Next Article in Journal
The Immune Response in Two Models of Traumatic Injury of the Immature Brain
Previous Article in Journal
Therapeutic Effect of Boron Neutron Capture Therapy on Boronophenylalanine Administration via Cerebrospinal Fluid Circulation in Glioma Rat Models
Previous Article in Special Issue
Molecular Mechanisms of Autophagy Decline during Aging
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Natural Autophagy Activators to Fight Age-Related Diseases

by
Vianey M. Mundo Rivera
1,
José Roberto Tlacuahuac Juárez
2,
Nadia Mireya Murillo Melo
1,3,
Norberto Leyva Garcia
3,
Jonathan J. Magaña
1,3,
Joaquín Cordero Martínez
2,* and
Guadalupe Elizabeth Jiménez Gutierrez
3,*
1
Departamento de Bioingeniería, Escuela de Ingeniería y Ciencias, Tecnologico de Monterrey, Campus Ciudad de México, Mexico City 14380, Mexico
2
Laboratorio de Bioquímica Farmacológica, Departamento de Bioquímica, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City 11340, Mexico
3
Laboratorio de Medicina Genómica, Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra, Mexico City 14389, Mexico
*
Authors to whom correspondence should be addressed.
Cells 2024, 13(19), 1611; https://doi.org/10.3390/cells13191611
Submission received: 24 August 2024 / Revised: 20 September 2024 / Accepted: 23 September 2024 / Published: 26 September 2024
(This article belongs to the Special Issue Autophagy Meets Aging 2024)

Abstract

:
The constant increase in the elderly population presents significant challenges in addressing new social, economic, and health problems concerning this population. With respect to health, aging is a primary risk factor for age-related diseases, which are driven by interconnected molecular hallmarks that influence the development of these diseases. One of the main mechanisms that has attracted more attention to aging is autophagy, a catabolic process that removes and recycles damaged or dysfunctional cell components to preserve cell viability. The autophagy process can be induced or deregulated in response to a wide range of internal or external stimuli, such as starvation, oxidative stress, hypoxia, damaged organelles, infectious pathogens, and aging. Natural compounds that promote the stimulation of autophagy regulatory pathways, such as mTOR, FoxO1/3, AMPK, and Sirt1, lead to increased levels of essential proteins such as Beclin-1 and LC3, as well as a decrease in p62. These changes indicate the activation of autophagic flux, which is known to be decreased in cardiovascular diseases, neurodegeneration, and cataracts. The regulated administration of natural compounds offers an adjuvant therapeutic alternative in age-related diseases; however, more experimental evidence is needed to support and confirm these health benefits. Hence, this review aims to highlight the potential benefits of natural compounds in regulating autophagy pathways as an alternative approach to combating age-related diseases.

1. Introduction

The global elderly population is rapidly increasing. According to the United Nations’ 2023 World Social Report, the number of people aged 65 and older is projected to double from 761 million in 2021 to 1.6 billion by 2050 [1]. This demographic shift presents significant challenges in addressing new social, economic, and health problems concerning the elderly. From the perspective of health care, aging is the primary risk factor for various age-related diseases (ARDs), including cardiovascular, neurodegenerative, and metabolic diseases [2]. Given this trend, it is urgent to understand more about the complex interplay between the molecular mechanisms and environmental factors implicated in the aging process to promote healthier aging and an improvement in quality of life.
The phenomenon of aging is driven by various interconnected molecular hallmarks that influence the development of ARDs. Key factors such as loss of proteostasis, deregulated nutrient sensing, mitochondrial dysfunction, cellular senescence, and impaired autophagy are gaining attention due to their direct impact on the progression of ARDs [3,4,5]. Various strategies have been proposed to counteract these effects; however, significant challenges and uncertainties remain.
One of the main mechanisms that has attracted more attention to aging is autophagy, a key catabolic process to remove and recycle damaged or dysfunctional cell components (protein aggregates, damaged mitochondria, peroxisomes, ribosomes, endoplasmic reticulum, endosomes, lipid droplets, and intracellular pathogens) [6] to support energy needs and maintain cellular homeostasis in order to preserve cell viability [7]. The three principal types of autophagy on eukaryotic cells are macroautophagy, microautophagy, and chaperone-mediated autophagy [8,9]. The macroautophagy process is described as the formation of the autophagosome and is distinguished by the involvement of the autophagy-related proteins (Atg) [10]. Microautophagy describes the direct translocation of cytosolic material into lysosomes via membrane invagination in order to contribute to cellular homeostasis [10,11]. In contrast, chaperone-mediated autophagy shows specificity, using only proteins as a target. This process does not depend on the autophagosome formation; therefore, the proteins are directly translocated into the lysosomes for degradation [12,13]. Macroautophagy is divided into five stages: (1) initiation, (2) nucleation, (3) expansion and elongation, (4) closure and fusion, and (5) degradation (Figure 1) [14]. The initiation stage involves the participation of protein complexes such as ULK-Atg1, Class III PI3-K complex, ATG9, Rab1, TRAPPIII, and ubiquitin-like molecules (LC3A-D), and is characterized by the formation of the omegasome, which is an enriched endoplastic reticulum domain with phosphatidylinositol-3-phosphate. The omegasome will serve as an initiation site for the formation of the phagophore, which is the membrane precursor of the autophagosome, and is generated at multiple sites throughout the cytoplasm [6,15]. In the nucleation stage, the membrane begins to expand by recruiting proteins and lipids, transforming into a primary double membrane sequestering compartment structure named the phagophore [16]. In this stage, the most important event is the recruiting of the proteins for the ATG14-Class III/PI3-K complex. This complex produces the phosphatidylinositol-3-phosphate needed for macroautophagy and for the recruiting of vacuolar protein sorting VPS15 and VPS34, as well as the release of Beclin1 and AMBRA1 from their inhibitor Bcl-2 [17]. During the expansion and elongation process, the membrane of the phagophore expands and closes around its cargo to form the autophagosome [16]. Also, the objective of this stage is to determine the site of lipidation of LC3, which is determined by the activity of the elongation complex ATG5-12/16L1. LC3 lipidation (LC3-II) is required for its association with the autophagosomal membrane; therefore, the membrane-associated LC3 will allow completion of the autophagosomal formation, also interacting with p62/SQSTM1 inside the autophagosome [18,19,20]. The closure and fusion stage are characterized by the maturation of the autophagosomes and their fusion with mature lysosomes to become autolysosomes [21]. The process is regulated by SNARE proteins and STX17, which colocalizes to complete the autophagosome formation; also, STX17 interacts with SNAP29 and lysosomal VAMP8, leading to the autophagosome–lysosome complete fusion [22]. Finally, in the degradation stage, macromolecules contained in the autolysosome are degraded by lysosomal hydrolases into monomeric units and returned to the cytoplasm to be recycled (Figure 1) [23].
The autophagy process can be induced, impeded, or deregulated in response to a wide range of internal or external stimuli [24], such as starvation, oxidative stress, hypoxia, damaged organelles, the presence of infectious pathogens, and aging [25]. Autophagy is known to decline with age, directly contributing to impaired protein homeostasis [26] that in turn leads to protein aggregates and dysfunctional protein turnover, which are molecular hallmarks of aging and increase the incidence of age-related diseases (ARDs) [27,28].
In this review, we provide an update about the role of natural compounds in the modulation of autophagy in common age-related diseases.

