed preserved retinal morphology and improved functionality via ERG, evidenced by a reduction in cone PR loss and ROS levels in the retina, as assessed through oxidative stress markers such as SOD1, SOD2, and acrolein. Subsequently, the authors investigated both flavonoids in another animal model, Tvrm4 mice, which are heterozygous for an I307N dominant mutation of Rho. In this study, they assessed the effects of each flavonoid individually and in combination, evaluating functional, morphological, and molecular biological parameters. When administered separately, the flavonoids reduced oxidative stress and apoptosis markers. However, co-treatment resulted in a state described as “anti-oxidative stress,” well-known as anti-oxidative imbalance (50 + 100 mg/kg/day of naringenin and quercetin, respectively) (Piano et al., 2022). In acute models of light-induced retinal degeneration, intraperitoneally administration of quercetin and myricetin at a dose of 20 mg/kg 30 minutes before light exposure showed incredible results in the rescue of photoreceptors, preserving ERG responses, and reducing cell death pathways via apoptosis and inflammatory response (Ortega et al., 2021). In a model of all-trans-retinal induced photoreceptor atrophy and degeneration, quercetin was able to alleviate cell death by reducing ROS generation (Yang et al., 2024). Another flavonoid, kaempferol, when administered at 20 mg/kg for 5 days after light exposure, resulted in a reduction of PR cell death and significant ERG improvements (Noguchi et al., 2023). Those results summarize the potential of flavonoids in treating PR degenerating diseases.

Curcumin

Curcumin is considered a bioflavonoid, abundant in plant-derived curry spice, the turmeric. This compound has been extensively studied due to its multi-targeted applicability, known as anti-inflammatory, antioxidant, anti-diabetic, anti-cancer, and anti-aging (Kotha and Luthria, 2019; Figure 2D). Based on this description, curcumin was utilized as a treatment for AD (Allegrini et al., 2022), Parkinson’s disease, and multiple sclerosis (Bássoli et al., 2023).

For RP, curcumin presented exciting results in animal models: when administered orally to P23H rats, curcumin was able to cross the BRB being found in the retina, RPE, sclera and vitreous. Curcumin induced a dissociation of mutated protein aggregation, ultimately diminishing endoplasmic reticulum stress. Further improved retinal structure and function, as well as the gene expression and localization of rhodopsin and other important genes to photoreceptors; this was achieved with a dose of 100 mg/kg orally for 40 days (Vasireddy et al., 2011). The prenatal exposure in embryonic transgenic P23H miniswine the same dose of curcumin was also able to reduce the degeneration, preventing the mutant rhodopsin mistrafficking, reaching similarity to control, with rod preserved morphology (Scott et al., 2015). Further supporting flavonoids in NUT mutation-independent, curcumin was also evaluated in Rd1 mice. A 7-day treatment by intravitreal injection (10 µM/L) leads to potent immunomodulatory effects in microglial cells, including suppression of their activation and amelioration of rod loss, resulting in a better ERG response (Wang et al., 2017).

Resveratrol

Resveratrol is a polyphenol found in various plants, including grapes, blueberries, raspberries, and peanuts (Figure 2E). Similarly to the other compounds described before, it presents a multi-targeted applicability. Unique to this compound is its property to activate Sirt1, a NAD+-dependent deacetylase. Sirt1 activity is stimulated by high levels of NAD+, which deacetylates lysine residues on histone and cytoplasmic proteins, thereby influencing histone stability, transcriptional activity, and translocation of specific proteins. Sirt1 is reported to be located not only in the nucleus, but also in the mitochondria and plays a role in stress response pathways, such as apoptosis and inflammation, highlighting this polyphenol as a promising therapeutic target (Najafi et al., 2021). In a light-induced retinal degeneration model, pre-treatment with 50 mg/kg for 5 days prior to exposure resulted in reduced cell death and improved ERG responses (Kubota et al., 2010).

Liu et al. (2018) evaluated both in vitro with 661W photoreceptors lineage cells and in vivo light-induced retinal degeneration mice and Sirt1 pathways involved. In vitro analysis demonstrated potential neuroprotective effects, reducing cell death via ROS reduction, improving the ratio of reduced/oxidized glutathione/glutathione disulfide, and mitigating mitochondrial dysfunction. In vivo injections intraperitoneally of resveratrol for seven consecutive days at a dose of 22.4 mg/kg before the light damage inhibited retinal dysfunction and photoreceptor cell loss, reducing the expression of parthanatos-related proteins. In the Rd10 animal model, treatment via subretinal injection at a dose of 5 mM of resveratrol showed positive dose-dependent effects on function and retinal preservation (Valdés-Sánchez et al., 2019).

The positive results mentioned above were not observed in the MNU rat model when resveratrol was administered by gavage at a dose of 50 mg/kg. This discrepancy is discussed as a matter of the model itself, attributed to the MNU-induced depletion of NAD+, which prevented the deacetylation of SIRT1 and subsequently hindered the beneficial effects (Yan et al., 2023). More recently, a novel acylated resveratrol prodrug, piceid octanoate, has been utilized in both in vitro studies and in Rd10 mice models, demonstrating several positive outcomes. These include promoting SIRT1 nuclear translocation, preserving the NAD+/NADH ratio, and suppressing intracellular ROS formation. These effects are attributed to the upregulation of antioxidant genes and the preservation of mitochondrial function. In rd10 mice, a single intravitreal injection of PIC-OCT at a dose of 10 mM inhibited PARP formation, increased Sirt1 expression, significantly reduced the number of TUNEL-positive cells, preserved ERG responses, and enhanced performance in light chamber tests (Moshtaghion et al., 2024).

These results underscore the potential properties of polyphenols, especially flavonoids, in the treatment of photoreceptor degeneration, independent of the mutation carried, by its multifaceted nature potential, modulating primarily an antioxidant response in the retina, affecting also other important pathways that ultimately lead to retinal degeneration.

Epigallocatechin gallate

Epigallocatechin gallate (EGCG) is the most abundant catechin-based flavonoid in green tea leaves, described as antioxidant, anti-inflammatory, neuroprotective, and others (Figure 2H). The systemic treatment with EGCG in a dose of 25 mg/kg every week in the P23H animal model showed better visual acuity and retinal electrical function when compared to the vehicle group, these results being linked to balancing of antioxidant enzymes and oxidative damage (Perdices et al., 2022). When RP-induced degeneration by MNU model was pre-treated orally with green tea extract, which includes approximately 54% of EGCG, resulted in a decreased number of TUNEL-positive cells and ONL preservation, with the suppression of PR cell death (Emoto et al., 2015). Another study used EGCG in a chronic isolated or a co-treatment with lutein, another natural compound classified as a carotenoid, demonstrated synergic effects, rescuing the ONL by reducing pro-inflammatory cytokines, this was achieved at doses of 500 mg/kg of EGCG (Jing et al., 2023a). In support of these findings, the study demonstrated an increased presence of EGCG in the retina of treated animals, confirming that the green tea extract successfully crossed the blood–retinal barrier. Specifically, animals received a dose of 250 mg/kg/day for 3 days prior to MNU injection, resulting in retinal EGCG levels of 117.7 nM at 1 hour and 0.213 nM at 2 hours post-administration (Emoto et al., 2014). When administered seven days before light-induced damage at 50 mg/kg, EGCG attenuated the PR degeneration (Qi et al., 2017).

Anthocyanins

Anthocyanins, plant pigments abundant in vegetables and fruits such as sweet potato and eggplant, are commonly applied as food colorants. These compounds serve as both exogenous and intrinsic antioxidants, with documented benefits extending to visual acuity (Shen et al., 2022). This class of flavonoids has been described to have several positive impacts on degenerative processes of the retina. These include reducing GFAP expression and inhibiting important apoptosis mediators in photoreceptor cells (Paik et al., 2012; Tao et al., 2016a). Treatment with peonidin at the dose of 80 mg/kg after 4 days of MNU administration showed some interesting results, ameliorating the PR degeneration and preserving the inner visual signal pathways (Figure 2I). Using a multi-electrode array and ERG showed preserved retinal function, later demonstrated by histological analysis, impacting both scotopic and photopic functions (Paik et al., 2012; Tao et al., 2016b). This positive effect of anthocyanins was also described with the anthocyanins of black soybean and kuromanin chloride (Figure 2J; Paik et al., 2012; Lee et al., 2014). Orally administered cyanidin-3-glucoside for 3 weeks before light exposure prevented retinal damage (Wang et al., 2016).

Nanocarriers and Synergies Between Therapies as Innovative Strategies for Delivering Nutraceuticals

The applicability of flavonoids remains a concern, primarily due to their lower bioavailability resulting from poor gastrointestinal absorption, which is attributable to their hydrophobicity, or even first-pass metabolism. In this sense, several strategies have been proposed to overcome this issue, nanocarrier systems have been proposed as a significant one (Koirala et al., 2016; Puri et al., 2022). Among the main nanotechnologies applied to flavonoid delivery, several reports highlight phytosomes (Chen et al., 2022b), nanodiamonds (Gismondi et al., 2015; Chipaux et al., 2018), inorganic nanoparticles (Sysak et al., 2023), and others have shown promising results in enhancing the effectiveness of bioactive compounds, for example enhancing anti-inflammatory and antioxidant effects (Bunkar et al., 2019; Ghezzi et al., 2021; Gorantla et al., 2021; Kaushal et al., 2022; Ashfaq et al., 2023; Rakotondrabe et al., 2023; Sarabia-Vallejo et al., 2023; Yuan et al., 2024; Additional Table 6).

Additional Table 6.

Nanocarriers, compositions, advantages, and limitations

Carries Compositions Synthesis methods Advantages Limitations References
Liposomes Liposomes comprise phospholipid bilayers, which can encapsulate hydrophilic and hydrophobic compounds. Polyphenols and flavonoids are typically encapsulated within the aqueous core or the lipid bilayers. Typically formed by hydrating a thin film of lipids (e.g., phosphatidylcholine) in an aqueous solution containing polyphenols or flavonoids. This is followed by sonication or extrusion to form small vesicles. Enhanced bioavailability: protects polyphenols and flavonoids from degradation and enhances absorption. Targeted delivery: can be surface-modified to target specific tissues or cells. Biocompatible: generally well-tolerated by the body. Concentration: higher loads of both hydrophilic and hydrophobic drugs, controlled release. Limited stability: Liposomes can be susceptible to degradation over time. Production costs: can be expensive to produce on a large scale. Size variability: size distribution can vary, affecting consistency in dosing. Gorantla et al., 2021; Yuan et al., 2024; Ashfaq et al., 2023; Kaushal et al., 2022
Micelles Micelles are formed from amphiphilic molecules (surfactants) in aqueous solutions, with a hydrophobic core and hydrophilic shell. They can encapsulate hydrophobic compounds like certain polyphenols and flavonoids. Micelles self-assemble in aqueous solutions above a critical concentration (critical micelle concentration), forming spherical structures with a core-shell morphology. Solubilization: improves the solubility of hydrophobic polyphenols and flavonoids. Enhanced bioavailability: facilitates absorption through biological membranes. Ease of formulation: generally simple to prepare and stabilize Stability concerns: can be sensitive to dilution and environmental factors. Limited loading capacity: may not accommodate high concentrations of polyphenols or flavonoids. Biocompatibility: Surfactants used in micelle formation may have toxicity concerns at higher concentrations. Bunkar et al., 2019; Gorantla et al., 2021; Kaushal et al., 2022
Phytosomes Phytosomes are complexes of polyphenols or flavonoids incorporated into the polar heads of phospholipids, typically phosphatidylcholine. Prepared by combining polyphenols or flavonoids with phospholipids under controlled conditions to form complexes, such as solvent evaporation, freeze-drying, and antisolvent precipitation. Enhanced bioavailability: protects polyphenols and flavonoids from degradation, enhances permeability, stability, absorption, and solubility. Targeted delivery: can be designed for specific absorption pathways. Specificity of phospholipids: The choice of phospholipid can affect the characteristics of the phytosome. Production complexity: requires careful formulation and manufacturing processes. Potential variability: composition and performance can vary based on formulation and manufacturing conditions. Yuan et al., 2024; Chen et al., 2022b; Kaushal et al., 2022
Niosomes Niosomes are non-ionic surfactant vesicles that encapsulate hydrophilic and hydrophobic compounds like polyphenols and flavonoids. Formed by hydration of a mixture of non-ionic surfactants and cholesterol, followed by sonication or extrusion to create vesicles. Biocompatibility: generally well-tolerated and biodegradable. Improved bioavailability and bioaccessibility: enhances absorption in the gastrointestinal tract. Versatility: can encapsulate a wide range of bioactive compounds. Cost- effective: less expensive compared to other vesicular systems like liposomes. Potential instability: may aggregate or fuse over time, affecting stability. Variable release profiles: requires careful formulation to achieve the desired release. Scalability: Large-scale production can present challenges. Gorantla et al., 2021;
Nanoemulsions or Microemulsions Lipid-based colloidal dispersions of oil and water stabilized by a mixture of surfactants or emulsifiers, with droplet sizes in the nanometer range. Prepared by methods such as high-pressure homogenization or sonication, where polyphenols or flavonoids are dispersed in an oil phase and emulsified in an aqueous phase. Enhanced stability: long-term stability of encapsulated compounds, protection from adverse storage conditions, degradation, light, pH, and elevated temperature. Improved bioavailability and bioaccessibility: enhances absorption due to small droplet size and increased surface area. Versatility: can encapsulate both hydrophilic and hydrophobic compounds Production complexity: requires specialized equipment for homogenization or sonication. Surfactant toxicity: potential toxicity concerns with surfactants used to stabilize nanoemulsions. Cost: production costs can be higher compared to conventional formulations. Ashfaq et al., 2023; Kaushal et al., 2022
Self- nanoemulsifying system Formulations that spontaneously form nanoemulsions in the gastrointestinal tract under mild agitation or digestion conditions. Typically contain a mixture of oil, surfactant, co-surfactant, and polyphenols or flavonoids, which form a nanoemulsion upon exposure to aqueous media/biological fluids. Enhanced absorption: facilitates solubilization and absorption ofpoorly water- soluble polyphenols or flavonoids. Improved stability: protects compounds from degradation in the gastrointestinal environment. Reduced variability: provides consistent dosing due to self-emulsification properties. Digestive variability: The efficiency of self-emulsification can vary depending on digestive conditions. Bioavailability challenges: ensuring adequate absorption across intestinal membranes can be complex, also low loading capacity and leakage. Ashfaq et al., 2023; Kaushal et al., 2022
Solid lipid nanoparticles (SLNs) SLNs are composed of solid lipids (e.g., triglycerides or waxes) with a crystalline nature, that encapsulate polyphenols or flavonoids within a solid lipid matrix. SLNs are typically prepared by dispersing melted lipids containing polyphenols or flavonoids in an aqueous surfactant solution, followed by homogenization or sonication to form nanoparticles. High loading: efficient encapsulation of polyphenols and flavonoids due to high lipid content. Improved stability: protects encapsulated compounds from degradation. Sustained release: can provide sustained release profiles. Potential lipid crystallization: may affect long-term stability. Limited drug loading: Some polyphenols or flavonoids may not efficiently load into SLNs. Size distribution: Control over particle size distribution can be challenging. Ashfaq et al., 2023; Kaushal et al., 2022; Yuan et al., 2024;
Nanostructured lipid carriers (NLCs) Nanostructured lipid carriers (NLCs), introduced as the next generation LNPs, are composed ofa core containing a combination of solid and liquid lipids at room temperature. NLCs are typically produced by high- pressure homogenization or solvent evaporation methods, where lipid mixtures are melted and mixed with the bioactive compounds, followed by solidification. Aim to overcome the shortcomings of SLNs, for instance, increased drug loading capacity and improved stability by preventing drug leakage from the nanoparticles during storage High drug loading capacity: can incorporate high amounts ofpolyphenols/flavonoids. Enhanced stability & bioavailability: protects encapsulated compounds from degradation. Controlled release: provides sustained and controlled release of the bioactive. Complex formulation: requires optimization of lipid mixtures and surfactant ratios. Polymorphic transitions: potential changes in lipid crystal forms can affect stability. Scalability issues: Large- scale production can be challenging. Bunkar et al., 2019; Ashfaq et al., 2023; Yuan et al., 2024;
Exosomes Exosomes are nanosized extracellular vesicles naturally secreted by cells, composed of lipid bilayers containing proteins, lipids, and nucleic acids. Exosomes are isolated from cell culture supernatants or bodily fluids. They can potentially encapsulate bioactive molecules like polyphenols and flavonoids through engineering or loading processes. Biological compatibility: natural carriers for cell-to-cell communication. Targeting capability: can be engineered to target specific cells or tissues. Stability: exhibits stability in circulation, biological fluids, and low immunogenicity. Complex isolation: isolation and purification methods can be challenging. Loading efficiency: The loading of polyphenols and flavonoids into exosomes can be variable. Yuan et al., 2024
Polymeric micelle Polymeric micelles are self- assembled nanoparticles formed from amphiphilic block copolymers (e.g., PEG-PLA, PEG-PCL), which can encapsulate hydrophobic drugs such as polyphenols and flavonoids in their core. Prepared by dissolving block copolymers in a solvent, followed by evaporation or dialysis to form micelles. Enhanced solubility: improves the solubility of hydrophobic polyphenols and flavonoids. Targeted delivery: can be surface-modified for targeted delivery to specific tissues. Concentration: high loading capacity. Enhanced stability: protection from adverse conditions, like pH and elevated temperature. Potential toxicity: Some polymers used may have toxicity concerns. Size distribution: Control over size distribution can be challenging. Biocompatibility: compatibility with biological systems needs careful consideration, due to low stability and poor permeability. High cost. Yuan et al., 2024; Kaushal et al., 2022; Gorantla et al., 2021.
Polymeric nanoparticle Polymeric micelles are self- assembled nanoparticles formed from amphiphilic block copolymers (e.g., PEG-PLA, PEG-PCL), which can encapsulate hydrophobic drugs such as polyphenols and flavonoids in their core. Typically formed by methods such as nanoprecipitation, emulsion/solvent evaporation, or electrospraying, where polymers and polyphenols or flavonoids are mixed under controlled conditions. Biocompatibility: Biodegradable polymers are generally well-tolerated. Versatility: can encapsulate a wide range ofcompounds, including polyphenols and flavonoids. Controlled release: allows high encapsulation efficiency, controlled and sustained release. Potential toxicity of polymers: Depending on the polymer used, toxicity concerns may arise. Formulation complexity: requires optimization of formulation parameters. Complex production process. Gorantla et al., 2021; Ghezzi et al., 2021;
Dendrimers Dendrimers are branched, tree-like synthetic macromolecules with high surface functionality, suitable for drug delivery applications. Dendrimers are synthesized through repetitive branching reactions, creating multiple generations with numerous surface functional groups for drug attachment. High functionalization: multiple sites for drug conjugation enhance loading capacity. Controlled size and structure: well-defined architecture aids in the predictability of behavior. Enhanced solubility: improves the solubility and bioavailability ofpolyphenols and flavonoids Synthesis complexity: requires precise and multi-step synthetic processes. Potential toxicity: higher generations may exhibit cytotoxicity, and are also non-biodegradable. Purification challenges: extensive purification is needed to remove by-products. Gorantla et al., 2021; Ghezzi et al., 2021;
Nanocrystals Nanocrystals are pure solid drug particles of nanometer size, stabilized by surfactants or polymers. Nanocrystals are produced through top- down methods like high-pressure homogenization and milling, or bottom-up methods like antisolvent precipitation, involving reducing drug particle size to nanometer scale. Enhanced solubility: greatly improves solubility and dissolution rates of poorly water-soluble polyphenols and flavonoids. High drug loading: consists of nearly 100% drug, leading to high loading capacity. Bioavailability: increases bioavailability due to the large surface area. Stability issues: potential for aggregation, Ostwald ripening over time, and limited control over release profile. Processing complexity: requires high energy input and specialized equipment. Scalability: Large-scale production can be challenging and expensive. Stability issues like agglomeration. Yuan et al., 2024
Nanodiamonds Nanodiamonds are crystalline carbon nanoparticles with sizes typically less than 100 nanometers, suitable for drug delivery and imaging applications. Produced by detonation synthesis or chemical vapor deposition, and polyphenols or flavonoids can be encapsulated or surface-modified. Biocompatibility: well-tolerated by biological systems. Surface functionalization: can be functionalized with drugs or targeting moieties. High surface area: enables high drug loading capacity. Production cost: can be expensive compared to other nanoparticles. Size control: Achieving uniform size distribution can be challenging. Long-term effects: long-term biocompatibility and clearance need thorough investigation. Chipaux et al., 2018; Gismondi et al., 2015
Co-crystal Co-crystals are crystalline structures composed of the active pharmaceutical ingredient (e.g., polyphenols or flavonoids) and a co-former molecule, bonded through non-covalent interactions. Prepared by solvent evaporation, grinding, or slurry methods, where the active compound and co-former are mixed in a suitable solvent and allowed to crystallize. Improved properties: enhances the solubility and stability of polyphenols and flavonoids. Tailored properties: allows for precise control over physical and chemical properties. Enhanced bioavailability: protects polyphenols and flavonoids from degradation and enhances absorption. Synthesis complexity: requires optimization of conditions for co-crystal formation. Yuan et al., 2024
Cyclodextrin complex Cyclodextrin complexes are inclusion complexes formed by encapsulating polyphenols or flavonoids within cyclodextrins (e.g., P-cyclodextrin). Complexation occurs through methods such as kneading, co-precipitation, or freeze-drying, where polyphenols or flavonoids are mixed with cyclodextrins. Enhanced solubility: improves solubility and stability of polyphenols and flavonoids. Protection from degradation: increases stability against light, heat, and oxidation. Improved bioavailability and bioaccessibility: enhances absorption in the gastrointestinal tract. Speciflcity of cyclodextrins: Selection of the appropriate cyclodextrin and complexation conditions is critical. Complexation efficiency: not all polyphenols or flavonoids may form stable complexes, also has leakage problems. Toxicity: exhibit nephrotoxicity. Yuan et al., 2024; Sarabia- Vallejo et al., 2023
Metal nanoparticles (silver, gold, metal oxides, and others) Metal nanoparticles are composed of atoms arranged in nanoscale dimensions, often functionalized with ligands or coatings. Synthesized by reducing metal salts in the presence of stabilizing agents, allowing for the encapsulation of polyphenols or flavonoids. Biocompatibility: generally compatible with biological systems. Surface functionalization: This can be functionalized by targeting ligands for specific delivery. Therapeutic potential: used in imaging, diagnostics, and therapy. Synergy: allows multiple combinations of polyphenols. Improve the biological effects of polyphenols. Biological fate: clearance and biodistribution and long term toxicity, in vivo need thorough study. Environmental impact: potential environmental concerns related to nanoparticle disposal. Cost: production costs can be higher compared to other nanoparticles. Regulatory considerations. Sysak et al., 2023; Gorantla et al., 2021