2. Cardiopathies

Cardiac diseases are among the most common ARDs and one of the top three causes of death worldwide. It is expected that cardiac diseases will impose a burden on the global economy and health care systems [2]. Cardiomyopathies are characterized by the suppression of autophagy in cardiac cells, which plays a crucial role in maintaining cardiac homeostasis. The disruption of this process is associated with various cardiac conditions such as cardiac hypertrophy, myocardial infarction, diabetic cardiomyopathy, and ultimately, cardiac failure [29].
Human aging is typically accompanied by cardiac hypertrophic remodeling and a progressive decline of left ventricular (LV) diastolic function [30,31]. In addition, cardiomyocytes, the essential cellular constituents of the cardiovascular system, are primarily found in a post-mitotic state, causing them to heavily depend on intact autophagy and mitophagy to preserve their physiological functions [30]. As we age, the cardiovascular system’s efficiency declines. To combat this, autophagy plays a crucial role in promoting the survival of dormant cells within the cardiovascular system and counteracting cell death caused by apoptosis or necrosis following injury [30].
The molecular mechanism of cardiovascular diseases includes two essential pathways: metabolic factors and mutations in cardiovascular disease-related sites. This section focuses on signaling cascades of PI3K/AKT/mTOR, epidermal growth factor (EGFR), insulin-like growth factor (IGF), AMPK/mTOR, and mitogen-activated protein kinases (MAPKs), among others [32]. Focusing on autophagy, the AMPK/mTOR signaling pathway plays an essential role in autophagy regulation, cell growth, cell proliferation, and metabolism. Several studies have shown that this signaling pathway participates in the process of cardiovascular diseases [32]. On the other hand, the silent information regulator 1 (Sirt1) also exerts a cardioprotective effect in various cardiovascular diseases via multiple cell activities [33].
Previous evidence shows that cells cope with environmental stress by targeting abundant transcription factors, such as the Forkhead box O (FoxO) proteins, which play a critical role in the cardiac pathologies associated with aging by regulating both metabolic and proliferative events in endothelial cells [34]. Indeed, a study by Hariharan et al. in 2010 demonstrated that Sirt1 mediates glucose starvation-induced autophagy by deacetylating FoxO in cardiomyocytes [35]. Additionally, Sirt1 overexpression protects the heart from ischemia-reperfusion injury by inhibiting proapoptotic molecules, demonstrating that Sirt1 is involved in cardio protection.
Next, we highlight some natural compounds known to induce autophagy and that have been reported to offer beneficial effects for age-related heart diseases.

2.1. Curcumin

Curcumin (1,7-bis-(4-hydroxy-3-methoxyphenyl) is the main biocompound that comes from the spice turmeric, and it has been widely used in Indian and Chinese traditional medicine because of its diverse biological benefits, such as anti-inflammatory, antioxidant, and anticancer properties [36,37,38,39]. Research has also highlighted curcumin’s ability to induce autophagy, particularly in cancer cells, where autophagy is a precursor to apoptosis [40,41,42]. This process is mediated through the AMPK signaling pathway and the activation of transcription factor EB (TFEB), both of which positively regulate autophagy [43,44]. Research on the effect of curcumin on autophagy has explored its effect on lifespan in models like C. elegans, D. melanogaster, yeast, and even mice, showing influence on pathways like mTOR, PKA, and FoxO [45,46,47,48]. Furthermore, studies made on various diseases (chronic kidney disease and insulin resistance in non-alcoholic fatty liver disease), demonstrated that the administration of this compound increased the ATP levels and inhibited their depletion and ameliorated mitochondrial dysfunction [49,50,51].
Evidence has highlighted curcumin as a molecule that activates autophagic flux and attenuates oxidative damage via AKT/mTOR pathway regulation, as shown by research in diabetic rats with myocardial damage, where curcumin treatment in combination with high intensity interval training (HIIT) demonstrated improvement in the expression of important autophagic proteins such as ATG-5 and Beclin-1 [52]. In mice with doxorubicin-induced cardiomyopathy, where curcumin attenuated oxidative stress and inflammation, curcumin also restored the alterations in autophagy, as shown by increased levels of Beclin-1 and of the LC3II/LC3I ratio and by activation of the AKT/mTOR pathway [53]. More evidence extrapolates the benefits of curcumin on reducing senescent cardiomyocytes via AMPK/mTOR in a dose-dependent manner, which suggests the role of curcumin as an anti-cardiac aging molecule (Figure 2) [54]. All of this evidence demonstrates the advantages of curcumin administration on cardiac homeostasis via autophagy activation. It is worth noting that the Food and Drug Administration (FDA) has not approved the use of curcumin for any medical condition; however, it is available in the United States as a dietary supplement [55].

2.2. Resveratrol

Resveratrol (3,5,4-trihydroxystilbene) is a natural polyphenol produced by various plants and fruits in response to external stimuli. It is commonly found in peanuts, berries, grapes, cranberries, and other products, such as red wine [56]. In the last decades, resveratrol has attracted much attention due to its beneficial properties. It has been shown to decrease oxidative stress through upregulation of antioxidant enzymes like catalase (CAT) (EC 1.11.1.6) and superoxide dismutase (SOD) (EC 1.15.1.1) [57,58]. Moreover, its anti-inflammatory effects occur through the modulation of Sirt1, which in turn inhibits the NF-κB/STAT pathway, thereby reducing the production of inflammatory molecules [59]. Additionally, resveratrol modulates diverse pathways that converge in cancer, such as Raf/MAK, ERK/p-38, and PI3K-AKT, among others [60].
Much evidence about resveratrol has pointed to a direct relationship with autophagy induction via inhibition of the mTOR-ULK1 pathway and stimulation of LC3-II production [61,62]. Moreover, the effects of resveratrol were studied in mitochondrial dysfunction and oxidative stress, and the results showed that a resveratrol treatment increased citrate synthase activity, basal oxygen consumption, and mitochondrial ATP production [63]. In line with this, resveratrol has been demonstrated to be a good candidate to fight ARDs.
In recent years, natural compounds with cardioprotective properties have been widely studied [64,65], and resveratrol is no exception. In studies in vivo on diabetic rats with cardiomyopathy and in vitro assays using neonatal rat ventricular myocytes, resveratrol has shown to negatively regulate the PI3K/Akt pathway in order to stimulate autophagic flux, decreasing apoptosis and ameliorating cardiac dysfunction [66]. Nevertheless, the effects of resveratrol also extrapolate to other pathways that converge in autophagy activation. For example, through the activation of the master autophagy regulator TFEB, resveratrol lessens endothelial oxidative damage of cardiovascular models [67]. Additionally, the activation of Sirt1 directly helps to increase the expression of autophagic molecules such as LC3 and Beclin-1 (Figure 2) [68]. This evidence suggests that the use of resveratrol could help to improve health in cardiomyopathies.