NLCs: Nanostructured lipid carriers; PEG-PCL: polyethylene glycol-poly(ε-caprolactone); PEG-PLA: polyethylene glycol-polylactic acid; SLNs: solid lipid nanoparticles.

In general, nanocarriers were developed in an attempt to overcome physic-chemical limitations such as reduced aqueous solubility, protection from environmental degradation, and/or biological metabolism. Furthermore, new perspectives were open in terms of improved therapeutic effects since some of these delivery systems evoked changes in polyphenol biopharmaceutical profiles or even showed therapeutic potential when administered alone. These delivery systems must be designed to ensure easy patient compliance and retain the bioactive properties of the compound. Furthermore, they should target the site of action to maximize benefits and provide controlled release, depending on the treatment requirements (Yang et al., 2017). Yuan et al. (2024) extensively reviewed and summarized the potential of these delivery systems in general. Overall, these delivery systems are beneficial in protecting highly sensitive polyphenols, thereby ensuring enhanced bioavailability, sustainable release, and storage stability. Figure 3 displays the most used nanocarriers (with different compositions) and therapy associations.

Figure 3.

Figure 3

Potential delivery systems for polyphenols and conjugated therapies with nutraceuticals.

The top section illustrates various nanotechnology-based systems that enhance the efficacy, bioavailability, and safety of polyphenols and flavonoids as nutraceuticals. These systems include inorganic nanoparticles, lipid-based carriers, polymeric formulations, and nanodiamonds. The bottom section highlights the combination of therapies with nutraceuticals for improved effectiveness. Gene therapy employs advanced technologies like the CRISPR/Cas system for in vivo gene editing, facilitating personalized medicine by correcting patient-specific mutations. Concurrently, nutraceuticals can mitigate related pathophysiological processes. Cell transplantation strategies aim to replace lost cells using autologous hiPSCs, retinal organoids, and 3D bioprinting to reconstruct retinal structures. Additionally, flavonoids can work synergistically with other flavonoids, pharmaceuticals, and natural compounds in NUT therapy, enhancing overall therapeutic outcomes. Created with BioRender.com. 3D: Three-dimentional; CRISPR: clustered regularly interspaced short palindromic repeats; hiPSC: human induced pluripotent stem cells.

Among different nanosystems applied to flavonoid encapsulation, one of the most reported strategies is referred to as colloidal carriers, such as lipid nanocarriers, including liposomes, nanoemulsions, nanostructured lipid carriers (NLC), and solid lipid nanoparticles. Liposomes are lipid vesicles with high biocompatibility and improved capability for encapsulating flavonoids into both internal aqueous compartments and lipid bilayers, protecting them from chemical degradation. Most articles discuss flavonoid-loaded liposomes designed for skin delivery (Chen et al., 2022), but the main limitations of their applications as nutraceuticals rely on physicochemical instability against pH variation throughout the gastrointestinal tract.

In an attempt to overcome those limitations, solid lipid nanoparticles and NLC were proposed for flavonoid delivery due to their ability to promote flavonoid permeation across the intestinal barrier. Interesting results were achieved by using NLC for delivering quercetin, naringenin, and hesperidin since the bioavailability of those compounds was higher after encapsulation in NLC when compared to solid lipid nanoparticles (Aditya et al., 2014). In fact, those promising results can be attributed to the adequate transition temperature of the lipids mixture (at physiological temperature) incorporated into the NLC core, which promotes the physico-chemical stability of the compound and, consequently, enhances the transepithelial permeation (Hu et al., 2021).

Other recently studied colloidal carriers are polymeric micelles. Most reasons for proposing micelles as flavonoid delivery systems rely on their capability to improve the aqueous solubility of molecules, even considering short circulation time of the micelle. In particular, their reduced dimensions (< 100 nm), compared to other nanocarriers, favor intestinal permeation, as well as their composition (natural polymers such as chitosan, pullulan, and cellulose-derivatives), determines adhesion and enhances the contact between the nanocarrier and intestinal mucosa (Kaushal et al., 2022).

Despite the therapeutic potential of colloidal carriers, factors inherent to gastrointestinal administration drive the main biopharmaceutical limitations to be overcome are: (i) their chemical integrity and colloidal stability even considering the pH changes from stomach to intestinal mucosa; (ii) the dilution processes that occur during intestinal peristalsis; (iii) the contact with mucin layer on the intestinal surface mucosa; (iv) targeted-cellular delivery modulated by nanocarriers components coating. Other strategies were developed, such as metallic nanoparticles silver, gold, and zinc. Similarly, quercetin and hesperidin are used, especially for colorectal cancer treatment. Another important point is the possibility of combining stimuli-responsive components (pH, temperature, and oxidative stress sensors) for coating metallic nanoparticles, expanding their applications for intestinal delivery (Sysak et al., 2023).

The synergy between two different therapeutic strategies occurs when the combined effect of both therapies is greater than the sum of their individual effects. This synergistic interaction can enhance the overall efficacy of treatment, reduce side effects, and address multiple pathways through complementary and supportive actions (Leena et al., 2020). Such integration is well-utilized in fields such as cancer therapy, where chemotherapy induces cell death and immunotherapy enhances the immune response (Di Napoli et al., 2023). Also seen in chronic diseases such as diabetes, where lifestyle interventions and pharmacology are combined (Wronka et al., 2022). Additionally, there is growing interest in the synergistic effects of polyphenols and flavonoids when combined with other dietary natural products (Ulrich-Merzenich et al., 2009; Hidalgo, Sánchez-Moreno and de Pascual-Teresa, 2010; Vue et al., 2015; Leena et al., 2020; Piano et al., 2022; Zhang et al., 2023b).

There is growing evidence that nutraceutical therapies, when used synergistically with other treatments, can contribute to more integrated and effective medical care (Yang et al., 2017; Hussain et al., 2020; Di Napoli et al., 2023; Li et al., 2024). Multi-target treatments could be more effective than single-drug treatments, especially for neurodegeneration like RP (Maneu et al., 2022). While NUT utilizing polyphenols and flavonoids appear to be a reliable treatment option for RP or other PR degenerative conditions, it may not be effective for severe or late-stage retinal degeneration. In these cases, it could be beneficial to combine NUT with other emerging therapies, such as genetic and cell-based therapies, like transplantation of autologous-reprogrammed retinal cells, incorporating retinal organoids and 3D bioprint for cell sheet transplantation (Voisin et al., 2023; Bovi Dos Santos et al., 2024). This approach aims to ameliorate the pathophysiology of retinopathies while addressing cell loss and suppressing the immunological response to avoid rejection (Hyon et al., 2006; Hodge et al., 2019). Finally, genetic therapy could be aligned with nutraceuticals – treating the pathogenesis (genetic mutations) and the pathophysiological process (neuroinflammation and oxidative stress).

Doses, Safety Assessments, and Potential Adverse Effects of Polyphenols and Flavonoids

Although polyphenols and flavonoids show significant potential in the nutraceutical management of photoreceptor degeneration, current literature highlights a substantial gap in detailed dose-response and safety data specific to retinal degeneration. Pre-clinical studies in retinal degeneration models have used doses ranging from 20 to 500 mg/kg (Additional Table 5), demonstrating neuroprotective effects without significant toxicity. However, these doses are relatively high when compared to typical dietary intake. For instance, the average daily consumption of total flavonoids is estimated to be between 300 and 1800 mg, varying according to the geographical region studied (Grosso et al., 2014; Escobar‐Cévoli et al., 2017; Anacleto et al., 2019; Del Bo et al., 2019; Hejazi et al., 2020; Huang et al., 2020; Kapolou et al., 2021; Carnauba et al., 2023). To illustrate, to achieve a therapeutic dose of 100 mg/kg of a specific polyphenol in a 70 kg adult, one would theoretically need to consume approximately 7 g of anthocyanins per day, a quantity 4 to 23 times higher than the normal dietary intake. In contrast, clinical trials that utilize polyphenol supplementation typically range from 10 to 1200 mg/day (Additional Tables 14), which is far less than the doses used in animal models – none of the pre-clinical nor clinical trials published/showed adverse effects of the doses implemented.

Moreover, the challenges related to bioavailability and metabolic processing further complicate the translation of these doses to human clinical applications – challenges that nanocarrier-based delivery systems may help overcome (Figure 3 and Additional Table 6). Therefore, rigorous clinical trials focusing on dose optimization, long-term safety, and monitoring of potential adverse events are essential to establish practical guidelines for their use in PR degenerating diseases like RP. However, extrapolation to clinical use requires caution, as these compounds may present adverse effects such as pro-oxidant activity, interference with cytochrome P450 enzymes, and reduced mineral absorption at high concentrations. In cases of overdose, their protective effects may be countered by toxicological consequences (Abotaleb et al., 2018; Andres et al., 2018; Del Bo et al., 2019; Kapolou et al., 2021; Cianciosi et al., 2022).

Although many studies have confirmed the safety of flavonoids, their potential toxicity remains an important area of research due to the highly complex mechanisms involved. Current evidence indicates that natural flavonoid glycosides act on multiple molecular targets, with their effects varying according to dose in both in vivo and in vitro experiments. While most flavonoids are considered safe, those marketed as food supplements must have their tolerable upper intake levels thoroughly assessed, as there have been reports of toxic effects associated with excessive intake. It is also important to note that, although the observed adverse effects of polyphenols have not shown significant impacts on neurons, caution is warranted. Some commercially available flavonoid supplements may pose public health concerns due to the limited information available on adverse side effects and drug interactions in both in vivo experimental settings and clinical trials (Abotaleb et al., 2018; Andres et al., 2018; Del Bo et al., 2019; Kapolou et al., 2021; Cianciosi et al., 2022).

Concluding Remarks

The complex nature of neurodegenerative diseases increases the difficulty of finding suitable treatments that benefit most diagnosed patients. Nutraceutical therapies aim to use natural food-based products with medicinal benefits to improve health, prevent chronic diseases, delay the aging process, and support the structure and function of the body. This review highlights the potential of nutraceutical therapies, particularly polyphenolic compounds such as flavonoids, in addressing neurodegeneration in the retina, focusing on RP. These bioactive compounds show promise due to their multi-targeted roles, including antioxidant, anti-inflammatory, and neuroprotective effects.

However, significant challenges remain, particularly in enhancing their bioavailability and developing reliable delivery methods with cellular-specific recognition. Integrating advanced delivery systems, such as nanoformulations and biomaterials, offers a promising avenue to overcome challenges inherent to nanocarrier design and intestinal mucosa barrier. Moreover, the synergistic use of nutraceuticals with other emerging therapies, such as genetic and cell-based treatments, could provide a more comprehensive approach to treating retinal degenerative diseases. Considering those advancements, nutraceuticals could become a viable and accessible option for patients suffering from RP and other retinal diseases, independent of the mutation displayed, ultimately improving their quality of life. Still, while flavonoids and polyphenols present a promising therapeutic option for RP, further research and clinical trials are essential to optimize their delivery systems and establish their efficacy.

Additional files:

Additional Table 1: Clinical trials based on dietary supplements or nutraceutical therapy for AMD.

Additional Table 2: Clinical trials based on dietary supplements or nutraceutical therapy for diabetic retinopathy.

Additional Table 2.

Clinical trials based on dietary supplements or nutraceutical therapy for diabetic retinopathy

Clinical data ID Compounds Methods Results Years
NCT00893724 Inosine; tocopherols, tocotrienol, CoQ10 combination capsule; niacinamide SR; vitamin C; N-acetyl cysteine; complete multivitamin with all minerals and minocycline Group 1 (supplement): Oral administration of inosine [500 mg], tocopherols [200IU], tocotrienol [10 mg], CoQ10 [50 mg], niacinamide [750 mg] vitamin C [1000 mg], N-acetyl cysteine [600 mg], complete multivitamin with all minerals; Group 2 (supplement + minocycline): Oral administration of supplement + minocycline [50 mg] Completed, no results published. 2009
NCT01646047 Multi-component nutritional supplement in capsule (vitamin C, mixed tocopherols/tocotrienols, vitamin D, fish oil, lutein, zeaxanthin, pine bark extract, benfotiamine, green tea extract, curcumin). Two capsules containing nutritional supplements per day for 6 months. Improvements in visual function, hsCRP and peripheral neuropathy in patients with diabetes, both with and without retinopathy, and without affecting glycaemic control. 2012
NCT01880372 Alpha lipoic acid. 600 mg oral administration of lipoic acid daily for 12 months. Terminated, no results published. 2013
NCT03533478 Oral treatment Alzer and Diamel. Alzer group: Oral administration of one 500 mg tablet 3 times a day (1500 mg daily) after breakfast, lunch and dinner for one year. Diamel group: Oral administration of two 660 mg capsules 3 times a day (3960 mg daily) before breakfast, lunch and dinner for one year. No statistically significant, but clinical: severe macular edema did not progress in the alzer and diamel. 2018
NCT04117022 Carotenoid supplement of DVS formula [vitamins C, D3 and E (d-a tocopherol), zinc oxide, eicosapentaenoic acid, docosahexaenoic acid, a-lipoic acid (racemic mixture), coenzyme Q10, mixed tocotrienols/tocopherols, zeaxanthin, lutein, benfotiamine, N-acetyl cysteine, grape seed extract, resveratrol, turmeric root extract, green tea leaf, and pycnogenol (patented French 2 soft gels per day for 6 months. In progress. 2019
NCT03866005 Maritime Pine Bark extract, sp Pinus pinaster, Horphag Research, Geneva, Switzerland)]. Diabetes Visual Function Supplement (DiVFuSS), containing the macular carotenoids lutein and zeaxanthin, as well as antioxidants (vitamins B1, B12, C, D, E, lipoic acid, coenzyme Q10, resveratrol), omega3 fatty acids (EPA/DHA) and botanical extracts (Pycnogenol™ [patented extract of French maritime pine bark, Pinus pinaster], grape seed and green tea extracts, curcumin). Two DiVFuSS soft gels per day, during 24 months. Unknown status, no results published. 2019
NCT04742829 INTRAVIT(2 tablets: Curcumin 380mg, artemisia dracunculus L. 160mg, bromelain 160 mg, pineapple comosus stem d.e.i. 40 mg, total bromelain activity 394 GDU, pipeline 3.8 mg). 2 tablets per day morning and evening before meals, 6 months. 6 months of a dietary complementary supplement additionally to the present therapy for the treatment of diabetes, improves CRT, BCVA, and VD in patients with mild DME. 2021
NCT06376240 Pyridoxamine Treatment with A (either 300 mg pyridoxamine or placebo per day) in period 1 followed by treatment with B (either 300 mg pyridoxamine or placebo per day) in period 2. Recruiting 2024

Additional Table 3: Clinical trials based on dietary supplements or nutraceutical therapy for glaucoma.

Additional Table 3.

Clinical trials based on dietary supplements or nutraceutical therapy for glaucoma

Clinical data ID Compounds Methods Results Years
NCT00476138 Epigallocatechin-gallate (EGCG). Oral administration of EGCG treatment (200 mg/day) for 3 months. Reduced intraocular pressure. 2007
NCT01254006 Forskolin, rutin and vitamins B1 and B2. Oral administration of two tablets per day of a food supplement (15 mg forskolin, 200 mg of rutin, 0.7 mg of vitamin B1 and 0.8 mg of vitamin B2) for 30 days. Reduced intraocular pressure fluctuations. 2010
NCT01630551 Omega-3 fatty acid. 90-day supply of daily oral administration of a fish oil nutritional supplement. Withdrawn. 2012
NCT01930487 Optic Nerve Formula (composed of essential vitamins and minerals, omega-3poly unsaturated fatty acids, polyphenolic nutrients, amino acid, botanical extracts, and flax seed oil). Oral administration of the antioxidants, for one- month of antioxidants administered - three-weeks washout period. Increases in biomarkers of ocular blood flow within retinal and retrobulbar vascular beds. 2013
NCT02984813 GlaucoHealth (alpha lipoic acid, citicoline, Co-enzyme Q10, Ginkgo biloba extract, grape seed extract, N- acetyl-cysteine, curcumin, and green tea extract) & GlaucoSelect (curcumin, bilberry extract, and grape seed extract). 2 pills once daily in the morning for 3 months. Terminated, no results published. 2016
NCT04088084 Palmitoylethanolamide PEA 600 mg supplementation for 3 months. Protective effect on ganglion cells. 2019
NCT04380025 Mirtogenol (bilberry with pycnogenol). Daily dietary supplementation of mirtogenol and one drop of bimatoprost 0.01% for 24 weeks. No results found. 2020
NCT04460365 Lutein, zeaxanthin and meso-zexanthin Daily oral administration of one softgel capsule (10 mg Lutein, 2 mg zeaxanthin, 10 mg meso-zeaxanthin) for 18 months. Macular pigment optical density alterations, an increase in mesopic contrast sensitivity under glare. 2020
NCT04676126 Lumega-Z A macular pigment-containing medical food (Lumega-Z) was prescribed in combination with a topical carbonic anhydrase inhibitor. No results. No subjects enrolled. 2020
NCT04846179 Ginkgo biloba extract. Webber natural ginkgo biloba 120 mg soft gel, 1 tablet twice a day for 4 months. No significant increase in perfusion density of the macula and optic nerve head. 2021
NCT05080153 Ocufolin forte. 1 capsule/day after RVP measurement, till 90 tablets are finished (3 months) Significantly reduce retinal vascular pressure in the setting of glaucoma. 2021
NCT05527106 Citicoline and docosahexaenoic acid (DHA) Oral administration for 3 months of citicoline and DHA vs. citicoline and DHA vs. vitamin C. Improvement of visual field indices. 2022
NCT05695027 Nicotinamide and pyruvate Oral administration for 87 wk (20 months). Enrolling. 2023

DHA: Docosahexaenoic acid; EGCG: epigallocatechm-gallate.

Additional Table 4: Clinical trials based on dietary supplements or nutraceutical therapy for RP.

Additional Table 5: Pre-clinical data utilizing in vivo experimentation with retinal degeneration models and polyphenols as treatment.

Additional Table 6: Nanocarriers, compositions, advantages and limitations.

Acknowledgments:

The authors would like to acknowledge the support provided by their respective institutions during the preparation of this manuscript. They are especially grateful to Erick Bovi dos Santos (Universidade Federal do ABC, Brazil) for his valuable assistance in acquiring data for the meta-analysis.

Funding Statement

Funding: AHK is grateful for grants from Fundação de Amparo à Pesquisa do Estado de São Paulo (FAPESP, Brazil, #2020/11667-0) and Universidade Federal do ABC (UFABC, Brazil). The following authors were recipients of fellowships from FAPESP: THLV (#2021/11969-9 and #2024/00828-3), GBS (#2021/14227-3), and GMB (#2024/10858-7). The following authors were recipients of fellowships from Coordenação de Aperfeiçoamento de Pessoal de Nível Superior (CAPES, Brazil): MIM (Finance Code 001, #88887.597402/2021-00). The following authors were recipients of fellowships from Conselho Nacional de Desenvolvimento Científico e Tecnológico (CNPq, Brazil.): GKD (#145164/2024-1), and DRA (#308819/2022-5). The funders had no role in study design, data collection and analysis, decision to publish, or manuscript preparation.

Footnotes

Conflicts of interest: The authors declare no conflicts of interest.

C-Editors: Zhao M, Liu WJ, Qiu Y; T-Editor: Zhou H

Data availability statement:

Not applicable.