2.3. Quercetin

Quercetin (2-(3,4-dihydroxyphenyl)-5,7-dihydroxy-4H-1-benzopyran-4-one) is one of the most common flavonoids present in fruits like grapes, berries, cherries and other citric fruits, in vegetables like onions and broccoli, and in seeds and other plants [69,70]. Antioxidant and anti-inflammatory properties are the most studied in quercetin [71]. Research has also highlighted its potential as an anti-diabetic and anti-carcinogenic agent [72,73,74,75]. Interestingly, quercetin has also been shown to stimulate autophagy via signaling pathways that converge in the inhibition of mTOR through the activation of AMPK and Sirt1, ultimately leading to an increase in the LC3-II/LC3-I ratio [76,77]. Additionally, quercetin was used to treat lipopolysaccharide-induced oxidative damage, and the results showed that quercetin improved ATP levels, leading to a beneficial effect in mitochondrial function [78].
In recent years, quercetin has attracted attention to cardiovascular diseases due to its demonstrated effect on autophagic stimulation. Specifically, quercetin treatment has been shown to ameliorate myocardial injury in diabetic rats through the activation of the AMPK pathway, leading to increased LC3 and Beclin-1 protein levels and amelioration of oxidative stress [79]. Furthermore, quercetin has been shown to suppress the PI3K/Akt pathway that results in autophagy activation and attenuation of myocardial ischemia in Sprague-Dawley rats [80]. Also, quercetin has been shown to promote autophagy by increasing the LC3 I/II ratio, improving endothelial function over time and decreasing oxidative stress to attenuate hypertension in Wistar rats [81]. Evidence has shown that quercetin prevents myocardial fibrosis via the modulation of miR-223-3p/FOXO3, which promotes the regulation of the autophagic effector enzyme ATG7 that, in turn, increases LC3 levels. This research was performed on cardiac tissues of patients with atrial fibrillation and in aged rats [82].
All of these research findings highlight the beneficial autophagic effects of quercetin’s function as a promising adjuvant to prevent and treat age-related cardiopathies (Figure 2).

2.4. Polyamines

Polyamines (PAs) are small aliphatic molecules containing amino groups throughout their structure. They can be found in a wide range of foods, including fruits and vegetables with a higher content of polyamines, such as mushrooms, green peppers, green peas, and citrus fruits [83]. At physiological pH, these amino groups become protonated and acquire the capacity to interact with negatively charged biomolecules such as nucleic acids, proteins, and phospholipids [84,85,86]. The principal PAs in mammalian cells are putrescine, spermidine, and spermine; they are interconverted through a stepwise process, typically starting with arginine [87]. Deregulation of PAs, in particular spermidine and spermine, is related to cellular senescence, impaired proteostasis, dysregulated nutrient sensing, dysfunctional mitochondria, and dysfunctional autophagy [88]. Spermidine is mechanically related to autophagy activation through the posttranslational modification of the eIF5A, which, in turn, promotes the translation and synthesis of TFEB, a master autophagy regulator. Spermidine also regulates cell viability by significantly increasing cell metabolic activity and ATP production in a mitophagy deficit induced by disease-associated tau protein and cardiac aging [89,90].
Moreover, spermidine inhibits the histone acetyltransferase P300 (EP300) to promote the activation of various autophagy-related genes [91]. Additionally, spermidine inhibits Beclin-1 degradation to promote autophagy initiation [92].
Spermidine and spermine have been shown to exert cardioprotective effects on aged rats by preventing myocardial fibrosis and morphological myocardial alterations [93]; similar effects were also reported in aged mice where spermidine stimulated autophagy [31]. Furthermore, spermidine limits atherosclerotic lipidic plaque formation by inducing autophagy, as shown by the increase in the LC3 II/I ratio of mouse aortic vascular smooth muscle cells [94]. Additionally, spermidine has been demonstrated to ameliorate myocardial injury and cardiac dysfunction in mice by promoting autophagy mediated by the AMPK/mTOR pathway (Figure 2) [95]. All of this evidence makes spermidine a good candidate to prevent and ameliorate cardiovascular diseases.

3. Age-Related Neurodegenerative Diseases: Alzheimer’s and Parkinson’s

Aging is the primary risk factor for most neurodegenerative diseases, including Alzheimer’s disease (AD) and Parkinson’s disease (PD) [96]. One in ten individuals aged ≥65 years has AD, and its prevalence increases with age. Meanwhile, more than 10 million people worldwide are living with PD; additionally, older men are 1.5 times more likely to have PD than women. In both diseases, pathogenesis presents similar features, such as oxidative stress, mitochondrial dysfunction, and protein aggregation, all of which are events related to autophagy [96,97,98].
Autophagy is an essential process in preserving the homeostatic requirements of post-mitotic neurons [97]; it cooperates with the ubiquitin-proteasome system (UPS) to degrade toxic proteins, and its deficiency causes the accumulation of ubiquitinated proteins, axon dystrophy, abnormal synaptic transmission, and subsequent degeneration [99]. Furthermore, autophagic responses extinguish neuroinflammatory reactions, which directly contribute to the pathogenesis of PD and AD [30]. Moreover, autophagy is associated with other signaling pathways such as AMPK, mTOR, and Sirt1. Human studies suggest that the mTOR signaling pathway is involved in AD, as it is inhibited in the cortex and hippocampus of adult mice with the disease [100,101]. In PD, the accumulation of alpha-synuclein is known to play a key role in the death of dopamine-producing neurons. Therefore, enhancing autophagy, particularly through the mTOR signaling pathway, may help prevent the toxic accumulation of alpha-synuclein [102]. Autophagy-enhancing effects have been linked to Sirt1, which aids in the degradation of Aβ and promotes synaptic formation and synaptic activity. Therefore, Sirt1 can reduce the progression of various degenerative diseases [103,104]. Sirt1 plays an essential role in Aβ and tau metabolism through the deacetylation of site-specific lysine residues of histone and non-histone proteins [104]. As shown, the inhibition of Sirt1, or its decline in enzymatic activity observed with age, results in a reduction in the deacetylation of tau. This reduction might block tau polyubiquitination and tau proteasomal degradation, leading to p-tau accumulation. Consequently, this accumulation can result in the formation of neurofibrillary tangles (NFTs) [104]. Also, genetic variation in Sirt1 is linked with the development of PD along with genetic mutations in PD, which have been reported to alter the expression of the Sirt1 promoter with a subsequent reduction of the neuroprotective role of Sirt1 [103].

3.1. Curcumin

Regarding age-related neurodegenerative diseases, evidence indicates that oral intake of curcumin has beneficial effects on conditions like AD by preventing short-term memory deterioration [105]. Moreover, curcumin has been shown to reverse cognitive disorders in an AD mice model induced by cisplatin. This effect is achieved through the activation of the AMPK-JNK signaling pathway to inhibit mTOR and Bcl-2 that, in turn, modulate autophagy as demonstrated by the overexpression of LC3 and downregulation of p62 [106]. The beneficial effects of curcumin on autophagy extend to PD. In mouse models, curcumin treatment led to an increased expression of LC3-II, facilitating enhanced clearance of α-synuclein proteins. Notably, this was accompanied by improvements in mitochondrial function (Figure 2) [107]. Collectively, these findings underscore curcumin’s potential as a therapeutic candidate for various neurodegenerative diseases.

3.2. Resveratrol

Among neurodegenerative diseases, the role of resveratrol has been widely studied in AD due to its effect on pathways that converge on autophagy activation [108]. New research on animal models has shown that resveratrol enhances cognitive function in rats by stimulating the AKT/mTOR pathway [109]. Moreover, in Wistar rats with AD induced by colchicine, the combination of resveratrol and donepezil significantly increased the number of astrocytes and decreased microglia. This suggests that activating the Sirt1 pathway enhances mitophagy, leading to improved neuroprotection [110]. In the Drosophila model that overexpresses the amyloid precursor protein (APP) for a similar AD phenotype, resveratrol treatment showed improvements in sleep and memory that were dependent on the ortholog Sir2, a sirtuin ortholog that in turn stimulates autophagy by activating HSP70 and ATG4, also interacting to LC3 and p62 [111,112]. Meanwhile, a randomized trial of people affected by AD who orally consumed 500 mg of resveratrol once a day reported good tolerability. Notably, its principal metabolites can penetrate the blood-brain barrier, implying potential beneficial effects on the central nervous system (CNS) [113]. Benefits of resveratrol have also been described in PD; studies in mice models showed that resveratrol ameliorates the deficit in cognitive and motor function in a dose-dependent manner; moreover, the effects of resveratrol also demonstrated a decrease in α-synuclein aggregation [114]. A study using state-of-the-art hydrogen/deuterium exchange mass spectrometry technology demonstrated that resveratrol interferes with amyloid aggregation [115]. The effects of autophagy and resveratrol in PD were demonstrated in a 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced mouse model of the disease, where LC3 promoted autophagic degradation of α-synuclein via the activation of the Sirt1 pathway (Figure 2) [116]. More recently, in two cell models of PD (SH-Sy5Y and SK-N-SH cell lines treated with MPP+ (1-Methyl-4-phenylpyridium)), resveratrol promoted autophagy as shown by the increase of the protein expression of Beclin-1 and LC3-II/LC3-I ratio, as well as the reduction in p62 levels; interestingly, the effects of resveratrol also reduced the expression of the lncRNA SNHG1 (small nucleolar RNA host gene 1), which positively regulates the SNCA gene that encodes for α-synuclein [117].
All of this evidence suggests that resveratrol is a promising natural compound with significant properties for the clearance of amyloid aggregates through autophagy stimulation.

3.3. Quercetin

Quercetin holds promise as a mitigator for neurodegenerative diseases. It activates Sirt1, a crucial regulator that influences the expression of key genes like FOXO1 and NRF2. These genes play interconnected roles in the autophagy pathway, suggesting quercetin’s potential to support neurological health through these regulatory mechanisms [118,119].
Recent research into the therapeutic potential of quercetin for AD has unveiled a whole network of molecular targets, with alpha serine/threonine-protein kinase-1 (AKT1) prominently featured (Figure 2). AKT1 is known to interact with Atg7, a critical enzyme that regulates the Atg proteins necessary to initiate autophagy [120,121]. Additionally, other research has shown that quercetin reduces the expression of Abelson kinase (Abl kinase), a significant molecule implicated in Alzheimer’s disease pathogenesis. By decreasing the expression of Abl kinase, quercetin helps to prevent tau phosphorylation and neuronal apoptosis triggered by Aβ, primarily through activation of the autophagy process [122,123]. Quercetin modified with Palladium nanomaterial (PdNPs) coated on polysorbate 80 has been shown to enhance autophagy and facilitate the clearance of Aβ in SH-SY5Y cells, a model for AD [124]. Furthermore, research on a PC12 cell model of AD has indicated that quercetin treatment improves cell survival and proliferation, mitigates Aβ toxicity, and enhances the expression of Sirt1 [125,126]. Moreover, evidence has shown that quercetin ameliorates neuronal death, mitochondrial dysfunction, and α-synuclein accumulation in animal and diverse cell models of PD by activating autophagy [127]. This effect is demonstrated by increased levels of ULK1(Ser555)/ULK1, p-TBK1(Ser172)/TBK1, and LC3 [128]. Furthermore, in a rotenone-induced rat model, quercetin treatment not only increased autophagy but also attenuated behavioral impairments [129].

3.4. Polyamines

Polyamines are crucial for central nervous system (CNS) function; among their different roles, they modulate N-methyl-D-aspartate (NMDA) receptors that, in turn, regulate diverse neurological functions, including memory [130,131]. Additionally, they promote the regulation of glutamate signaling, which also enhances excitability and memory [130].
It has long been known that PA levels are disrupted in the human brain with AD [132]; in fact, other research on mice models of the disease have demonstrated altered polyamine levels, as shown by increased anabolism and upregulation of spermidine synthase (EC 2.5.1.16) and elevated acetylspermidine levels, suggesting a breakdown issue [133]. Notably, the distinction between these acetylated polyamine forms and biogenic polyamines has a differential impact on tau protein accumulation [90].
Research suggests that oral supplementation of spermidine and spermine promotes autophagy in the hippocampus, thereby delaying brain aging and maintaining cognitive function in mice [134]. Additionally, evidence has shown the benefits of spermidine treatment on mitochondrial impairment through autophagy activation, as seen by elevated LC3 and Beclin levels compared to the control in SY5Y cells stably expressing a mutant form of human tau protein [90]. Research using high-resolution imaging demonstrated that spermidine could induce autophagy and clear amyloid precursor protein clusters, a key pathological feature of Alzheimer’s disease [135]. These findings suggest that spermidine, by promoting autophagy and restoring polyamine homeostasis, may hold promise as a therapeutic strategy for Alzheimer’s disease.