References

  1. Abdul-Latif R, Stupans I, Allahham A, Adhikari B, Thrimawithana T. Natural antioxidants in the management of Parkinson’s disease: Review of evidence from cell line and animal models. J Integr Med. 2021;19:300–310. doi: 10.1016/j.joim.2021.03.007. [DOI] [PubMed] [Google Scholar]
  2. Abotaleb M, Samuel SM, Varghese E, Varghese S, Kubatka P, Liskova A, Büsselberg D. Flavonoids in cancer and apoptosis. Cancers (Basel) 2018;11:28. doi: 10.3390/cancers11010028. [DOI] [PMC free article] [PubMed] [Google Scholar]
  3. Ahluwalia K, Martinez-Camarillo JC, Thomas BB, Naik A, Gonzalez-Calle A, Pollalis D, Lebkowski J, Lee SY, Mitra D, Louie SG, Humayun MS. Polarized RPE secretome preserves photoreceptors in retinal dystrophic RCS rats. Cells. 2023;12:1689. doi: 10.3390/cells12131689. [DOI] [PMC free article] [PubMed] [Google Scholar]
  4. Aidoo R, Abe-Inge V, Kwofie EM, Baum JI, Kubow S. Sustainable healthy diet modeling for a plant-based dietary transitioning in the United States. NPJ Sci Food. 2023;7:61. doi: 10.1038/s41538-023-00239-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  5. Ali MU, Rahman MSU, Cao J, Yuan PX. Genetic characterization and disease mechanism of retinitis pigmentosa; current scenario. 3 Biotech. 2017;7:251. doi: 10.1007/s13205-017-0878-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  6. Alizadeh SR, Ebrahimzadeh MA. Quercetin derivatives: Drug design, development, and biological activities, a review. Eur J Med Chem. 2022;229:114068. doi: 10.1016/j.ejmech.2021.114068. [DOI] [PubMed] [Google Scholar]
  7. Allegrini D, Raimondi R, Borgia A, Sorrentino T, Montesano G, Tsoutsanis P, Cancian G, Verma Y, De Rosa FP, Romano MR. Curcumin in retinal diseases: A comprehensive review from bench to bedside. Int J Mol Sci. 2022;23:3557. doi: 10.3390/ijms23073557. [DOI] [PMC free article] [PubMed] [Google Scholar]
  8. Anacleto SL, Lajolo FM, Hassimotto NMA. Estimation of dietary flavonoid intake of the Brazilian population: A comparison between the USDA and Phenol-Explorer databases. J Food Compost Anal. 2019;78:1–8. [Google Scholar]
  9. Andres S, Pevny S, Ziegenhagen R, Bakhiya N, Schäfer B, Hirsch-Ernst KI, Lampen A. Safety aspects of the use of quercetin as a dietary supplement. Mol Nutr Food Res. 2018 doi: 10.1002/mnfr.201700447. doi: 10.1002/mnfr.201700447. [DOI] [PubMed] [Google Scholar]
  10. Aoun M, Passerini I, Chiurazzi P, Karali M, De Rienzo I, Sartor G, Murro V, Filimonova N, Seri M, Banfi S. Inherited retinal diseases due to RPE65 variants: from genetic diagnostic management to therapy. Int J Mol Sci. 2021;22:7207. doi: 10.3390/ijms22137207. [DOI] [PMC free article] [PubMed] [Google Scholar]
  11. Ascunce K, Dhodapkar RM, Huang D, Hafler BP. Innate immune biology in age-related macular degeneration. Front Cell Dev Biol. 2023;11:1118524. doi: 10.3389/fcell.2023.1118524. [DOI] [PMC free article] [PubMed] [Google Scholar]
  12. Ashfaq R, Rasul A, Asghar S, Kovács A, Berkó S, Budai-Szűcs M. Lipid nanoparticles: an effective tool to improve the bioavailability of nutraceuticals. Int J Mol Sci. 2023;24:15764. doi: 10.3390/ijms242115764. [DOI] [PMC free article] [PubMed] [Google Scholar]
  13. Athanasiou D, Aguila M, Bellingham J, Li W, McCulley C, Reeves PJ, Cheetham ME. The molecular and cellular basis of rhodopsin retinitis pigmentosa reveals potential strategies for therapy. Prog Retin Eye Res. 2018;62:1–23. doi: 10.1016/j.preteyeres.2017.10.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  14. Atienzar-Aroca S, Flores-Bellver M, Serrano-Heras G, Martinez-Gil N, Barcia JM, Aparicio S, Perez-Cremades D, Garcia-Verdugo JM, Diaz-Llopis M, Romero FJ, Sancho-Pelluz J. Oxidative stress in retinal pigment epithelium cells increases exosome secretion and promotes angiogenesis in endothelial cells. J Cell Mol Med. 2016;20:1457–1466. doi: 10.1111/jcmm.12834. [DOI] [PMC free article] [PubMed] [Google Scholar]
  15. Augustin S, Lam M, Lavalette S, Verschueren A, Blond F, Forster V, Przegralek L, He Z, Lewandowski D, Bemelmans AP, Picaud S, Sahel JA, Mathis T, Paques M, Thuret G, Guillonneau X, Delarasse C, Sennlaub F. Melanophages give rise to hyperreflective foci in AMD, a disease-progression marker. J Neuroinflammation. 2023;20:28. doi: 10.1186/s12974-023-02699-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  16. Azam M, Jastrzebska B. Mechanisms of rhodopsin-related inherited retinal degeneration and pharmacological treatment strategies. Cells. 2025;14:49. doi: 10.3390/cells14010049. [DOI] [PMC free article] [PubMed] [Google Scholar]
  17. Bakri SJ, Thorne JE, Ho AC, Ehlers JP, Schoenberger SD, Yeh S, Kim SJ. Safety and efficacy of anti-vascular endothelial growth factor therapies for neovascular age-related macular degeneration: a report by the american academy of ophthalmology. Ophthalmology. 2019;126:55–63. doi: 10.1016/j.ophtha.2018.07.028. [DOI] [PubMed] [Google Scholar]
  18. Bardak H, Uğuz AC, Bardak Y. Curcumin regulates intracellular calcium release and inhibits oxidative stress parameters, VEGF, and caspase-3/-9 levels in human retinal pigment epithelium cells. Physiol Int. 2017;104:301–315. doi: 10.1556/2060.104.2017.4.3. [DOI] [PubMed] [Google Scholar]
  19. Barhoum R, Martínez-Navarrete G, Corrochano S, Germain F, Fernandez-Sanchez L, de la Rosa EJ, de la Villa P, Cuenca N. Functional and structural modifications during retinal degeneration in the rd10 mouse. Neuroscience. 2008;155:698–713. doi: 10.1016/j.neuroscience.2008.06.042. [DOI] [PubMed] [Google Scholar]
  20. Barwick SR, Smith SB. Comparison of mouse models of autosomal dominant retinitis pigmentosa due to the P23H mutation of rhodopsin. Adv Exp Med Biol. 2023;1415:341–345. doi: 10.1007/978-3-031-27681-1_49. [DOI] [PubMed] [Google Scholar]
  21. Bássoli RMF, Audi D, Ramalho BJ, Audi M, Quesada KR, Barbalho SM. The effects of curcumin on neurodegenerative diseases: a systematic review. J Herb Med. 2023;42:100771. [Google Scholar]
  22. Bazan NG, Gordon WC, Rodriguez de Turco EB. Docosahexaenoic acid uptake and metabolism in photoreceptors: retinal conservation by an efficient retinal pigment epithelial cell-mediated recycling process. Adv Exp Med Biol. 1992;318:295–306. doi: 10.1007/978-1-4615-3426-6_26. [DOI] [PubMed] [Google Scholar]
  23. Belmouhand M, Rothenbuehler SP, Dabbah S, Bjerager J, Sander B, Hjelmborg JB, Dalgård C, Jensen R, Larsen M. Small hard drusen and associated factors in early seniority. PLoS One. 2022;17:e0279279. doi: 10.1371/journal.pone.0279279. [DOI] [PMC free article] [PubMed] [Google Scholar]
  24. Blank T, Goldmann T, Koch M, Amann L, Schön C, Bonin M, Pang S, Prinz M, Burnet M, Wagner JE, Biel M, Michalakis S. Early microglia activation precedes photoreceptor degeneration in a mouse model of CNGB1-linked retinitis pigmentosa. Front Immunol. 2018;8:1930. doi: 10.3389/fimmu.2017.01930. [DOI] [PMC free article] [PubMed] [Google Scholar]
  25. Bloch SB, Lund-Andersen H, Sander B, Larsen M. Subfoveal fibrosis in eyes with neovascular age-related macular degeneration treated with intravitreal ranibizumab. Am J Ophthalmol. 2013;156:116–124. doi: 10.1016/j.ajo.2013.02.012. [DOI] [PubMed] [Google Scholar]
  26. Del Bo’ C, Bernardi S, Marino M, Porrini M, Tucci M, Guglielmetti S, Cherubini A, Carrieri B, Kirkup B, Kroon P, Zamora-Ros R, Liberona NH, Andres-Lacueva C, Riso P. Systematic review on polyphenol intake and health outcomes: is there sufficient evidence to define a health-promoting polyphenol-rich dietary pattern? Nutrients 11:1355. 2019 doi: 10.3390/nu11061355. [DOI] [PMC free article] [PubMed] [Google Scholar]
  27. Böhm EW, Buonfiglio F, Voigt AM, Bachmann P, Safi T, Pfeiffer N, Gericke A. Oxidative stress in the eye and its role in the pathophysiology of ocular diseases. Redox Biol. 2023;68:102967. doi: 10.1016/j.redox.2023.102967. [DOI] [PMC free article] [PubMed] [Google Scholar]
  28. Bovi Dos Santos G, de Lima-Vasconcellos TH, Móvio MI, Birbrair A, Del Debbio CB, Kihara AH. New perspectives in stem cell transplantation and associated therapies to treat retinal diseases: from gene editing to 3D bioprinting. Stem Cell Rev Rep. 2024;20:722–737. doi: 10.1007/s12015-024-10689-4. [DOI] [PubMed] [Google Scholar]
  29. Brandli A, Khong FL, Kong RCK, Kelly DJ, Fletcher EL. Transcriptomic analysis of choroidal neovascularization reveals dysregulation of immune and fibrosis pathways that are attenuated by a novel anti-fibrotic treatment. Sci Rep. 2022;12:859. doi: 10.1038/s41598-022-04845-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  30. Bravo-Gil N, González-Del Pozo M, Martín-Sánchez M, Méndez-Vidal C, Rodríguez-de la Rúa E, Borrego S, Antiñolo G. Unravelling the genetic basis of simplex Retinitis Pigmentosa cases. Sci Rep. 2017;7:41937. doi: 10.1038/srep41937. [DOI] [PMC free article] [PubMed] [Google Scholar]
  31. Brickman AM, Khan UA, Provenzano FA, Yeung LK, Suzuki W, Schroeter H, Wall M, Sloan RP, Small SA. Enhancing dentate gyrus function with dietary flavanols improves cognition in older adults. Nat Neurosci. 2014;17:1798–1803. doi: 10.1038/nn.3850. [DOI] [PMC free article] [PubMed] [Google Scholar]
  32. Broadgate S, Yu J, Downes SM, Halford S. Unravelling the genetics of inherited retinal dystrophies: Past, present and future. Prog Retin Eye Res. 2017;59:53–96. doi: 10.1016/j.preteyeres.2017.03.003. [DOI] [PubMed] [Google Scholar]
  33. Brunet AA, Harvey AR, Carvalho LS. Primary and secondary cone cell death mechanisms in inherited retinal diseases and potential treatment options. Int J Mol Sci. 2022;23:726. doi: 10.3390/ijms23020726. [DOI] [PMC free article] [PubMed] [Google Scholar]
  34. Bunkar N, Shandilya R, Bhargava A, Samarth RM, Tiwari R, Mishra DK, Srivastava RK, Sharma RS, Lohiya NK, Mishra PK. Front Biosci (Landmark Ed) 2019;24:1097–1157. doi: 10.2741/4771. [DOI] [PubMed] [Google Scholar]
  35. Buschini E, Fea AM, Lavia CA, Nassisi M, Pignata G, Zola M, Grignolo FM. Recent developments in the management of dry age-related macular degeneration. Clin Ophthalmol. 2015;9:563–574. doi: 10.2147/OPTH.S59724. [DOI] [PMC free article] [PubMed] [Google Scholar]
  36. Butler MR, Ma H, Yang F, Belcher J, Le YZ, Mikoshiba K, Biel M, Michalakis S, Iuso A, Križaj D, Ding XQ. Endoplasmic reticulum (ER) Ca2+-channel activity contributes to ER stress and cone death in cyclic nucleotide-gated channel deficiency. J Biol Chem. 2017;292:11189–11205. doi: 10.1074/jbc.M117.782326. [DOI] [PMC free article] [PubMed] [Google Scholar]
  37. Cai X, Conley SM, Naash MI. RPE65: role in the visual cycle, human retinal disease, and gene therapy. Ophthalmic Genet. 2009;30:57–62. doi: 10.1080/13816810802626399. [DOI] [PMC free article] [PubMed] [Google Scholar]
  38. Calderaro A, Patanè GT, Tellone E, Barreca D, Ficarra S, Misiti F, Laganà G. The neuroprotective potentiality of flavonoids on Alzheimer’s disease. Int J Mol Sci. 2022;23:14835. doi: 10.3390/ijms232314835. [DOI] [PMC free article] [PubMed] [Google Scholar]
  39. Campochiaro PA, Mir TA. The mechanism of cone cell death in Retinitis Pigmentosa. Prog Retin Eye Res. 2018;62:24–37. doi: 10.1016/j.preteyeres.2017.08.004. [DOI] [PubMed] [Google Scholar]
  40. Cantó A, Martínez-González J, Almansa I, López-Pedrajas R, Hernández-Rabaza V, Olivar T, Miranda M. Time-course changes in oxidative stress and inflammation in the retinas of rds mice: a retinitis pigmentosa model. Antioxidants (Basel) 2022;11:1950. doi: 10.3390/antiox11101950. [DOI] [PMC free article] [PubMed] [Google Scholar]
  41. Carnauba RA, Sarti FM, Hassimotto NMA, Lajolo FM. Estimated polyphenol intake and major food sources of the Brazilian population: changes between 2008-2009 and 2017-2018. Br J Nutr. 2023;130:147–154. doi: 10.1017/S0007114522003221. [DOI] [PubMed] [Google Scholar]
  42. Carullo G, Federico S, Relitti N, Gemma S, Butini S, Campiani G. Retinitis pigmentosa and retinal degenerations: deciphering pathways and targets for drug discovery and development. ACS Chem Neurosci. 2020;11:2173–2191. doi: 10.1021/acschemneuro.0c00358. [DOI] [PubMed] [Google Scholar]
  43. Chen L, Perera ND, Karoukis AJ, Feathers KL, Ali RR, Thompson DA, Fahim AT. Oxidative stress differentially impacts apical and basolateral secretion of angiogenic factors from human iPSC-derived retinal pigment epithelium cells. Sci Rep. 2022;12:12694. doi: 10.1038/s41598-022-16701-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  44. Chen RP, Chavda VP, Patel AB, Chen ZS. Phytochemical delivery through transferosome (Phytosome): an advanced transdermal drug delivery for complementary medicines. Front Pharmacol. 2022;13:850862. doi: 10.3389/fphar.2022.850862. [DOI] [PMC free article] [PubMed] [Google Scholar]
  45. Chen W, Liu P, Liu D, Huang H, Feng X, Fang F, Li L, Wu J, Liu L, Solow-Cordero DE, Hu Y. Maprotiline restores ER homeostasis and rescues neurodegeneration via Histamine Receptor H1 inhibition in retinal ganglion cells. Nat Commun. 2022;13:6796. doi: 10.1038/s41467-022-34682-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  46. Chen X, Shi C, He M, Xiong S, Xia X. Endoplasmic reticulum stress: molecular mechanism and therapeutic targets. Signal Transduct Target Ther. 2023;8:352. doi: 10.1038/s41392-023-01570-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
  47. Chiang JP, Trzupek K. The current status of molecular diagnosis of inherited retinal dystrophies. Curr Opin Ophthalmol. 2015;26:346–351. doi: 10.1097/ICU.0000000000000185. [DOI] [PubMed] [Google Scholar]
  48. Chipaux M, van der Laan KJ, Hemelaar SR, Hasani M, Zheng T, Schirhagl R. Nanodiamonds and their applications in cells. Small. 2018;14:e1704263. doi: 10.1002/smll.201704263. [DOI] [PubMed] [Google Scholar]
  49. Choo PP, Woi PJ, Bastion MC, Omar R, Mustapha M, Md Din N. Review of evidence for the usage of antioxidants for eye aging. Biomed Res Int. 2022;2022:5810373. doi: 10.1155/2022/5810373. [DOI] [PMC free article] [PubMed] [Google Scholar]
  50. Chung DC, et al. The natural history of inherited retinal dystrophy due to biallelic mutations in the RPE65 gene. Am J Ophthalmol. 2019;199:58–70. doi: 10.1016/j.ajo.2018.09.024. [DOI] [PMC free article] [PubMed] [Google Scholar]
  51. Cianciosi D, Forbes-Hernández TY, Regolo L, Alvarez-Suarez JM, Navarro-Hortal MD, Xiao J, Quiles JL, Battino M, Giampieri F. The reciprocal interaction between polyphenols and other dietary compounds: Impact on bioavailability, antioxidant capacity and other physico-chemical and nutritional parameters. Food Chem. 2022;375:131904. doi: 10.1016/j.foodchem.2021.131904. [DOI] [PubMed] [Google Scholar]
  52. Comitato A, Schiroli D, Montanari M, Marigo V. Calpain activation is the major cause of cell death in photoreceptors expressing a rhodopsin misfolding mutation. Mol Neurobiol. 2020;57:589–599. doi: 10.1007/s12035-019-01723-5. [DOI] [PubMed] [Google Scholar]
  53. Cortinhal T, Santos C, Vaz-Pereira S, Marta A, Duarte L, Miranda V, Costa J, Sousa AB, Peter VG, Kaminska K, Rivolta C, Carvalho AL, Saraiva J, Soares CA, Silva R, Murta J, Santos LC, Marques JP. Genetic profile of syndromic retinitis pigmentosa in Portugal. Graefes Arch Clin Exp Ophthalmol. 2024;262:1883–1897. doi: 10.1007/s00417-023-06360-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  54. Costa KA, Salles MV, Whitebirch C, Chiang J, Sallum JMF. Gene panel sequencing in Brazilian patients with retinitis pigmentosa. Int J Retina Vitreous. 2017;3:33. doi: 10.1186/s40942-017-0087-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  55. Daiger SP, Sullivan LS, Bowne SJ. Genes and mutations causing retinitis pigmentosa. Clin Genet. 2013;84:132–141. doi: 10.1111/cge.12203. [DOI] [PMC free article] [PubMed] [Google Scholar]
  56. Davis MD, Gangnon RE, Lee LY, Hubbard LD, Klein BE, Klein R, Ferris FL, Bressler SB, Milton RC; Age-Related Eye Disease Study Group The Age-Related Eye Disease Study severity scale for age-related macular degeneration: AREDS Report No. 17. Arch Ophthalmol. 2005;123:1484–1498. doi: 10.1001/archopht.123.11.1484. [DOI] [PMC free article] [PubMed] [Google Scholar]
  57. DeAngelis MM, Owen LA, Morrison MA, Morgan DJ, Li M, Shakoor A, Vitale A, Iyengar S, Stambolian D, Kim IK, Farrer LA. Genetics of age-related macular degeneration (AMD) Hum Mol Genet. 2017;26:R45–50. doi: 10.1093/hmg/ddx228. [DOI] [PMC free article] [PubMed] [Google Scholar]
  58. Detaram HD, Liew G, Lewis JR, Bondonno NP, Bondonno CP, Van Vu K, Burlutsky G, Hodgson JM, Mitchell P, Gopinath B. Dietary flavonoids are associated with longitudinal treatment outcomes in neovascular age-related macular degeneration. Eur J Nutr. 2021;60:4243–4250. doi: 10.1007/s00394-021-02582-4. [DOI] [PubMed] [Google Scholar]
  59. Dias MC, Pinto DCGA, Silva AMS. Plant flavonoids: chemical characteristics and biological activity. Molecules. 2021;26:5377. doi: 10.3390/molecules26175377. [DOI] [PMC free article] [PubMed] [Google Scholar]
  60. Díaz-Lezama N, Kajtna J, Wu J, Ayten M, Koch SF. Microglial and macroglial dynamics in a model of retinitis pigmentosa. Vision Res. 2023 Sep. 2023;210:108268. doi: 10.1016/j.visres.2023.108268. [DOI] [PubMed] [Google Scholar]
  61. Dong HJ, Brain K, Olsson M, Dragioti E, Gerdle B, Ghafouri B. Eating habits and the desire to eat healthier among patients with chronic pain: a registry-based study. Sci Rep. 2024;14:4705. doi: 10.1038/s41598-024-55449-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  62. Doyle SL, Campbell M, Ozaki E, Salomon RG, Mori A, Kenna PF, Farrar GJ, Kiang AS, Humphries MM, Lavelle EC, O’Neill LA, Hollyfield JG, Humphries P. NLRP3 has a protective role in age-related macular degeneration through the induction of IL-18 by drusen components. Nat Med. 2012;18:791–798. doi: 10.1038/nm.2717. [DOI] [PMC free article] [PubMed] [Google Scholar]
  63. Dryja TP, McGee TL, Reichel E, Hahn LB, Cowley GS, Yandell DW, Sandberg MA, Berson EL. A point mutation of the rhodopsin gene in one form of retinitis pigmentosa. Nature. 1990;343:364–366. doi: 10.1038/343364a0. [DOI] [PubMed] [Google Scholar]
  64. Edwards MM, McLeod DS, Shen M, Grebe R, Sunness JS, Bhutto IA, McDonnell E, Pado AM, Gregori G, Rosenfeld PJ, Lutty GA. Clinicopathologic findings in three siblings with geographic atrophy. Invest Ophthalmol Vis Sci. 2023;64:2. doi: 10.1167/iovs.64.3.2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  65. El-Emam MA, Sheta E, El-Abhar HS, Abdallah DM, El Kerdawy AM, Eldehna WM, Gowayed MA. Morin suppresses mTORc1/IRE-1α/JNK and IP3R-VDAC-1 pathways: Crucial mechanisms in apoptosis and mitophagy inhibition in experimental Huntington’s disease, supported by in silico molecular docking simulations. Life Sci. 2024;338:122362. doi: 10.1016/j.lfs.2023.122362. [DOI] [PubMed] [Google Scholar]
  66. Emoto Y, Yoshizawa K, Kinoshita Y, Yuri T, Yuki M, Sayama K, Shikata N, Tsubura A. Green tea extract suppresses N-methyl-N-nitrosourea-induced photoreceptor apoptosis in Sprague-Dawley rats. Graefes Arch Clin Exp Ophthalmol. 2014;252:1377–1384. doi: 10.1007/s00417-014-2702-7. [DOI] [PubMed] [Google Scholar]
  67. Emoto Y, Yoshizawa K, Kinoshita Y, Yuki M, Yuri T, Tsubura A. Green tea extract attenuates MNU-induced photoreceptor cell apoptosis via suppression of heme oxygenase-1. J Toxicol Pathol. 2015;29:61–65. doi: 10.1293/tox.2015-0052. [DOI] [PMC free article] [PubMed] [Google Scholar]
  68. Escobar-Cévoli R, Castro-Espín C, Béraud V, Buckland G, Zamora-Ros R, Béraud GB V. An overview of global flavonoid intake and its food sources. Flavonoids Biosynthesis Human Health [Preprint] 2017 doi: 10.5772/67655. [Google Scholar]
  69. Fan Q, et al. Contribution of common and rare variants to Asian neovascular age-related macular degeneration subtypes. Nat Commun. 2023;14:5574. doi: 10.1038/s41467-023-41256-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  70. Fanaro GB, Marques MR, Calaza KDC, Brito R, Pessoni AM, Mendonça HR, Lemos DEA, 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 (Basel) 2023;12:1237. doi: 10.3390/antiox12061237. [DOI] [PMC free article] [PubMed] [Google Scholar]
  71. Farnoodian M, Bose D, Barone F, Nelson LM, Boyle M, Jun B, Do K, Gordon W, Guerin MK, Perera R, Ji JX, Cogliati T, Sharma R, Brooks BP, Bazan NG, Bharti K. Retina and RPE lipid profile changes linked with ABCA4 associated Stargardt’s maculopathy. Pharmacol Ther. 2023;249:108482. doi: 10.1016/j.pharmthera.2023.108482. [DOI] [PMC free article] [PubMed] [Google Scholar]
  72. Fernández-Sánchez L, Lax P, Campello L, Pinilla I, Cuenca N. Astrocytes and Müller cell alterations during retinal degeneration in a transgenic rat model of retinitis pigmentosa. Front Cell Neurosci. 2015;9:484. doi: 10.3389/fncel.2015.00484. [DOI] [PMC free article] [PubMed] [Google Scholar]
  73. Ferreira C, Vieira P, Sá H, Malva J, Castelo-Branco M, Reis F, Viana S. Polyphenols: immunonutrients tipping the balance of immunometabolism in chronic diseases. Front Immunol. 2024;15:1360065. doi: 10.3389/fimmu.2024.1360065. [DOI] [PMC free article] [PubMed] [Google Scholar]
  74. Ferris FL, 3rd, Wilkinson CP, Bird A, Chakravarthy U, Chew E, Csaky K, Sadda SR; Beckman Initiative for Macular Research Classification Committee Clinical classification of age-related macular degeneration. Ophthalmology. 2013;120:844–851. doi: 10.1016/j.ophtha.2012.10.036. [DOI] [PMC free article] [PubMed] [Google Scholar]
  75. Finger RP, Daien V, Eldem BM, Talks JS, Korobelnik JF, Mitchell P, Sakamoto T, Wong TY, Pantiri K, Carrasco J. Anti-vascular endothelial growth factor in neovascular age-related macular degeneration - a systematic review of the impact of anti-VEGF on patient outcomes and healthcare systems. BMC Ophthalmol. 2020;20:294. doi: 10.1186/s12886-020-01554-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  76. Fleckenstein M, Keenan TDL, Guymer RH, Chakravarthy U, Schmitz-Valckenberg S, Klaver CC, Wong WT, Chew EY. Age-related macular degeneration. Nat Rev Dis Primers. 2021;7:31. doi: 10.1038/s41572-021-00265-2. [DOI] [PubMed] [Google Scholar]
  77. Gallenga CE, Lonardi M, Pacetti S, Violanti SS, Tassinari P, Di Virgilio F, Tognon M, Perri P. Molecular mechanisms related to oxidative stress in retinitis pigmentosa. Antioxidants (Basel) 2021;10:848. doi: 10.3390/antiox10060848. [DOI] [PMC free article] [PubMed] [Google Scholar]
  78. Gao Y, Eskici G, Ramachandran S, Poitevin F, Seven AB, Panova O, Skiniotis G, Cerione RA. Structure of the visual signaling complex between transducin and phosphodiesterase 6. Mol Cell. 2020;80:237–245. doi: 10.1016/j.molcel.2020.09.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  79. Gass JD, Agarwal A, Lavina AM, Tawansy KA. Focal inner retinal hemorrhages in patients with drusen: an early sign of occult choroidal neovascularization and chorioretinal anastomosis. Retina. 2003;23:741–751. doi: 10.1097/00006982-200312000-00001. [DOI] [PubMed] [Google Scholar]
  80. Ghezzi M, Pescina S, Padula C, Santi P, Del Favero E, Cantù L, Nicoli S. Polymeric micelles in drug delivery: An insight of the techniques for their characterization and assessment in biorelevant conditions. J Control Release. 2021;332:312–336. doi: 10.1016/j.jconrel.2021.02.031. [DOI] [PubMed] [Google Scholar]
  81. Gismondi A, Reina G, Orlanducci S, Mizzoni F, Gay S, Terranova ML, Canini A. Nanodiamonds coupled with plant bioactive metabolites: A nanotech approach for cancer therapy. Biomaterials. 2015;38:22–35. doi: 10.1016/j.biomaterials.2014.10.057. [DOI] [PubMed] [Google Scholar]
  82. Gómez-Pinilla F. Brain foods: the effects of nutrients on brain function. Nat Rev Neurosci. 2008;9:568–578. doi: 10.1038/nrn2421. [DOI] [PMC free article] [PubMed] [Google Scholar]
  83. Gopinath B, Liew G, Kifley A, Flood VM, Joachim N, Lewis JR, Hodgson JM, Mitchell P. Dietary flavonoids and the prevalence and 15-y incidence of age-related macular degeneration. Am J Clin Nutr. 2018;108:381–387. doi: 10.1093/ajcn/nqy114. [DOI] [PubMed] [Google Scholar]
  84. Gorantla S, Wadhwa G, Jain S, Sankar S, Nuwal K, Mahmood A, Dubey SK, Taliyan R, Kesharwani P, Singhvi G. Recent advances in nanocarriers for nutrient delivery. Drug Deliv Transl Res. 2021;12:2359–2384. doi: 10.1007/s13346-021-01097-z. [DOI] [PubMed] [Google Scholar]
  85. Grimm C, Remé CE. Light damage as a model of retinal degeneration. Methods Mol Biol. 2013;935:87–97. doi: 10.1007/978-1-62703-080-9_6. [DOI] [PubMed] [Google Scholar]
  86. Grosso G, Stepaniak U, Topor-Mądry R, Szafraniec K, Pająk A. Estimated dietary intake and major food sources of polyphenols in the Polish arm of the HAPIEE study. Nutrition. 2014;30:1398–1403. doi: 10.1016/j.nut.2014.04.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  87. Ha JH, Shil PK, Zhu P, Gu L, Li Q, Chung S. Ocular inflammation and endoplasmic reticulum stress are attenuated by supplementation with grape polyphenols in human retinal pigmented epithelium cells and in C57BL/6 mice. J Nutr. 2014;144:799–806. doi: 10.3945/jn.113.186957. [DOI] [PMC free article] [PubMed] [Google Scholar]
  88. Haidara K, Marion M, Gascon-Barré M, Denizeau F, Averill-Bates DA. Implication of caspases and subcellular compartments in tert-butylhydroperoxide induced apoptosis. Toxicol Appl Pharmacol. 2008;229:65–76. doi: 10.1016/j.taap.2008.01.010. [DOI] [PubMed] [Google Scholar]
  89. Hanna J, David LA, Touahri Y, Fleming T, Screaton RA, Schuurmans C. Beyond genetics: The role of metabolism in photoreceptor survival, development and repair. Front Cell Dev Biol. 2022;10:887764. doi: 10.3389/fcell.2022.887764. [DOI] [PMC free article] [PubMed] [Google Scholar]
  90. Hartmann KI, Gomez ML, Bartsch DU, Schuster AK, Freeman WR. Effect of change in drusen evolution on photoreceptor inner segment/outer segment junction. Retina. 2012;32:1492–1499. doi: 10.1097/IAE.0b013e318242b949. [DOI] [PMC free article] [PubMed] [Google Scholar]
  91. He M, Wu T, Zhang L, Ye W, Ma J, Zhao C, Liu J, Zhou J. Correlation between neutrophil-to-lymphocyte ratio and clinical manifestations and complications of retinitis pigmentosa. Acta Ophthalmol. 2022;100:e278–287. doi: 10.1111/aos.14880. [DOI] [PubMed] [Google Scholar]
  92. He X, Sun D, Chen S, Xu H. Activation of liver X receptor delayed the retinal degeneration of rd1 mice through modulation of the immunological function of glia. Oncotarget. 2017;8:32068–32082. doi: 10.18632/oncotarget.16643. [DOI] [PMC free article] [PubMed] [Google Scholar]
  93. Heier JS, Lad EM, Holz FG, Rosenfeld PJ, Guymer RH, Boyer D, Grossi F, Baumal CR, Korobelnik JF, Slakter JS, Waheed NK, Metlapally R, Pearce I, Steinle N, Francone AA, Hu A, Lally DR, Deschatelets P, Francois C, Bliss C, Staurenghi G, Monés J, Singh RP, Ribeiro R, Wykoff CC; OAKS and DERBY study investigators Pegcetacoplan for the treatment of geographic atrophy secondary to age-related macular degeneration (OAKS and DERBY): two multicentre, randomised, double-masked, sham-controlled, phase 3 trials. Lancet. 2023;402:1434–1448. doi: 10.1016/S0140-6736(23)01520-9. [DOI] [PubMed] [Google Scholar]
  94. Hejazi J, Ghanavati M, Hejazi E, Poustchi H, Sepanlou SG, Khoshnia M, Gharavi A, Sohrabpour AA, Sotoudeh M, Dawsey SM, Boffetta P, Abnet CC, Kamangar F, Etemadi A, Pourshams A, FazeltabarMalekshah A, Brennan P, Malekzadeh R, Hekmatdoost A. Habitual dietary intake of flavonoids and all-cause and cause-specific mortality: Golestan cohort study. Nutr J. 2020;19:108. doi: 10.1186/s12937-020-00627-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  95. Hepsomali P, Groeger JA. Diet and general cognitive ability in the UK Biobank dataset. Sci Rep. 2021;11:11786. doi: 10.1038/s41598-021-91259-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  96. Herrera-Hernández MG, Ramon E, Lupala CS, Tena-Campos M, Pérez JJ, Garriga P. Flavonoid allosteric modulation of mutated visual rhodopsin associated with retinitis pigmentosa. Sci Rep. 2017;7:11167. doi: 10.1038/s41598-017-11391-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  97. Hidalgo M, Sánchez-Moreno C, de Pascual-Teresa S. Flavonoid–flavonoid interaction and its effect on their antioxidant activity. Food Chem. 2010;121:691–696. [Google Scholar]
  98. Hippert C, Graca AB, Barber AC, West EL, Smith AJ, Ali RR, Pearson RA. Müller glia activation in response to inherited retinal degeneration is highly varied and disease-specific. PLoS One. 2015;10:e0120415. doi: 10.1371/journal.pone.0120415. [DOI] [PMC free article] [PubMed] [Google Scholar]
  99. Hodge G, Hodge S, Liu H, Nguyen P, Holmes-Liew CL, Holmes M. BOS is associated with decreased SIRT1 in peripheral blood proinflammatory T, NK, and NKT-like lymphocytes. Transplantation. 2019;103:2255–2263. doi: 10.1097/TP.0000000000002817. [DOI] [PubMed] [Google Scholar]
  100. Holz FG, et al. Imaging protocols in clinical studies in advanced age-related macular degeneration: recommendations from classification of atrophy consensus meetings. Ophthalmology. 2017;124:464–478. doi: 10.1016/j.ophtha.2016.12.002. [DOI] [PubMed] [Google Scholar]
  101. de Hoz R, Rojas B, Ramírez AI, Salazar JJ, Gallego BI, Triviño A, Ramírez JM. Retinal macroglial responses in health and disease. Biomed Res Int. 2016;2016:2954721. doi: 10.1155/2016/2954721. [DOI] [PMC free article] [PubMed] [Google Scholar]
  102. Hu Y, Park KK, Yang L, Wei X, Yang Q, Cho KS, Thielen P, Lee AH, Cartoni R, Glimcher LH, Chen DF, He Z. Differential effects of unfolded protein response pathways on axon injury-induced death of retinal ganglion cells. Neuron. 2012;73:445–452. doi: 10.1016/j.neuron.2011.11.026. [DOI] [PMC free article] [PubMed] [Google Scholar]
  103. Huang Q, Braffett BH, Simmens SJ, Young HA, Ogden CL. Dietary polyphenol intake in US adults and 10-year trends: 2007–2016. J Acad Nutr Diet. 2020;120:1821–1833. doi: 10.1016/j.jand.2020.06.016. [DOI] [PubMed] [Google Scholar]
  104. Huang Y, Wong CM, Lau CW, Yao X, Tsang SY, Su YL, Chen ZY. Inhibition of nitric oxide/cyclic GMP-mediated relaxation by purified flavonoids, baicalin and baicalein, in rat aortic rings. Biochem Pharmacol. 2004;67:787–794. doi: 10.1016/j.bcp.2003.10.002. [DOI] [PubMed] [Google Scholar]
  105. Huang Y, Chen Y, Shaw AM, Goldfine H, Tian J, Cai J. Enhancing TFEB-mediated cellular degradation pathways by the mTORC1 inhibitor quercetin. Oxid Med Cell Longev. 2018;2018:5073420. doi: 10.1155/2018/5073420. [DOI] [PMC free article] [PubMed] [Google Scholar]
  106. Hussain Y, Luqman S, Meena A. Research progress in flavonoids as potential anticancer drug including synergy with other approaches. Curr Top Med Chem. 2020;20:1791–1809. doi: 10.2174/1568026620666200502005411. [DOI] [PubMed] [Google Scholar]
  107. Hyon SH, Kim DH, Cui W, Matsumura K, Kim JY, Tsutsumi S. Preservation of rat aortic tissue transplant with green tea polyphenols. Cell Transplant. 2006;15:881–883. doi: 10.3727/000000006783981422. [DOI] [PubMed] [Google Scholar]
  108. Ibbett P, Goverdhan SV, Pipi E, Chouhan JK, Keeling E, Angus EM, Scott JA, Gatherer M, Page A, Teeling JL, Lotery AJ, Arjuna Ratnayaka J. A lasered mouse model of retinal degeneration displays progressive outer retinal pathology providing insights into early geographic atrophy. Sci Rep. 2019;9:7475. doi: 10.1038/s41598-019-43906-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  109. Iker Etchegaray J, et al. Phagocytosis in the retina promotes local insulin production in the eye. Nat Metab. 2023;5:207–218. doi: 10.1038/s42255-022-00728-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  110. Inana G, Murat C, An W, Yao X, Harris IR, Cao J. RPE phagocytic function declines in age-related macular degeneration and is rescued by human umbilical tissue derived cells. J Transl Med. 2018;16:63. doi: 10.1186/s12967-018-1434-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  111. Iraha S, Tu HY, Yamasaki S, Kagawa T, Goto M, Takahashi R, Watanabe T, Sugita S, Yonemura S, Sunagawa GA, Matsuyama T, Fujii M, Kuwahara A, Kishino A, Koide N, Eiraku M, Tanihara H, Takahashi M, Mandai M. Establishment of immunodeficient retinal degeneration model mice and functional maturation of human ESC-derived retinal sheets after transplantation. Stem Cell Reports. 2018;10:1059–1074. doi: 10.1016/j.stemcr.2018.01.032. [DOI] [PMC free article] [PubMed] [Google Scholar]
  112. Ivanescu AA, Fernández-Robredo P, Heras-Mulero H, Sádaba-Echarri LM, García-García L, Fernández-García V, Moreno-Orduna M, Redondo-Exposito A, Recalde S, García-Layana A. Modifying choroidal neovascularization development with a nutritional supplement in mice. Nutrients. 2015;7:5423–5442. doi: 10.3390/nu7075229. [DOI] [PMC free article] [PubMed] [Google Scholar]
  113. Jang S, Johnson RW. Can consuming flavonoids restore old microglia to their youthful state? Nutr Rev. 2010;68:719–728. doi: 10.1111/j.1753-4887.2010.00336.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  114. Jauregui R, Takahashi VKL, Park KS, Cui X, Takiuti JT, Lima de Carvalho JR, Jr, Tsang SH. Multimodal structural disease progression of retinitis pigmentosa according to mode of inheritance. Sci Rep. 2019;9:10712. doi: 10.1038/s41598-019-47251-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  115. Jauregui R, Park KS, Tsang SH. Two-year progression analysis of RPE65 autosomal dominant retinitis pigmentosa. Ophthalmic Genet. 2018;39:544–549. doi: 10.1080/13816810.2018.1484929. [DOI] [PMC free article] [PubMed] [Google Scholar]