4. Age-Related Ocular Diseases: Cataracts

Visual impairments are among the leading diseases in developed countries, and aging is the primary cause of their clinical manifestation [30]. Cataracts are the most common cause of visual impairment in the elderly [136], and it is estimated that from 2000 to 2020, 43.3 million people went blind due to cataracts and 295 million had moderate to severe vision impairment (MSVI) due to this condition [137].
Age-related cataracts form in the center of the lens, which stiffens as new layers of cortical fibers continue to proliferate. In this condition, autophagy plays a crucial role in maintaining cell integrity and preserving the lens’s physical properties by ensuring cellular homeostasis and the absence of fiber accumulation [138,139,140]. Additionally, the autophagic role in the eye is highlighted by the high expression of autophagy-related proteins in various eye structures, including the cornea, lens, retina, and orbit. This protein expression correlates with significant autophagic activity observed particularly within the lens [138,141].
Studies on mice revealed the impact of altered autophagic flux mediated by TBC1D20, a key regulator of autophagosome maturation. The mice exhibited cataracts in the lens nucleus, along with highly disorganized, degenerated lens fibers and an accumulation of ubiquitin aggregates. These findings strongly suggest that autophagy is not only crucial for maintaining intracellular homeostasis in the lens but also that the accumulation of residual material contributes to cataract formation [138,142]. Multiple signaling molecules and pathways, such as mTOR and AMPK, play significant roles in the development of cataracts. For instance, the mTOR pathway influences cataract development by modulating various cellular processes, such as cell growth, autophagy, and epithelial-mesenchymal transition.
Similarly, the AMPK pathway is central in the pathogenesis of ocular diseases, affecting various ocular tissues. Studies have linked age-related inactivation of AMPK and autophagy to lens epithelial cell senescence and age-related cataracts [143,144]. Furthermore, Sirt1 has been detected in multiple eye structures in both mice and humans, including the cornea, lens, ciliary body, retinal pigment epithelium (RPE), neuroretina, and melanocyte. Numerous studies have also linked Sirt1 dysfunction to ocular diseases, including cataracts and age-associated macular degeneration [145]. Notably, Sirt1 levels in the lens were significantly decreased in individuals aged ≥51 years, further highlighting its potential role in cataract formation due to its widespread distribution in different eye cell types.

4.1. Curcumin

Curcumin has been demonstrated to have protective effects against cataract development and its progression in numerous in vitro and in vivo cataract models. In an in vitro study, it was shown that curcumin inhibited a pleiotropic oxidative stress response in cultured lens epithelial cells (hLECs) [146]. In another study performed in rats, curcumin suppressed selenium-induced oxidative stress in the rats’ lenses and delayed the formation of cataracts by inhibiting non-enzymatic antioxidant depletion [146,147]. The above suggests that the anti-cataract effect of curcumin may originate from its antioxidant properties.
As it concerns autophagy, a study performed in RPE cells demonstrated that curcumin, compared with rapamycin, exerts a similar amount of neuroprotection [148]. Recent research on rats with glucose-induced cataracts highlights the potential significance of tetrahydrocurcumin (THC), a principal metabolite of curcumin, and a synthetic curcumin analog (C1) in promoting lysosomal biogenesis and activating TFEB, consequently mitigating cataract formation [149]. These studies offer a new perspective on age-related cataracts, suggesting that curcumin may safely stimulate autophagy and could be a promising treatment option.

4.2. Resveratrol

While much of the research has primarily emphasized the anti-inflammatory advantages of resveratrol in addressing various age-related eye conditions, its autophagic properties have also shown promise in treating other ocular diseases, including age-related macular degeneration (AMD) [150], diabetic retinopathy [151], glaucoma [152], and cataracts. In fact, resveratrol emerges as a good candidate in the prevention and treatment of cataracts. Evidence indicates that resveratrol promotes the phosphorylation of p38, which in turn mitigates oxidative damage induced by high levels of glucose in human lens epithelial cells (HLEC) by triggering autophagy, as evidenced by the increased protein levels of LC3 and Beclin-1 (Figure 2) [153]. These autophagic effects have also been observed in the ARPE-19 cell line [154]. Also, research has shown that resveratrol and three of its main metabolites (resveratrol-3-O-glucuronide, resveratrol-4′-O-glucuronide, and resveratrol-3-O-sulfate) can be measured in different eye tissues, such as the aqueous humor, vitreous humor, and conjunctiva, suggesting the feasibility of using dosage-effective resveratrol treatment for ocular diseases [155].
This evidence supports the idea that autophagic stimulation by resveratrol could be applied to treat age-related ocular diseases, including cataracts.

4.3. Quercetin

The effects of quercetin on autophagy in ocular diseases are still being explored, especially in cataracts. However, new research has demonstrated that quercetin activates the Sirt1/AMK signaling pathway, a positive regulator of autophagy (Figure 2) [77]. Additionally, research has shown that quercetin decreases oxidative species by promoting autophagy through the NRF2-PGC-1α-Sirt1 pathway in mice with macular degeneration [156].
Quercetin and its metabolites can protect the lens from the internal flow of calcium and sodium, thereby maintaining the lens’s ion balance and protein levels, crucial for lens transparency [157]. Moreover, quercetin stimulates the hypoxia-inducible factor-1 (HIF1) pathway in lens epithelial cells, activating and synthesizing downstream effectors and thereby potentially delaying cataract progression [157].
Extensive in vitro and in vivo studies in animal models have consistently demonstrated the lens-protective effects of quercetin’s antioxidant and chelating properties [157,158,159]. In a rat model of cataracts induced by sodium selenite, the antioxidant properties of quercetin significantly reduced crystal turbidity and prevented the formation of cataracts caused by selenite [160]. It also bolstered the antioxidant mechanism of the lens, safeguarding its integrity. Furthermore, quercetin has shown promising results in protecting against cataracts induced by H2O2 (causing lens opacification) and retinal lesions induced by diabetes [161]. This evidence strongly suggests quercetin as a potential therapeutic target for cataracts.
It is also important to highlight quercetin as an alternative in treating other specific ocular diseases due to its proven antioxidant, anti-inflammatory, antifibrosis, and pro-autophagic properties. Its antioxidant effects could significantly impact the management of conditions besides cataracts, such as dry eye disease and retinopathy, among others [157].