  116. Jin M, Li S, Moghrabi WN, Sun H, Travis GH. Rpe65 is the retinoid isomerase in bovine retinal pigment epithelium. Cell. 2005;122:449–459. doi: 10.1016/j.cell.2005.06.042. [DOI] [PMC free article] [PubMed] [Google Scholar]
  117. Jing H, Nie M, Wang X, Zhang Z, Xu Y, Zhang G, Li D, Dai Z. Lutein combined with EGCG improved retinitis pigmentosa against N-methyl-N nitrosourea-induced. Food Funct. 2023;14:9554–9566. doi: 10.1039/d3fo02716c. [DOI] [PubMed] [Google Scholar]
  118. Jones BW, Pfeiffer RL, Ferrell WD, Watt CB, Marmor M, Marc RE. Retinal remodeling in human retinitis pigmentosa. Exp Eye Res. 2016;150:149–165. doi: 10.1016/j.exer.2016.03.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  119. Jurga AM, Paleczna M, Kuter KZ. Overview of general and discriminating markers of differential microglia phenotypes. Front Cell Neurosci. 2020;14:198. doi: 10.3389/fncel.2020.00198. [DOI] [PMC free article] [PubMed] [Google Scholar]
  120. Kajtna J, Tsang SH, Koch SF. Late-stage rescue of visually guided behavior in the context of a significantly remodeled retinitis pigmentosa mouse model. Cell Mol Life Sci. 2022;79:148. doi: 10.1007/s00018-022-04161-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  121. Kaleem M, Ahmad A. Flavonoids as nutraceuticals. Ther Probiotic Unconv Food. 2018 doi:10.1016/B978-0-12-814625-5.00008-X. [Google Scholar]
  122. Kanan Y, Hackett SF, Hsueh HT, Khan M, Ensign LM, Campochiaro PA. Reduced inspired oxygen decreases retinal superoxide radicals and promotes cone function and survival in a model of retinitis pigmentosa. Free Radic Biol Med. 2023;198:118–122. doi: 10.1016/j.freeradbiomed.2023.01.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  123. Kannan R, Hinton DR. Sodium iodate induced retinal degeneration: new insights from an old model. Neural Regen Res. 2014;9:2044–2045. doi: 10.4103/1673-5374.147927. [DOI] [PMC free article] [PubMed] [Google Scholar]
  124. Kapolou A, Karantonis HC, Rigopoulos N, Koutelidakis AE. Association of mean daily polyphenols intake with Mediterranean diet adherence and anthropometric indices in healthy Greek adults: A retrospective study. Appl Sci. 2021;11:4664. [Google Scholar]
  125. Kaushal N, Singh M, Singh Sangwan R. Flavonoids: Food associations, therapeutic mechanisms, metabolism and nanoformulations. Food Res Int. 2022;157:111442. doi: 10.1016/j.foodres.2022.111442. [DOI] [PubMed] [Google Scholar]
  126. Kim EK, Kim H, Kwon O, Chang N. Associations between fruits, vegetables, vitamin A, β-carotene and flavonol dietary intake, and age-related macular degeneration in elderly women in Korea: the Fifth Korea National Health and Nutrition Examination Survey. Eur J Clin Nutr. 2017;72:161–167. doi: 10.1038/ejcn.2017.152. [DOI] [PubMed] [Google Scholar]
  127. Klein R, Chou CF, Klein BE, Zhang X, Meuer SM, Saaddine JB. Prevalence of age-related macular degeneration in the US population. Arch Ophthalmol. 2011;129:75–80. doi: 10.1001/archophthalmol.2010.318. [DOI] [PubMed] [Google Scholar]
  128. Kocherlakota S, Das Y, Swinkels D, Vanmunster M, Callens M, Vinckier S, Vaz FM, Sinha D, Van Veldhoven PP, Fransen M, Baes M. The murine retinal pigment epithelium requires peroxisomal β-oxidation to maintain lysosomal function and prevent dedifferentiation. Proc Natl Acad Sci U S A. 2023;120:e2301733120. doi: 10.1073/pnas.2301733120. [DOI] [PMC free article] [PubMed] [Google Scholar]
  129. Koirala N, Thuan NH, Ghimire GP, Thang DV, Sohng JK. Methylation of flavonoids: Chemical structures, bioactivities, progress and perspectives for biotechnological production. Enzyme Microb Technol. 2016;86:103–116. doi: 10.1016/j.enzmictec.2016.02.003. [DOI] [PubMed] [Google Scholar]
  130. Kotha RR, Luthria DL. Curcumin: Biological, pharmaceutical, nutraceutical, and analytical aspects. Molecules. 2019;24:2930. doi: 10.3390/molecules24162930. [DOI] [PMC free article] [PubMed] [Google Scholar]
  131. Koyanagi Y, et al. Genetic characteristics of retinitis pigmentosa in 1204 Japanese patients. J Med Genet. 2019;56:662–670. doi: 10.1136/jmedgenet-2018-105691. [DOI] [PubMed] [Google Scholar]
  132. Kubota S, Kurihara T, Ebinuma M, Kubota M, Yuki K, Sasaki M, Noda K, Ozawa Y, Oike Y, Ishida S, Tsubota K. Resveratrol prevents light-induced retinal degeneration via suppressing activator protein-1 activation. Am J Pathol. 2010;177:1725–31. doi: 10.2353/ajpath.2010.100098. [DOI] [PMC free article] [PubMed] [Google Scholar]
  133. Kuehlewein L, Zobor D, Stingl K, Kempf M, Nasser F, Bernd A, Biskup S, Cremers FPM, Khan MI, Mazzola P, Schäferhoff K, Heinrich T, Haack TB, Wissinger B, Zrenner E, Weisschuh N, Kohl S. Clinical phenotype of PDE6B-associated retinitis pigmentosa. Int J Mol Sci. 2021;22:2374. doi: 10.3390/ijms22052374. [DOI] [PMC free article] [PubMed] [Google Scholar]
  134. Lambert NG, ElShelmani H, Singh MK, Mansergh FC, Wride MA, Padilla M, Keegan D, Hogg RE, Ambati BK. Risk factors and biomarkers of age-related macular degeneration. Prog Retin Eye Res. 2016;54:64–102. doi: 10.1016/j.preteyeres.2016.04.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  135. Lang M, Harris A, Ciulla TA, Siesky B, Patel P, Belamkar A, Mathew S, Verticchio Vercellin AC. Vascular dysfunction in retinitis pigmentosa. Acta Ophthalmol. 2019;97:660–664. doi: 10.1111/aos.14138. [DOI] [PubMed] [Google Scholar]
  136. Lee SH, Jeong E, Paik SS, Jeon JH, Jung SW, Kim HB, Kim M, Chun MH, Kim IB. Cyanidin-3-glucoside extracted from mulberry fruit can reduce N-methyl-N-nitrosourea-Induced Retinal Degeneration in Rats. Curr Eye Res. 2014;39:79–87. doi: 10.3109/02713683.2013.825275. [DOI] [PubMed] [Google Scholar]
  137. Leena MM, Silvia MG, Vinitha K, Moses JA. Synergistic potential of nutraceuticals: mechanisms and prospects for futuristic medicine. Food Funct. 2020;11:9317–9337. doi: 10.1039/d0fo02041a. [DOI] [PubMed] [Google Scholar]
  138. Leinonen HO, Bull E, Fu Z. Neural and Müller glial adaptation of the retina to photoreceptor degeneration. Neural Regen Res. 2023;18:701–707. doi: 10.4103/1673-5374.354511. [DOI] [PMC free article] [PubMed] [Google Scholar]
  139. Lew DS, Mazzoni F, Finnemann SC. Microglia inhibition delays retinal degeneration due to MerTK phagocytosis receptor deficiency. Front Immunol. 2020;11:1463. doi: 10.3389/fimmu.2020.01463. [DOI] [PMC free article] [PubMed] [Google Scholar]
  140. Lewin AS, Rossmiller B, Mao H. Gene augmentation for adRP mutations in RHO. Cold Spring Harb Perspect Med. 2014;4:a017400. doi: 10.1101/cshperspect.a017400. [DOI] [PMC free article] [PubMed] [Google Scholar]
  141. Li S, Ma H, Yang F, Ding X. cGMP signaling in photoreceptor degeneration. Int J Mol Sci. 2023;24:11200. doi: 10.3390/ijms241311200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  142. Li X, Peng X, Zoulikha M, Boafo GF, Magar KT, Ju Y, He W. Multifunctional nanoparticle-mediated combining therapy for human diseases. Signal Transduct Target Ther. 2024;9:1. doi: 10.1038/s41392-023-01668-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  143. Liu D, Zhang C, Zhang J, Xu GT, Zhang J. Molecular pathogenesis of subretinal fibrosis in neovascular AMD focusing on epithelial-mesenchymal transformation of retinal pigment epithelium. Neurobiol Dis. 2023;185:106250. doi: 10.1016/j.nbd.2023.106250. [DOI] [PubMed] [Google Scholar]
  144. Liu SY, Song JY, Fan B, Wang Y, Pan YR, Che L, Sun YJ, Li GY. Resveratrol protects photoreceptors by blocking caspase- and PARP-dependent cell death pathways. Free Radic Biol Med. 2018;129:569–581. doi: 10.1016/j.freeradbiomed.2018.10.431. [DOI] [PubMed] [Google Scholar]
  145. Liu Y, Tai J, Yu C, Xu D, Xiao D, Pang J. Unlocking therapeutic potential: dual gene therapy for ameliorating the disease phenotypes in a mouse model of RPE65 Leber congenital amaurosis. Front Med (Lausanne) 2024;10:1291795. doi: 10.3389/fmed.2023.1291795. [DOI] [PMC free article] [PubMed] [Google Scholar]
  146. Lopez-Rodriguez R, Lantero E, Blanco-Kelly F, Avila-Fernandez A, Martin Merida I, Del Pozo-Valero M, Perea-Romero I, Zurita O, Jiménez-Rolando B, Swafiri ST, Riveiro-Alvarez R, Trujillo-Tiebas MJ, Carreño Salas E, García-Sandoval B, Corton M, Ayuso C. RPE65-related retinal dystrophy: Mutational and phenotypic spectrum in 45 affected patients. Exp Eye Res. 2021;212:108761. doi: 10.1016/j.exer.2021.108761. [DOI] [PubMed] [Google Scholar]
  147. Ma H, Butler MR, Thapa A, Belcher J, Yang F, Baehr W, Biel M, Michalakis S, Ding XQ. cGMP/protein kinase G signaling suppresses inositol 1,4,5-trisphosphate receptor phosphorylation and promotes endoplasmic reticulum stress in photoreceptors of cyclic nucleotide-gated channel-deficient mice. J Biol Chem. 2015;290:20880–20892. doi: 10.1074/jbc.M115.641159. [DOI] [PMC free article] [PubMed] [Google Scholar]
  148. Ma W, Zhao L, Wong WT. Microglia in the outer retina and their relevance to pathogenesis of age-related macular degeneration. Adv Exp Med Biol. 2012;723:37–42. doi: 10.1007/978-1-4614-0631-0_6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  149. Maneu V, Lax P, De Diego AMG, Cuenca N, García AG. Combined drug triads for synergic neuroprotection in retinal degeneration. Biomed Pharmacother. 2022;149:112911. doi: 10.1016/j.biopha.2022.112911. [DOI] [PubMed] [Google Scholar]
  150. Marsh-Armstrong B, Murrell KS, Valente D, Jonnal RS. Using directional OCT to analyze photoreceptor visibility over AMD-related drusen. Sci Rep. 2022;12:9763. doi: 10.1038/s41598-022-13106-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  151. Martínez-Fernández de la Cámara C, Salom D, Sequedo MD, Hervás D, Marín-Lambíes C, Aller E, Jaijo T, Díaz-Llopis M, Millán JM, Rodrigo R. Altered antioxidant-oxidant status in the aqueous humor and peripheral blood of patients with retinitis pigmentosa. PLoS One. 2013;8:e74223. doi: 10.1371/journal.pone.0074223. [DOI] [PMC free article] [PubMed] [Google Scholar]
  152. Martin-Merida I, Avila-Fernandez A, Del Pozo-Valero M, Blanco-Kelly F, Zurita O, Perez-Carro R, Aguilera-Garcia D, Riveiro-Alvarez R, Arteche A, Trujillo-Tiebas MJ, Tahsin-Swafiri S, Rodriguez-Pinilla E, Lorda-Sanchez I, Garcia-Sandoval B, Corton M, Ayuso C. Genomic landscape of sporadic retinitis pigmentosa: Findings from 877 Spanish cases. Ophthalmology. 2019;126:1181–1188. doi: 10.1016/j.ophtha.2019.03.018. [DOI] [PubMed] [Google Scholar]
  153. Matos AL, Bruno DF, Ambrósio AF, Santos PF. ‘The benefits of flavonoids in diabetic retinopathy. Nutrients. 2020;12:3169. doi: 10.3390/nu12103169. [DOI] [PMC free article] [PubMed] [Google Scholar]
  154. McMurtrey JJ, Tso MOM. A review of the immunologic findings observed in retinitis pigmentosa. Surv Ophthalmol. 2018;63:769–781. doi: 10.1016/j.survophthal.2018.03.002. [DOI] [PubMed] [Google Scholar]
  155. McNamee SM, Akula M, Love Z, Nasraty N, Nystuen K, Singh P, Upadhyay AK, DeAngelis MM, Haider NB. Evaluating therapeutic potential of NR2E3 doses in the rd7 mouse model of retinal degeneration. Sci Rep. 2024;14:16490. doi: 10.1038/s41598-024-67095-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  156. Medrano-Jiménez E, Meza-Sosa KF, Urbán-Aragón JA, Secundino I, Pedraza-Alva G, Pérez-Martínez L. Microglial activation in Alzheimer’s disease: The role of flavonoids and microRNAs. J Leukoc Biol. 2022;112:47–77. doi: 10.1002/JLB.3MR1021-531R. [DOI] [PubMed] [Google Scholar]
  157. Minocha T, Birla H, Obaid AA, Rai V, Sushma P, Shivamallu C, Moustafa M, Al-Shehri M, Al-Emam A, Tikhonova MA, Yadav SK, Poeggeler B, Singh D, Singh SK. Flavonoids as promising neuroprotectants and their therapeutic potential against Alzheimer’s disease. Oxid Med Cell Longev. 2022;2022:6038996. doi: 10.1155/2022/6038996. [DOI] [PMC free article] [PubMed] [Google Scholar]
  158. Mitter SK, Song C, Qi X, Mao H, Rao H, Akin D, Lewin A, Grant M, Dunn W, Jr, Ding J, Bowes Rickman C, Boulton M. Dysregulated autophagy in the RPE is associated with increased susceptibility to oxidative stress and AMD. Autophagy. 2014;10:1989–2005. doi: 10.4161/auto.36184. [DOI] [PMC free article] [PubMed] [Google Scholar]
  159. Mohan KV, Mishra A, Muniyasamy A, Sinha P, Sahu P, Kesarwani A, Jain K, Nagarajan P, Scaria V, Agarwal M, Akhter NS, Gupta C, Upadhyay P. Immunological consequences of compromised ocular immune privilege accelerate retinal degeneration in retinitis pigmentosa. Orphanet J Rare Dis. 2022;17:378. doi: 10.1186/s13023-022-02528-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  160. Moshtaghion SM, Caballano-Infantes E, Plaza Reyes Á, Valdés-Sánchez L, Fernández PG, de la Cerda B, Riga MS, Álvarez-Dolado M, Peñalver P, Morales JC, Díaz-Corrales FJ. Piceid octanoate protects retinal cells against oxidative damage by regulating the sirtuin 1/poly-ADP-ribose polymerase 1 axis in vitro and in rd10 mice. Antioxidants (Basel) 2024;13:201. doi: 10.3390/antiox13020201. [DOI] [PMC free article] [PubMed] [Google Scholar]
  161. Móvio MI, de Lima-Vasconcellos TH, Dos Santos GB, Echeverry MB, Colombo E, Mattos LS, Resende RR, Kihara AH. Retinal organoids from human-induced pluripotent stem cells: From studying retinal dystrophies to early diagnosis of Alzheimer’s and Parkinson’s disease. Semin Cell Dev Biol. 2023;144:77–86. doi: 10.1016/j.semcdb.2022.09.011. [DOI] [PubMed] [Google Scholar]
  162. Muñoz-Reyes D, Morales AI, Prieto M. Transit and metabolic pathways of quercetin in tubular cells: involvement of its antioxidant properties in the kidney. Antioxidants (Basel) 2021;10:909. doi: 10.3390/antiox10060909. [DOI] [PMC free article] [PubMed] [Google Scholar]
  163. Murakami Y, Ishikawa K, Nakao S, Sonoda KH. Innate immune response in retinal homeostasis and inflammatory disorders. Prog Retin Eye Res. 2020;74:100778. doi: 10.1016/j.preteyeres.2019.100778. [DOI] [PubMed] [Google Scholar]
  164. Murenu E, Gerhardt MJ, Biel M, Michalakis S. ‘More than meets the eye: The role of microglia in healthy and diseased retina. Front Immunol. 2022;13:1006897. doi: 10.3389/fimmu.2022.1006897. [DOI] [PMC free article] [PubMed] [Google Scholar]
  165. Murota K, Nakamura Y, Uehara M. Flavonoid metabolism: the interaction of metabolites and gut microbiota. Biosci Biotechnol Biochem. 2018;82:600–610. doi: 10.1080/09168451.2018.1444467. [DOI] [PubMed] [Google Scholar]
  166. Nagai N, Kubota S, Tsubota K, Ozawa Y. Resveratrol prevents the development of choroidal neovascularization by modulating AMP-activated protein kinase in macrophages and other cell types. J Nutr Biochem. 2014;25:1218–1225. doi: 10.1016/j.jnutbio.2014.05.015. [DOI] [PubMed] [Google Scholar]
  167. Nagata JM, Bashir A, Weinstein S, Al-Shoaibi AAA, Shao IY, Ganson KT, Testa A, Garber AK. Social epidemiology of the Mediterranean-dietary approaches to stop hypertension intervention for neurodegenerative delay (MIND) diet among early adolescents: the Adolescent Brain Cognitive Development Study. Pediatr Res. 2024;96:230–236. doi: 10.1038/s41390-023-02959-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  168. Najafi M, Nikpayam O, Tavakoli-Rouzbehani OM, Papi S, Amrollahi Bioky A, Ahmadiani ES, Sohrab G. A comprehensive insight into the potential effects of resveratrol supplementation on SIRT-1: A systematic review. Diabetes Metab Syndr. 2021;15:102224. doi: 10.1016/j.dsx.2021.102224. [DOI] [PubMed] [Google Scholar]
  169. Nakagawa T, Zhu H, Morishima N, Li E, Xu J, Yankner BA, Yuan J. Caspase-12 mediates endoplasmic-reticulum-specific apoptosis and cytotoxicity by amyloid-beta. Nature. 2000;403:98–103. doi: 10.1038/47513. [DOI] [PubMed] [Google Scholar]
  170. Di Napoli R, Balzano N, Mascolo A, Cimmino C, Vitiello A, Zovi A, Capuano A, Boccellino M. What is the role of nutraceutical products in cancer patients? A systematic review of randomized clinical trials. Nutrients. 2023;15:3249. doi: 10.3390/nu15143249. [DOI] [PMC free article] [PubMed] [Google Scholar]
  171. Naróg D, Sobkowiak A. Electrochemistry of flavonoids. Molecules. 2023;28:7618. doi: 10.3390/molecules28227618. [DOI] [PMC free article] [PubMed] [Google Scholar]
  172. Navneet S, Wilson K, Rohrer B. Müller glial cells in the macula: their activation and cell-cell interactions in age-related macular degeneration. Invest Ophthalmol Vis Sci. 2024;65:42. doi: 10.1167/iovs.65.2.42. [DOI] [PMC free article] [PubMed] [Google Scholar]
  173. Noguchi JL, Seu MY, Qiao JB, Tan IR, Swaminathan SR, McDonnell JF, Tan Z, Bu P. Kaempferol protects against retinal photoreceptor degeneration in a mouse model of light-induced retinal injury. J Ocul Pharmacol Ther. 2023;39:80–85. doi: 10.1089/jop.2022.0128. [DOI] [PubMed] [Google Scholar]
  174. Nones J, Stipursky J, Costa SL, Gomes FC. Flavonoids and astrocytes crosstalking: Implications for brain development and pathology. Neurochem Res. 2010;35:955–966. doi: 10.1007/s11064-010-0144-0. [DOI] [PubMed] [Google Scholar]
  175. Okonkwo ON, Hassan AO, Agweye CT, Victor U, Akanbi T. Clinical presentation and macular morphology in retinitis pigmentosa patients. Ann Afr Med. 2023;22:451–455. doi: 10.4103/aam.aam_181_22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  176. Olivares-González L, Velasco S, Campillo I, Salom D, González-García E, Soriano Del Castillo JM, Rodrigo R. Nutraceutical supplementation ameliorates visual function, retinal degeneration, and redox status in rd10 mice. Antioxidants (Basel) 2021;10:1033. doi: 10.3390/antiox10071033. [DOI] [PMC free article] [PubMed] [Google Scholar]
  177. Olivares-González L, Velasco S, Campillo I, Rodrigo R. ‘Retinal inflammation, cell death and inherited retinal dystrophies. Int J Mol Sci. 2021;22:2096. doi: 10.3390/ijms22042096. [DOI] [PMC free article] [PubMed] [Google Scholar]
  178. Oncel D, Corradetti G, Wakatsuki Y, Nittala MG, Velaga SB, Stambolian D, Pericak-Vance MA, Haines JL, Sadda SR. Drusen morphometrics on optical coherence tomography in eyes with age-related macular degeneration and normal aging. Graefes Arch Clin Exp Ophthalmol. 2023;261:2525–2533. doi: 10.1007/s00417-023-06088-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  179. O’Neal TB, Tripathy K, Luther EE. StatPearls [Internet] Treasure Island (FL): StatPearls Publishing; 2024. Retinitis Pigmentosa. [PubMed] [Google Scholar]
  180. Ortega JT, Parmar T, Golczak M, Jastrzebska B. Protective effects of flavonoids in acute models of light-induced retinal degeneration. Mol Pharmacol. 2021;99:60–77. doi: 10.1124/molpharm.120.000072. [DOI] [PMC free article] [PubMed] [Google Scholar]
  181. Ortega JT, Parmar T, Carmena-Bargueño M, Pérez-Sánchez H, Jastrzebska B. Flavonoids improve the stability and function of P23H rhodopsin slowing down the progression of retinitis pigmentosa in mice. J Neurosci Res. 2022;100:1063–1083. doi: 10.1002/jnr.25021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  182. Ortega JT, Jastrzebska B. Neuroinflammation as a therapeutic target in retinitis pigmentosa and quercetin as its potential modulator. Pharmaceutics. 2021;13:1935. doi: 10.3390/pharmaceutics13111935. [DOI] [PMC free article] [PubMed] [Google Scholar]
  183. Ortega JT, Parmar T, Jastrzebska B. Flavonoids enhance rod opsin stability, folding, and self-association by directly binding to ligand-free opsin and modulating its conformation. J Biol Chem. 2019;294:8101–8122. doi: 10.1074/jbc.RA119.007808. [DOI] [PMC free article] [PubMed] [Google Scholar]
  184. Paik SS, Jeong E, Jung SW, Ha TJ, Kang S, Sim S, Jeon JH, Chun MH, Kim IB. Anthocyanins from the seed coat of black soybean reduce retinal degeneration induced by N-methyl-N-nitrosourea. Exp Eye Res. 2012;97:55–62. doi: 10.1016/j.exer.2012.02.010. [DOI] [PubMed] [Google Scholar]
  185. Paquet-Durand F, Silva J, Talukdar T, Johnson LE, Azadi S, van Veen T, Ueffing M, Hauck SM, Ekström PA. Excessive activation of poly(ADP-ribose) polymerase contributes to inherited photoreceptor degeneration in the retinal degeneration 1 mouse. J Neurosci. 2007;27:10311–10319. doi: 10.1523/JNEUROSCI.1514-07.2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  186. Paquet-Durand F, Hauck SM, van Veen T, Ueffing M, Ekström P. PKG activity causes photoreceptor cell death in two retinitis pigmentosa models. J Neurochem. 2009;108:796–810. doi: 10.1111/j.1471-4159.2008.05822.x. [DOI] [PubMed] [Google Scholar]
  187. Parmar T, Ortega JT, Jastrzebska B. Retinoid analogs and polyphenols as potential therapeutics for age-related macular degeneration. Exp Biol Med (Maywood) 2020;245:1615–1625. doi: 10.1177/1535370220926938. [DOI] [PMC free article] [PubMed] [Google Scholar]
  188. Pawloff M, Gerendas BS, Deak G, Bogunovic H, Gruber A, Schmidt-Erfurth U. Performance of retinal fluid monitoring in OCT imaging by automated deep learning versus human expert grading in neovascular AMD. Eye (Lond) 2023;37:3793–3800. doi: 10.1038/s41433-023-02615-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  189. Pawlowska E, Szczepanska J, Koskela A, Kaarniranta K, Blasiak J. Dietary polyphenols in age-related macular degeneration: protection against oxidative stress and beyond. Oxid Med Cell Longev. 2019;2019:9682318. doi: 10.1155/2019/9682318. [DOI] [PMC free article] [PubMed] [Google Scholar]
  190. Peddada KV, Brown A, Verma V, Nebbioso M. Therapeutic potential of curcumin in major retinal pathologies. Int Ophthalmol. 2019;39:725–734. doi: 10.1007/s10792-018-0845-y. [DOI] [PubMed] [Google Scholar]
  191. Peng B, Xiao J, Wang K, So KF, Tipoe GL, Lin B. Suppression of microglial activation is neuroprotective in a mouse model of human retinitis pigmentosa. J Neurosci. 2014;34:8139–8150. doi: 10.1523/JNEUROSCI.5200-13.2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  192. Pennington KL, DeAngelis MM. Epidemiology of age-related macular degeneration (AMD): associations with cardiovascular disease phenotypes and lipid factors. Eye Vis (Lond) 2016;3:34. doi: 10.1186/s40662-016-0063-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  193. Pennington KL, DeAngelis MM. Epidemiology of age-related macular degeneration (AMD): associations with cardiovascular disease phenotypes and lipid factors. Eye Vis (Lond) 2016;3:34. doi: 10.1186/s40662-016-0063-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  194. Perdices L, Fuentes-Broto L, Segura F, Cavero A, Orduna-Hospital E, Insa-Sánchez G, Sánchez-Cano AI, Fernández-Sánchez L, Cuenca N, Pinilla I. Systemic epigallocatechin gallate protects against retinal degeneration and hepatic oxidative stress in the P23H-1 rat. Neural Regen Res. 2022;17:625–631. doi: 10.4103/1673-5374.320990. [DOI] [PMC free article] [PubMed] [Google Scholar]
  195. Piano I, D’Antongiovanni V, Testai L, Calderone V, Gargini C. A nutraceutical strategy to slowing down the progression of cone death in an animal model of retinitis pigmentosa. Front Neurosci. 2019;13:461. doi: 10.3389/fnins.2019.00461. [DOI] [PMC free article] [PubMed] [Google Scholar]
  196. Piano I, Corsi F, Polini B, Gargini C. Nutraceutical molecules slow down retinal degeneration, in tvrm4 mice a model of retinitis pigmentosa, by genetic modulation of anti-oxidant pathway. Front Neurosci. 2022;16:868750. doi: 10.3389/fnins.2022.868750. [DOI] [PMC free article] [PubMed] [Google Scholar]
  197. Piccolella S, Crescente G, Candela L, Pacifico S. Nutraceutical polyphenols: New analytical challenges and opportunities. J Pharm Biomed Anal. 2019;175:112774. doi: 10.1016/j.jpba.2019.07.022. [DOI] [PubMed] [Google Scholar]
  198. Pierrache LHM, Ghafaryasl B, Khan MI, Yzer S, van Genderen MM, Schuil J, Boonstra FN, Pott JWR, de Faber JTHN, Tjon-Fo-Sang MJH, Vermeer KA, Cremers FPM, Klaver CCW, van den Born LI. Longitudinal study of rpe65-associated inherited retinal degenerations. Retina. 2020;40:1812–1828. doi: 10.1097/IAE.0000000000002681. [DOI] [PubMed] [Google Scholar]
  199. Ploier B, Caro LN, Morizumi T, Pandey K, Pearring JN, Goren MA, Finnemann SC, Graumann J, Arshavsky VY, Dittman JS, Ernst OP, Menon AK. Dimerization deficiency of enigmatic retinitis pigmentosa-linked rhodopsin mutants. Nat Commun. 2016;7:12832. doi: 10.1038/ncomms12832. [DOI] [PMC free article] [PubMed] [Google Scholar]
  200. Prajapati R, Seong SH, Park SE, Paudel P, Jung HA, Choi JS. Isoliquiritigenin, a potent human monoamine oxidase inhibitor, modulates dopamine D1, D3, and vasopressin V1A receptors. Sci Rep. 2021;11:23528. doi: 10.1038/s41598-021-02843-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  201. Provis JM, Penfold PL, Cornish EE, Sandercoe TM, Madigan MC. Anatomy and development of the macula: specialisation and the vulnerability to macular degeneration. Clin Exp Optom. 2005;88:269–281. doi: 10.1111/j.1444-0938.2005.tb06711.x. [DOI] [PubMed] [Google Scholar]
  202. Punzo C, Kornacker K, Cepko CL. Stimulation of the insulin/mTOR pathway delays cone death in a mouse model of retinitis pigmentosa. Nat Neurosci. 2009;12:44–52. doi: 10.1038/nn.2234. [DOI] [PMC free article] [PubMed] [Google Scholar]
  203. Puri V, Nagpal M, Singh I, Singh M, Dhingra GA, Huanbutta K, Dheer D, Sharma A, Sangnim T. A comprehensive review on nutraceuticals: therapy support and formulation challenges. Nutrients. 2022;14:4637. doi: 10.3390/nu14214637. [DOI] [PMC free article] [PubMed] [Google Scholar]
  204. Qi S, Wang C, Song D, Song Y, Dunaief JL. Intraperitoneal injection of (−)-Epigallocatechin-3-gallate protects against light-induced photoreceptor degeneration in the mouse retina. Mol Vis. 2017;23:171–178. [PMC free article] [PubMed] [Google Scholar]
  205. Rakotondrabe TF, Fan MX, Muema FW, Guo MQ. Modulating inflammation-mediated diseases via natural phenolic compounds loaded in nanocarrier systems. Pharmaceutics. 2023;15:699. doi: 10.3390/pharmaceutics15020699. [DOI] [PMC free article] [PubMed] [Google Scholar]
  206. Retina International 2025 Available at: https://retina-international.org/ (Accessed: 13 May 2024) [Google Scholar]
  207. Reyes-Reveles J, Dhingra A, Alexander D, Bragin A, Philp NJ, Boesze-Battaglia K. Phagocytosis-dependent ketogenesis in retinal pigment epithelium. J Biol Chem. 2017;292:8038–8047. doi: 10.1074/jbc.M116.770784. [DOI] [PMC free article] [PubMed] [Google Scholar]
  208. Roche SL, Wyse-Jackson AC, Byrne AM, Ruiz-Lopez AM, Cotter TG. Alterations to retinal architecture prior to photoreceptor loss in a mouse model of retinitis pigmentosa. Int J Dev Biol. 2016;60:127–139. doi: 10.1387/ijdb.150400tc. [DOI] [PubMed] [Google Scholar]
  209. Roche SL, Ruiz-Lopez AM, Moloney JN, Byrne AM, Cotter TG. Microglial-induced Müller cell gliosis is attenuated by progesterone in a mouse model of retinitis pigmentosa. Glia. 2018;66:295–310. doi: 10.1002/glia.23243. [DOI] [PubMed] [Google Scholar]
  210. Ruan Y, Jiang S, Gericke A. Age-related macular degeneration: role of oxidative stress and blood vessels. Int J Mol Sci. 2021;22:1296. doi: 10.3390/ijms22031296. [DOI] [PMC free article] [PubMed] [Google Scholar]
  211. Sadda SR, Chakravarthy U, Birch DG, Staurenghi G, Henry EC, Brittain C. Clinical endpoints for the study of geographic atrophy secondary to age-related macular degeneration. Retina. 2016;36:1806–1822. doi: 10.1097/IAE.0000000000001283. [DOI] [PMC free article] [PubMed] [Google Scholar]
  212. Sadigh S, Luo X, Cideciyan AV, Sumaroka A, Boxley SL, Hall LM, Sheplock R, Feuer WJ, Stambolian DS, Jacobson SG. Drusen and photoreceptor abnormalities in African-Americans with intermediate non-neovascular age-related macular degeneration. Curr Eye Res. 2015;40:398–406. doi: 10.3109/02713683.2014.925934. [DOI] [PMC free article] [PubMed] [Google Scholar]
  213. Sancho-Pelluz J, Alavi MV, Sahaboglu A, Kustermann S, Farinelli P, Azadi S, van Veen T, Romero FJ, Paquet-Durand F, Ekström P. Excessive HDAC activation is critical for neurodegeneration in the rd1 mouse. Cell Death Dis. 2010;1:e24. doi: 10.1038/cddis.2010.4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  214. Sanges D, Comitato A, Tammaro R, Marigo V. Apoptosis in retinal degeneration involves cross-talk between apoptosis-inducing factor (AIF) and caspase-12 and is blocked by calpain inhibitors. Proc Natl Acad Sci U S A. 2006;103:17366–17371. doi: 10.1073/pnas.0606276103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  215. Dos Santos NCL, Malta SM, Franco RR, Silva HCG, Silva MH, Rodrigues TS, de Oliveira RM, Araújo TN, Augusto SC, Espindola FS, Ueira-Vieira C. Antioxidant and anti-Alzheimer’s potential of Tetragonisca angustula (Jataí) stingless bee pollen. Sci Rep. 2024;14:308. doi: 10.1038/s41598-023-51091-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  216. Sarabia-Vallejo Á, Caja MDM, Olives AI, Martín MA, Menéndez JC. Cyclodextrin inclusion complexes for improved drug bioavailability and activity: synthetic and analytical aspects. Pharmaceutics. 2023;15:2345. doi: 10.3390/pharmaceutics15092345. [DOI] [PMC free article] [PubMed] [Google Scholar]
  217. Schnichels S, Paquet-Durand F, Löscher M, Tsai T, Hurst J, Joachim SC, Klettner A. Retina in a dish: Cell cultures, retinal explants and animal models for common diseases of the retina. Prog Retin Eye Res. 2021;81:100880. doi: 10.1016/j.preteyeres.2020.100880. [DOI] [PubMed] [Google Scholar]
  218. Scott PA, Kaplan HJ, McCall MA. Prenatal exposure to curcumin protects rod photoreceptors in a transgenic Pro23His swine model of retinitis pigmentosa. Transl Vis Sci Technol. 2015;4:5. doi: 10.1167/tvst.4.5.5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  219. Scrivo A, Bourdenx M, Pampliega O, Cuervo AM. Selective autophagy as a potential therapeutic target for neurodegenerative disorders. Lancet Neurol. 2018;17:802–815. doi: 10.1016/S1474-4422(18)30238-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  220. Selvam S, Kumar T, Fruttiger M. Retinal vasculature development in health and disease. Prog Retin Eye Res. 2018;63:1–19. doi: 10.1016/j.preteyeres.2017.11.001. [DOI] [PubMed] [Google Scholar]
  221. Senabouth A, et al. Transcriptomic and proteomic retinal pigment epithelium signatures of age-related macular degeneration. Nat Commun. 2022;13:4233. doi: 10.1038/s41467-022-31707-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  222. Serafini M, Peluso I, Raguzzini A. Flavonoids as anti-inflammatory agents. Proc Nutr Soc. 2010;69:273–278. doi: 10.1017/S002966511000162X. [DOI] [PubMed] [Google Scholar]
  223. 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. doi: 10.1016/j.foodchem.2022.132531. [DOI] [PubMed] [Google Scholar]
  224. Sheu SJ, Liu NC, Chen JL. Resveratrol protects human retinal pigment epithelial cells from acrolein-induced damage. J Ocul Pharmacol Ther. 2010;26:231–236. doi: 10.1089/jop.2009.0137. [DOI] [PubMed] [Google Scholar]
  225. Shi Y, Zhang Q, Zhou H, Wang L, Chu Z, Jiang X, Shen M, Thulliez M, Lyu C, Feuer W, de Sisternes L, Durbin MK, Gregori G, Wang RK, Rosenfeld PJ. Correlations between choriocapillaris and choroidal measurements and the growth of geographic atrophy using swept source OCT imaging. Am J Ophthalmol. 2021;224:321–331. doi: 10.1016/j.ajo.2020.12.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  226. Sinha T, Naash MI, Al-Ubaidi MR. The symbiotic relationship between the neural retina and retinal pigment epithelium is supported by utilizing differential metabolic pathways. iScience. 2020;23:101004. doi: 10.1016/j.isci.2020.101004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  227. Somasundaran S, Constable IJ, Mellough CB, Carvalho LS. Retinal pigment epithelium and age‐related macular degeneration: A review of major disease mechanisms. Clin Exp Ophthalmol. 2020;48:1043–1056. doi: 10.1111/ceo.13834. [DOI] [PMC free article] [PubMed] [Google Scholar]
  228. Song K, Na JY, Kim S, Kwon J. Rutin upregulates neurotrophic factors resulting in attenuation of ethanol‐induced oxidative stress in HT22 hippocampal neuronal cells. J Sci Food Agric. 2015;95:2117–2123. doi: 10.1002/jsfa.6927. [DOI] [PubMed] [Google Scholar]
  229. Stasheff SF, Shankar M, Andrews MP. Developmental time course distinguishes changes in spontaneous and light-evoked retinal ganglion cell activity in rd1 and rd10 mice. J Neurophysiol. 2011;105:3002–3009. doi: 10.1152/jn.00704.2010. [DOI] [PubMed] [Google Scholar]
  230. Storti F, Raphael G, Griesser V, Klee K, Drawnel F, Willburger C, Scholz R, Langmann T, von Eckardstein A, Fingerle J, Grimm C, Maugeais C. Regulated efflux of photoreceptor outer segment-derived cholesterol by human RPE cells. Exp Eye Res. 2017;165:65–77. doi: 10.1016/j.exer.2017.09.008. [DOI] [PubMed] [Google Scholar]