4.4. Polyamines

Although direct studies on the autophagic role of spermidine in age-related cataracts have not been conducted, there is indirect evidence regarding its positive regulation of Sirt1 [88,162]. This regulatory role of spermidine should be considered crucial in the development of the disease because the downregulation of Sirt1 in age-related cataracts has been described in research [163,164].
It is also important to mention that there is evidence on the anti-aging and protective properties of PA in treating ocular diseases [165], particularly in cataracts. Research indicates that PAs are detectable in intraocular fluid [166], and a decrease in its levels is linked to the development of age-related cataracts [167]. Furthermore, studies in mice and rabbits with cataracts have shown a remarkable decrease in the level of spermidine [168,169]. This decrease is likely caused by increased catabolism activity and transglutaminase type 2 (EC: 2.3.2.13), which catalyzes lens crystallin cross-linking. Furthermore, studies show that supplementing with spermidine can delay lens opacification by replenishing spermidine levels and reducing lens crystallin cross-linking, which maintains lens transparency, suggesting its potential as a treatment for age-related cataracts [169,170].
Even though information about PA is limited for cataracts, these new findings hold promise for improving eye health, preventing vision loss, and potentially restoring sight, ultimately leading to a better quality of life.
In Table 1, we provide a summary of the benefits associated with the natural compounds previously discussed in relation to ARDs.

5. Natural Autophagy Activators in Other ARDs

The benefits of natural compounds that activate autophagy discussed in this review also extend to other ARDs such as osteoarthritis, sarcopenia, and type II diabetes (Table 2). We have described the involvement of key autophagic regulators, such as the Akt/mTOR, AMPK, FOXO3a and Sirt1, as the main effectors in enhancing the autophagic flux and ameliorating the consequences of these ARDs. These findings highlight the potential systemic health benefits that would bring new hope for ARDs.
Further research into the effects of natural compounds that stimulate autophagy could bring hope for better and healthier aging in fighting the associated diseases.

6. Conclusions

Age-related diseases such as cardiovascular, neurodegenerative, and ocular diseases represent a global social, economic, and health challenge [2]. Although the molecular mechanisms involved in aging and related diseases are not yet fully understood, it is known that processes like autophagy play a crucial role in the development of these pathologies. In fact, a decrease in the protein levels of key molecules in the autophagic flow, such as LC3 I/II, is one of their distinctive features [184]. As a result, various efforts have been made to propose new therapeutic targets that promote autophagic flow to restore cellular homeostasis in age-related diseases.
Our work focuses on compiling the most recent evidence regarding curcumin, resveratrol, polyamines, and quercetin as natural autophagy regulators for the treatment of age-related diseases. Natural molecular targets are of great interest due to their nutritional value and historical role as precursors in the development of new drugs. Molecules such as curcumin, quercetin, resveratrol, and some polyamines have been shown to enhance autophagy in both cellular and animal models [185]. Our work gathers recent evidence on the effects of administering these natural compounds that promote the stimulation of autophagy regulatory pathways, such as mTOR, FOXO1/3, AMPK, and Sirt1, leading to increased levels of key proteins such as Beclin-1 and LC3, as well as a decrease in p62. These effects of natural compounds have benefits on cardiovascular diseases, neurodegeneration, and cataracts [186]. Furthermore, other natural compounds like berberine have been shown to enhance autophagy AMPK and Sirt1 activation. Additionally, berberine is reported to extend lifespan and offer potential therapeutic benefits for ARDs such as sarcopenia and cardiovascular and neurodegenerative disorders [187,188,189,190,191,192].
To further explore the potential use of natural compounds as an adjuvant therapeutic strategy for ARDs, and given their better tolerability compared to other drugs, more research on long-term dosing is essential. For example, while curcumin and quercetin have been approved by the Food and Drug Administration (FDA) as “Generally Recognized As Safe” (GRS) [193,194], in clinical trials they have shown significant variability regarding dosage across different countries [195,196]. In contrast, evidence about resveratrol (8 mg/day) and spermidine (1.2 mg/day) did not seem to have major side effects in one-year and three-month clinical trials, respectively [197,198].
The combination of natural compounds with autophagy-enhancing drugs like rapamycin may be a promising strategy to fight ARDs due to their potential synergistic effects in promoting autophagy. However, there is currently insufficient evidence regarding these effects on ARDs. Most studies have primarily focused on anticancer therapies, where they have reported mechanisms of cell death via apoptosis or various forms of autophagy-related cell death [199,200,201,202]. However, natural compounds that stimulate autophagy also provide additional benefits, including anti-inflammatory and antioxidant properties, as well as improvements in mitochondrial dysfunction [59,203,204,205]. These cellular processes are closely linked to autophagy.
However, it is important to highlight that one of the main disadvantages of using natural compounds is their low bioavailability; thus, their effects may be mild. To address this, various efforts based on nanotechnology have been made to improve the bioavailability of these compounds. Other important factors to consider include the potential interactions with other drugs or supplements that could be harmful to health. Additionally, more experimental evidence is needed to support and confirm the benefits of curcumin, resveratrol, polyamines, and quercetin in age-related diseases.

Author Contributions

Conceptualization, G.E.J.G. and J.C.M.; investigation, V.M.M.R., J.R.T.J. and N.M.M.M.; resources, J.C.M., J.J.M. and N.L.G.; writing—original draft preparation, J.J.M., N.L.G. and N.M.M.M.; writing—review and editing, G.E.J.G., J.C.M., V.M.M.R., J.R.T.J., J.J.M., N.L.G. and N.M.M.M.; visualization, J.J.M., G.E.J.G. and J.C.M.; supervision, G.E.J.G. and J.C.M.; funding acquisition, J.C.M. All authors have read and agreed to the published version of the manuscript.

Funding

The APC was funded by COFAA-Instituto Politécnico Nacional.

Informed Consent Statement

Not applicable.

Conflicts of Interest

The authors declare no conflicts of interest.