  231. Sulaiman RS, Basavarajappa HD, Corson TW. Natural product inhibitors of ocular angiogenesis. Exp Eye Res. 2014;129:161–171. doi: 10.1016/j.exer.2014.10.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  232. Sundaram V, Wilde C, Aboshiha J, Cowing J, Han C, Langlo CS, Chana R, Davidson AE, Sergouniotis PI, Bainbridge JW, Ali RR, Dubra A, Rubin G, Webster AR, Moore AT, Nardini M, Carroll J, Michaelides M. Retinal structure and function in achromatopsia: implications for gene therapy. Ophthalmology. 2014;121:234–245. doi: 10.1016/j.ophtha.2013.08.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  233. Sysak S, Czarczynska-Goslinska B, Szyk P, Koczorowski T, Mlynarczyk DT, Szczolko W, Lesyk R, Goslinski T. Metal nanoparticle-flavonoid connections: synthesis, physicochemical and biological properties, as well as potential applications in medicine. Nanomaterials (Basel) 2023;13:1531. doi: 10.3390/nano13091531. [DOI] [PMC free article] [PubMed] [Google Scholar]
  234. Tan W, Zou J, Yoshida S, Jiang B, Zhou Y. The role of inflammation in age-related macular degeneration. Int J Biol Sci. 2020;16:2989–3001. doi: 10.7150/ijbs.49890. [DOI] [PMC free article] [PubMed] [Google Scholar]
  235. Tao Y, Chen T, Fang W, Peng G, Wang L, Qin L, Liu B, Fei Huang Y. The temporal topography of the N-Methyl- N-nitrosourea induced photoreceptor degeneration in mouse retina. Sci Rep. 2015;5:18612. doi: 10.1038/srep18612. [DOI] [PMC free article] [PubMed] [Google Scholar]
  236. Tao Y, Chen T, Yang GQ, Peng GH, Yan ZJ, Huang YF. Anthocyanin can arrest the cone photoreceptor degeneration and act as a novel treatment for retinitis pigmentosa. Int J Ophthalmol. 2016;9:153–158. doi: 10.18240/ijo.2016.01.25. [DOI] [PMC free article] [PubMed] [Google Scholar]
  237. Tao Y, Chen T, Liu Z, Xu W, Qin L, Peng G, Huang YF. Integrating Topographic Measures to Explore the Protective Effects of peonidin against the N-methyl-N-nitrosourea induced photoreceptor degeneration. Cell Physiol Biochem. 2016;38:893–908. doi: 10.1159/000443043. [DOI] [PubMed] [Google Scholar]
  238. Tomita Y, Qiu C, Bull E, Allen W, Kotoda Y, Talukdar S, Smith LEH, Fu Z. Müller glial responses compensate for degenerating photoreceptors in retinitis pigmentosa. Exp Mol Med. 2021;53:1748–1758. doi: 10.1038/s12276-021-00693-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
  239. Trachsel-Moncho L, Benlloch-Navarro S, Fernández-Carbonell Á, Ramírez-Lamelas DT, Olivar T, Silvestre D, Poch E, Miranda M. Oxidative stress and autophagy-related changes during retinal degeneration and development. Cell Death Dis. 2018;9:812. doi: 10.1038/s41419-018-0855-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  240. Ulrich-Merzenich G, Zeitler H, Vetter H, Kraft K. Synergy research: vitamins and secondary plant components in the maintenance of the redox-homeostasis and in cell signaling. Phytomedicine. 2009;16:2–16. doi: 10.1016/j.phymed.2008.11.007. [DOI] [PubMed] [Google Scholar]
  241. Uren PJ, Lee JT, Doroudchi MM, Smith AD, Horsager A. A profile of transcriptomic changes in the rd10 mouse model of retinitis pigmentosa. Mol Vis. 2014;20:1612–1628. [PMC free article] [PubMed] [Google Scholar]
  242. Valdés-Sánchez L, García-Delgado AB, Montero-Sánchez A, de la Cerda B, Lucas R, Peñalver P, Morales JC, Bhattacharya SS, Díaz-Corrales FJ. The resveratrol prodrug JC19 delays retinal degeneration in rd10 mice. Adv Exp Med Biol. 2019;1185:457–462. doi: 10.1007/978-3-030-27378-1_75. [DOI] [PubMed] [Google Scholar]
  243. Vasireddy V, Chavali VR, Joseph VT, Kadam R, Lin JH, Jamison JA, Kompella UB, Reddy GB, Ayyagari R. Rescue of photoreceptor degeneration by curcumin in transgenic rats with P23H rhodopsin mutation. PLoS One. 2011;6:e21193. doi: 10.1371/journal.pone.0021193. [DOI] [PMC free article] [PubMed] [Google Scholar]
  244. Vasudevan S, Senapati S, Pendergast M, Park PS. Aggregation of rhodopsin mutants in mouse models of autosomal dominant retinitis pigmentosa. Nat Commun. 2024;15:1451. doi: 10.1038/s41467-024-45748-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  245. Verbakel SK, van Huet RAC, Boon CJF, den Hollander AI, Collin RWJ, Klaver CCW, Hoyng CB, Roepman R, Klevering BJ. Prog Retin Eye Res. 2018;66:157–186. doi: 10.1016/j.preteyeres.2018.03.005. [DOI] [PubMed] [Google Scholar]
  246. Vingolo EM, Casillo L, Contento L, Toja F, Florido A. Retinitis pigmentosa (RP): The role of oxidative stress in the degenerative process progression. Biomedicines. 2022;10:582. doi: 10.3390/biomedicines10030582. [DOI] [PMC free article] [PubMed] [Google Scholar]
  247. Voisin A, Pénaguin A, Gaillard A, Leveziel N. Stem cell therapy in retinal diseases. Neural Regen Res. 2023;18:1478–1485. doi: 10.4103/1673-5374.361537. [DOI] [PMC free article] [PubMed] [Google Scholar]
  248. Vosler PS, Sun D, Wang S, Gao Y, Kintner DB, Signore AP, Cao G, Chen J. Calcium dysregulation induces apoptosis-inducing factor release: cross-talk between PARP-1- and calpain-signaling pathways. Exp Neurol. 2009;218:213–220. doi: 10.1016/j.expneurol.2009.04.032. [DOI] [PMC free article] [PubMed] [Google Scholar]
  249. Vu JT, Wang E, Wu J, Sun YJ, Velez G, Bassuk AG, Lee SH, Mahajan VB. Calpains as mechanistic drivers and therapeutic targets for ocular disease. Trends Mol Med. 2022;28:644–661. doi: 10.1016/j.molmed.2022.05.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  250. Vue B, Zhang S, Chen QH. Synergistic effects of dietary natural products as anti-prostate cancer agents. Nat Prod Commun. 2015;10:2179–2188. [PubMed] [Google Scholar]
  251. Wang D, Chen Y, Li J, Wu E, Tang T, Singla RK, Shen B, Zhang M. Natural products for the treatment of age-related macular degeneration. Phytomedicine. 2024;130:155522. doi: 10.1016/j.phymed.2024.155522. [DOI] [PubMed] [Google Scholar]
  252. Wang G, Wang Y, Yao L, Gu W, Zhao S, Shen Z, Lin Z, Liu W, Yan T. Pharmacological activity of quercetin: an updated review. Evid Based Complement Alternat Med. 2022;2022:3997190. doi: 10.1155/2022/3997190. [DOI] [PMC free article] [PubMed] [Google Scholar]
  253. Wang J, Li M, Geng Z, Khattak S, Ji X, Wu D, Dang Y. Role of oxidative stress in retinal disease and the early intervention strategies: a review. Oxid Med Cell Longev. 2022;2022:7836828. doi: 10.1155/2022/7836828. [DOI] [PMC free article] [PubMed] [Google Scholar]
  254. Wang TH, Tseng WC, Leu YL, Chen CY, Lee WC, Chi YC, Cheng SF, Lai CY, Kuo CH, Yang SL, Yang SH, Shen JJ, Feng CH, Wu CC, Hwang TL, Wang CJ, Wang SH, Chen CC. The flavonoid corylin exhibits lifespan extension properties in mouse. Nat Commun. 2022;13:1238. doi: 10.1038/s41467-022-28908-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  255. Wang Y, Huo Y, Zhao L, Lu F, Wang O, Yang X, Ji B, Zhou F. Cyanidin-3-glucoside and its phenolic acid metabolites attenuate visible light-induced retinal degeneration in vivo via activation of Nrf2/HO-1 pathway and NF-κB suppression. Mol Nutr Food Res. 2016;60:1564–1577. doi: 10.1002/mnfr.201501048. [DOI] [PubMed] [Google Scholar]
  256. Wang Y, Yin Z, Gao L, Sun D, Hu X, Xue L, Dai J, Zeng Y, Chen S, Pan B, Chen M, Xie J, Xu H. Curcumin delays retinal degeneration by regulating microglia activation in the retina of rd1 mice. Cell Physiol Biochem. 2017;44:479–493. doi: 10.1159/000485085. [DOI] [PubMed] [Google Scholar]
  257. Whelan K, Bancil AS, Lindsay JO, Chassaing B. Ultra-processed foods and food additives in gut health and disease. Nat Rev Gastroenterol Hepatol. 2024;21:406–427. doi: 10.1038/s41575-024-00893-5. [DOI] [PubMed] [Google Scholar]
  258. Wong WL, Su X, Li X, Cheung CM, Klein R, Cheng CY, Wong TY. Global prevalence of age-related macular degeneration and disease burden projection for 2020 and 2040: a systematic review and meta-analysis. Lancet Glob Health. 2014;2:e106–116. doi: 10.1016/S2214-109X(13)70145-1. [DOI] [PubMed] [Google Scholar]
  259. Wright AF, Chakarova CF, Abd El-Aziz MM, Bhattacharya SS. Photoreceptor degeneration: genetic and mechanistic dissection of a complex trait. Nat Rev Genet. 2010;11:273–284. doi: 10.1038/nrg2717. [DOI] [PubMed] [Google Scholar]
  260. Wronka M, Krzemińska J, Młynarska E, Rysz J, Franczyk B. The influence of lifestyle and treatment on oxidative stress and inflammation in diabetes. Int J Mol Sci. 2022;23:15743. doi: 10.3390/ijms232415743. [DOI] [PMC free article] [PubMed] [Google Scholar]
  261. Wu L, Chu L, Pang Y, Huo J, Cao H, Tian Q, Gao Q. Effects of dietary supplements, foods, and dietary patterns in Parkinson’s disease: meta-analysis and systematic review of randomized and crossover studies. Eur J Clin Nutr. 2024;78:365–375. doi: 10.1038/s41430-024-01411-1. [DOI] [PubMed] [Google Scholar]
  262. Wu Q, Gao ZJ, Yu X, Wang P. Dietary regulation in health and disease. Signal Transduct Target Ther. 2022;7:252. doi: 10.1038/s41392-022-01104-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
  263. Xing W, Gao W, Zhao Z, Xu X, Bu H, Su H, Mao G, Chen J. Dietary flavonoids intake contributes to delay biological aging process: analysis from NHANES dataset. J Transl Med. 2023;21:492. doi: 10.1186/s12967-023-04321-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  264. Xu H, Cao L, Chen Y, Zhou C, Xu J, Zhang Z, Li X, Liu L, Lu J. Single-cell RNA sequencing reveals the heterogeneity and interactions of immune cells and Müller glia during zebrafish retina regeneration. Neural Regen Res. 2024;20:3635–3648. doi: 10.4103/NRR.NRR-D-23-02083. [DOI] [PMC free article] [PubMed] [Google Scholar]
  265. Xu J, Morris L, Thapa A, Ma H, Michalakis S, Biel M, Baehr W, Peshenko IV, Dizhoor AM, Ding XQ. cGMP accumulation causes photoreceptor degeneration in CNG channel deficiency: evidence of cGMP cytotoxicity independently of enhanced CNG channel function. J Neurosci. 2013;33:14939–14948. doi: 10.1523/JNEUROSCI.0909-13.2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  266. Xu J, Tu Y, Wang Y, Xu X, Sun X, Xie L, Zhao Q, Guo Y, Gu Y, Du J, Du S, Zhu M, Song E. Prodrug of epigallocatechin-3-gallate alleviates choroidal neovascularization via down-regulating HIF-1α/VEGF/VEGFR2 pathway and M1 type macrophage/microglia polarization. Biomed Pharmacother. 2020;121:109606. doi: 10.1016/j.biopha.2019.109606. [DOI] [PubMed] [Google Scholar]
  267. Xu Q, Fu Q, Li Z, Liu H, Wang Y, Lin X, He R, Zhang X, Ju Z, Campisi J, Kirkland JL, Sun Y. The flavonoid procyanidin C1 has senotherapeutic activity and increases lifespan in mice. Nat Metab. 2021;3:1706–1726. doi: 10.1038/s42255-021-00491-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  268. Xu XJ, Wang SM, Jin Y, Hu YT, Feng K, Ma ZZ. Melatonin delays photoreceptor degeneration in a mouse model of autosomal recessive retinitis pigmentosa. J Pineal Res. 2017 doi: 10.1111/jpi.12428. doi: 10.1111/jpi.12428. [DOI] [PubMed] [Google Scholar]
  269. Xu Y, Guan L, Shen T, Zhang J, Xiao X, Jiang H, Li S, Yang J, Jia X, Yin Y, Guo X, Wang J, Zhang Q. Mutations of 60 known causative genes in 157 families with retinitis pigmentosa based on exome sequencing. Hum Genet. 2014;133:1255–1271. doi: 10.1007/s00439-014-1460-2. [DOI] [PubMed] [Google Scholar]
  270. Yan J, Günter A, Das S, Mühlfriedel R, Michalakis S, Jiao K, Seeliger MW, Paquet-Durand F. Inherited retinal degeneration: PARP-dependent activation of calpain requires CNG channel activity. Biomolecules. 2022;12:455. doi: 10.3390/biom12030455. [DOI] [PMC free article] [PubMed] [Google Scholar]
  271. Yan W, Sun Y, Wang Y, Liang W, Xia Y, Yan W, Chen M, Chen T, Li D. The impacts of resveratrol on the retinal degeneration in a rat model of retinitis pigmentosa induced by alkylation: an in-vivo study. Anim Cells Syst (Seoul) 2023;27:138–148. doi: 10.1080/19768354.2023.2226695. [DOI] [PMC free article] [PubMed] [Google Scholar]
  272. Yang B, Yang K, Xi R, Chen J, Wu Y. Quercetin alleviates all- trans-retinal-induced photoreceptor apoptosis and retinal degeneration by inhibiting the ER stress-related PERK signaling. Int J Mol Sci. 2024;25:13624. doi: 10.3390/ijms252413624. [DOI] [PMC free article] [PubMed] [Google Scholar]
  273. Yang F, Ma H, Butler MR, Ding XQ. Preservation of endoplasmic reticulum (ER) Ca2+ stores by deletion of inositol-1,4,5-trisphosphate receptor type 1 promotes ER retrotranslocation, proteostasis and protein outer segment localization in cyclic nucleotide-gated channel-deficient cone photoreceptors. FASEB J. 2021;35:e21579. doi: 10.1096/fj.202002711R. [DOI] [PMC free article] [PubMed] [Google Scholar]
  274. Yang N, Sampathkumar K, Loo SCJ. Recent advances in complementary and replacement therapy with nutraceuticals in combating gastrointestinal illnesses. Clin Nutr. 2017;36:968–979. doi: 10.1016/j.clnu.2016.08.020. [DOI] [PubMed] [Google Scholar]
  275. Yi YS. Flavonoids: Nutraceuticals for rheumatic diseases via targeting of inflammasome activation. Int J Mol Sci. 2021;22:488. doi: 10.3390/ijms22020488. [DOI] [PMC free article] [PubMed] [Google Scholar]
  276. Yu C, Saban DR. Microglia preserve visual function in a mouse model of retinitis pigmentosa with rhodopsin-P23H mutant. Adv Exp Med Biol. 2023;1415:421–425. doi: 10.1007/978-3-031-27681-1_62. [DOI] [PubMed] [Google Scholar]
  277. Yuan D, Guo Y, Pu F, Yang C, Xiao X, Du H, He J, Lu S. Opportunities and challenges in enhancing the bioavailability and bioactivity of dietary flavonoids: A novel delivery system perspective. Food Chem. 2024;430:137115. doi: 10.1016/j.foodchem.2023.137115. [DOI] [PubMed] [Google Scholar]
  278. Zhang IX, Herrmann A, Leon J, Jeyarajan S, Arunagiri A, Arvan P, Gilon P, Satin LS. ER stress increases expression of intracellular calcium channel RyR1 to modify Ca2+ homeostasis in pancreatic beta cells. J Biol Chem. 2023;299:105065. doi: 10.1016/j.jbc.2023.105065. [DOI] [PMC free article] [PubMed] [Google Scholar]
  279. Zhang Q, Presswalla F, Calton M, Charniga C, Stern J, Temple S, Vollrath D, Zacks DN, Ali RR, Thompson DA, Miller JML. Highly differentiated human fetal RPE cultures are resistant to the accumulation and toxicity of lipofuscin-like material. Invest Ophthalmol Vis Sci. 2019;60:3468–3479. doi: 10.1167/iovs.19-26690. [DOI] [PMC free article] [PubMed] [Google Scholar]
  280. Zhang Q, Pan J, Liu H, Jiao Z. Characterization of the synergistic antioxidant activity of epigallocatechin gallate (EGCG) and kaempferol. Molecules. 2023;28:5265. doi: 10.3390/molecules28135265. [DOI] [PMC free article] [PubMed] [Google Scholar]
  281. Zhang X, Wang H, Kilpatrick LA, Dong TS, Gee GC, Labus JS, Osadchiy V, Beltran-Sanchez H, Wang MC, Vaughan A, Gupta A. Discrimination exposure impacts unhealthy processing of food cues: crosstalk between the brain and gut. Nat Ment Health. 2023;1:841–852. doi: 10.1038/s44220-023-00134-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  282. Zhao L, Zabel MK, Wang X, Ma W, Shah P, Fariss RN, Qian H, Parkhurst CN, Gan WB, Wong WT. Microglial phagocytosis of living photoreceptors contributes to inherited retinal degeneration. EMBO Mol Med. 2015;7:1179–1197. doi: 10.15252/emmm.201505298. [DOI] [PMC free article] [PubMed] [Google Scholar]
  283. Zhao L, Hou C, Yan N. Neuroinflammation in retinitis pigmentosa: Therapies targeting the innate immune system. Front Immunol. 2022;13:1059947. doi: 10.3389/fimmu.2022.1059947. [DOI] [PMC free article] [PubMed] [Google Scholar]
  284. Zhen F, Zou T, Wang T, Zhou Y, Dong S, Zhang H. Rhodopsin-associated retinal dystrophy: Disease mechanisms and therapeutic strategies. Front Neurosci. 2023;17:1132179. doi: 10.3389/fnins.2023.1132179. [DOI] [PMC free article] [PubMed] [Google Scholar]
  285. Zhou T, Huang Z, Sun X, Zhu X, Zhou L, Li M, Cheng B, Liu X, He C. Microglia polarization with M1/M2 phenotype changes in rd1 mouse model of retinal degeneration. Front Neuroanat. 2017;11:77. doi: 10.3389/fnana.2017.00077. [DOI] [PMC free article] [PubMed] [Google Scholar]
  286. Zhou XE, Melcher K, Xu HE. Structure and activation of rhodopsin. Acta Pharmacol Sin. 2012;33:291–299. doi: 10.1038/aps.2011.171. [DOI] [PMC free article] [PubMed] [Google Scholar]
  287. Zong Z, Cheng X, Yang Y, Qiao J, Hao J, Li F. Association between dietary flavonol intake and mortality risk in the U.S. adults from NHANES database. Sci Rep. 2024;14:4572. doi: 10.1038/s41598-024-55145-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  288. Zujko ME, Witkowska AM, Waśkiewicz A, Mirończuk-Chodakowska I. Dietary antioxidant and flavonoid intakes are reduced in the elderly. Oxid Med Cell Longev. 2015;2015:843173. doi: 10.1155/2015/843173. [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Data Availability Statement

Not applicable.


Articles from Neural Regeneration Research are provided here courtesy of Wolters Kluwer -- Medknow Publications

RESOURCES