Glossary

AbbreviationMeaning
Abl Abelson kinase
AD Alzheimer’s disease
AKT Alpha serine-threonine protein kinase
AMBRA1 Activating molecule in Beclin1-regulated autophagy protein 1
AMD Age-related macular degeneration
AMPK 5′-adenosine monophosphate (AMP)-activated protein kinase
APP Amyloid precursor protein
ARDs Age-related diseases
Atg Agy-related proteins
Amyloid-β
Bcl-2 B-cell lymphoma 2 protein
BECN1 Beclin 1 gene
C1 curcumin analog
CAT Catalase
CNS Central nervous system
CXCL16 CXC motif chemokine ligand 16
EGFR Epidermal growth factor
eIF5A Initial Translate factor 5A Eukaryotic
EP300 Histone acetyltransferase P300
ERK Extracellular signal-regulated kinase
FDA Food and Drug Administration
FoxO Forkhead box O proteins
HIIT High intensity interval training
HLECs Cultured lens epithelial cells
HSP70 70-kDa heat shock proteins
IGF Insulin-like growth factor
JAK/STAT Janus kinase-signal transducers and activators of transcription
JNK c-jun-N-terminal kinase
LC3 Microtubule-associated protein 1A/1B-light chain 3 (LC3)
LV Left ventricular
MAPK Mitogen-activated protein kinase
MPP 1-methyl-4-phenylpyridium
MPTP 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine
MSVI Severe vision impairment
mTOR Mammalian target of rapamycin
NF-kB Nuclear factor κB
NFTs Neurofibrillary tangles
NMDA N-methyl-D-aspartate
NRF2 Nuclear factor erythroid 2-related factor 2
Ox-LDL Oxidized Low-Density lipoprotein
P62/SQSTM1 Sequestosome 1 (SQSTM1)/p62
PAs Polyamines
PD Parkinson´s disease
PdNPs Palladium nanomaterial
PI3K Phosphoinositide 3-kinase
PINK1 PTEN-induced putative kinase 1
PKA Protein kinase A
p-TBK1 phospho-TANK-binding kinase 1
RPE Retinal pigment epithelium
Sirt1 Silent information regulator 1
SNAP29 synaptosome associated protein 29
SNARE Soluble N-ethylmaleimide-sensitive factor attachment protein receptor
SNCA Synuclein alpha gene
SNHG1 small nucleolar RNA host gene 1
SOD Superoxide dismutase
STX Syntaxin 17
TBC1D20 TBC1 domain family, member 20
TFEB Transcriptional Factor EB
THC Tetrahydrocurcumin
TRAP Translocon-associated protein
ULK Ubiquitin-like-kinase
UPS Ubiquitin-proteasome system
UVRAG UV Radiation Resistance Associated
VAMP8 Vesicle associated membrane protein 8
VPS Vacuolar protein sorting

References

  1. United Nations Department of Economic; Social Affairs. World Social Report 2023; United Nations: New York, NY, USA, 2023. [Google Scholar]
  2. Chang, A.Y.; Skirbekk, V.F.; Tyrovolas, S.; Kassebaum, N.J.; Dieleman, J.L. Measuring population ageing: An analysis of the Global Burden of Disease Study 2017. Lancet Public Health 2019, 4, e159–e167. [Google Scholar] [CrossRef] [PubMed]
  3. Ahmed, T.; Zulfiqar, A.; Ishaq, S. Age-Related Diseases. In Nutrients and Nutraceuticals for Active & Healthy Ageing; Nabavi, S.M., D’Onofrio, G., Nabavi, S.F., Eds.; Springer: Singapore, 2020; pp. 27–51. [Google Scholar]
  4. Li, Z.; Zhang, Z.; Ren, Y.; Wang, Y.; Fang, J.; Yue, H.; Ma, S.; Guan, F. Aging and age-related diseases: From mechanisms to therapeutic strategies. Biogerontology 2021, 22, 165–187. [Google Scholar] [CrossRef] [PubMed]
  5. Di Ciaula, A.; Portincasa, P. The environment as a determinant of successful aging or frailty. Mech. Ageing Dev. 2020, 188, 111244. [Google Scholar] [CrossRef] [PubMed]
  6. Hurley, J.H.; Young, L.N. Mechanisms of Autophagy Initiation. Annu. Rev. Biochem. 2017, 86, 225–244. [Google Scholar] [CrossRef]
  7. Baba, M.; Takeshige, K.; Baba, N.; Ohsumi, Y. Ultrastructural analysis of the autophagic process in yeast: Detection of autophagosomes and their characterization. J. Cell Biol. 1994, 124, 903–913. [Google Scholar] [CrossRef]
  8. Chen, T.; Tu, S.; Ding, L.; Jin, M.; Chen, H.; Zhou, H. The role of autophagy in viral infections. J. Biomed. Sci. 2023, 30, 5. [Google Scholar] [CrossRef]
  9. Mizushima, N.; Komatsu, M. Autophagy: Renovation of cells and tissues. Cell 2011, 147, 728–741. [Google Scholar] [CrossRef]
  10. Yang, Y.; Pan, Z.; Sun, J.; Welch, J.; Klionsky, D.J. Autophagy and machine learning: Unanswered questions. Biochim. Biophys. Acta Mol. Basis Dis. 2024, 1870, 167263. [Google Scholar] [CrossRef]
  11. Wang, L.; Klionsky, D.J.; Shen, H.M. The emerging mechanisms and functions of microautophagy. Nat. Rev. Mol. Cell Biol. 2023, 24, 186–203. [Google Scholar] [CrossRef]
  12. Dice, J.F. Altered degradation of proteins microinjected into senescent human fibroblasts. J. Biol. Chem. 1982, 257, 14624–14627. [Google Scholar] [CrossRef]
  13. Valdor, R.; Martinez-Vicente, M. The Role of Chaperone-Mediated Autophagy in Tissue Homeostasis and Disease Pathogenesis. Biomedicines 2024, 12, 257. [Google Scholar] [CrossRef] [PubMed]
  14. Mulcahy Levy, J.M.; Thorburn, A. Autophagy in cancer: Moving from understanding mechanism to improving therapy responses in patients. Cell Death Differ. 2020, 27, 843–857. [Google Scholar] [CrossRef] [PubMed]
  15. Axe, E.L.; Walker, S.A.; Manifava, M.; Chandra, P.; Roderick, H.L.; Habermann, A.; Griffiths, G.; Ktistakis, N.T. Autophagosome formation from membrane compartments enriched in phosphatidylinositol 3-phosphate and dynamically connected to the endoplasmic reticulum. J. Cell Biol. 2008, 182, 685–701. [Google Scholar] [CrossRef] [PubMed]
  16. Parzych, K.R.; Klionsky, D.J. An overview of autophagy: Morphology, mechanism, and regulation. Antioxid Redox Signal. 2014, 20, 460–473. [Google Scholar] [CrossRef]
  17. Pyo, J.O.; Nah, J.; Jung, Y.K. Molecules and their functions in autophagy. Exp. Mol. Med. 2012, 44, 73–80. [Google Scholar] [CrossRef]
  18. Pankiv, S.; Clausen, T.H.; Lamark, T.; Brech, A.; Bruun, J.A.; Outzen, H.; Øvervatn, A.; Bjørkøy, G.; Johansen, T. p62/SQSTM1 binds directly to Atg8/LC3 to facilitate degradation of ubiquitinated protein aggregates by autophagy. J. Biol. Chem. 2007, 282, 24131–24145. [Google Scholar] [CrossRef]
  19. Fujita, N.; Itoh, T.; Omori, H.; Fukuda, M.; Noda, T.; Yoshimori, T. The Atg16L complex specifies the site of LC3 lipidation for membrane biogenesis in autophagy. Mol. Biol. Cell 2008, 19, 2092–2100. [Google Scholar] [CrossRef]
  20. Fahmy, A.M.; Labonté, P. The autophagy elongation complex (ATG5-12/16L1) positively regulates HCV replication and is required for wild-type membranous web formation. Sci. Rep. 2017, 7, 40351. [Google Scholar] [CrossRef]
  21. Ke, P.Y. Molecular Mechanism of Autophagosome-Lysosome Fusion in Mammalian Cells. Cells 2024, 13, 500. [Google Scholar] [CrossRef]
  22. Zhen, Y.; Stenmark, H. A dual-purpose fusion complex in autophagy. Cell Res. 2024, 34, 183–184. [Google Scholar] [CrossRef]
  23. Mizushima, N. Autophagy: Process and function. Genes Dev. 2007, 21, 2861–2873. [Google Scholar] [CrossRef] [PubMed]
  24. Ortega, M.A.; Fraile-Martinez, O.; de Leon-Oliva, D.; Boaru, D.L.; Lopez-Gonzalez, L.; García-Montero, C.; Alvarez-Mon, M.A.; Guijarro, L.G.; Torres-Carranza, D.; Saez, M.A.; et al. Autophagy in Its (Proper) Context: Molecular Basis, Biological Relevance, Pharmacological Modulation, and Lifestyle Medicine. Int. J. Biol. Sci. 2024, 20, 2532–2554. [Google Scholar] [CrossRef] [PubMed]
  25. He, C.; Klionsky, D.J. Regulation mechanisms and signaling pathways of autophagy. Annu. Rev. Genet. 2009, 43, 67–93. [Google Scholar] [CrossRef] [PubMed]
  26. López-Otín, C.; Blasco, M.A.; Partridge, L.; Serrano, M.; Kroemer, G. Hallmarks of aging: An expanding universe. Cell 2023, 186, 243–278. [Google Scholar] [CrossRef]
  27. Hipp, M.S.; Kasturi, P.; Hartl, F.U. The proteostasis network and its decline in ageing. Nat. Rev. Mol. Cell Biol. 2019, 20, 421–435. [Google Scholar] [CrossRef]
  28. Kumar, P.; Choudhary, A.; Kinger, S.; Jagtap, Y.A.; Dubey, A.R.; Gutti, R.K.; Chitkara, D.; Suresh, A.K.; Mishra, A. Proteostasis defects: Medicinal challenges of imperfect aging & neurodegeneration. Transl. Med. Aging 2023, 7, 87–97. [Google Scholar]
  29. Wu, X.; Liu, Z.; Yu, X.Y.; Xu, S.; Luo, J. Autophagy and cardiac diseases: Therapeutic potential of natural products. Med. Res. Rev. 2021, 41, 314–341. [Google Scholar] [CrossRef]
  30. Klionsky, D.J.; Petroni, G.; Amaravadi, R.K.; Baehrecke, E.H.; Ballabio, A.; Boya, P.; Bravo-San Pedro, J.M.; Cadwell, K.; Cecconi, F.; Choi, A.M.K.; et al. Autophagy in major human diseases. Embo J. 2021, 40, e108863. [Google Scholar] [CrossRef]
  31. Eisenberg, T.; Abdellatif, M.; Schroeder, S.; Primessnig, U.; Stekovic, S.; Pendl, T.; Harger, A.; Schipke, J.; Zimmermann, A.; Schmidt, A.; et al. Cardioprotection and lifespan extension by the natural polyamine spermidine. Nat. Med. 2016, 22, 1428–1438. [Google Scholar] [CrossRef]
  32. Jiang, B.; Zhou, X.; Yang, T.; Wang, L.; Feng, L.; Wang, Z.; Xu, J.; Jing, W.; Wang, T.; Su, H.; et al. The role of autophagy in cardiovascular disease: Cross-interference of signaling pathways and underlying therapeutic targets. Front. Cardiovasc. Med. 2023, 10, 1088575. [Google Scholar] [CrossRef]
  33. Luo, G.; Jian, Z.; Zhu, Y.; Zhu, Y.; Chen, B.; Ma, R.; Tang, F.; Xiao, Y. Sirt1 promotes autophagy and inhibits apoptosis to protect cardiomyocytes from hypoxic stress. Int. J. Mol. Med. 2019, 43, 2033–2043. [Google Scholar] [CrossRef] [PubMed]
  34. Farhan, M.; Silva, M.; Xingan, X.; Huang, Y.; Zheng, W. Role of FOXO Transcription Factors in Cancer Metabolism and Angiogenesis. Cells 2020, 9, 1586. [Google Scholar] [CrossRef] [PubMed]
  35. Hariharan, N.; Maejima, Y.; Nakae, J.; Paik, J.; Depinho, R.A.; Sadoshima, J. Deacetylation of FoxO by Sirt1 Plays an Essential Role in Mediating Starvation-Induced Autophagy in Cardiac Myocytes. Circ. Res. 2010, 107, 1470–1482. [Google Scholar] [CrossRef] [PubMed]
  36. Maithili Karpaga Selvi, N.; Sridhar, M.G.; Swaminathan, R.P.; Sripradha, R. Curcumin Attenuates Oxidative Stress and Activation of Redox-Sensitive Kinases in High Fructose- and High-Fat-Fed Male Wistar Rats. Sci. Pharm. 2015, 83, 159–175. [Google Scholar] [CrossRef]
  37. Kong, F.; Ye, B.; Cao, J.; Cai, X.; Lin, L.; Huang, S.; Huang, W.; Huang, Z. Curcumin Represses NLRP3 Inflammasome Activation via TLR4/MyD88/NF-κB and P2X7R Signaling in PMA-Induced Macrophages. Front. Pharmacol. 2016, 7, 369. [Google Scholar] [CrossRef]
  38. Jang, S.Y.; Kim, J.; Hong, E.; Lee, K.; Na, Y.; Yeom, C.H.; Park, S. Curcumin inhibits human cancer cell growth and migration through downregulation of SVCT2. Cell Biochem. Funct. 2023, 41, 696–703. [Google Scholar] [CrossRef]
  39. Akaberi, M.; Sahebkar, A.; Emami, S.A. Turmeric and Curcumin: From Traditional to Modern Medicine. Adv. Exp. Med. Biol. 2021, 1291, 15–39. [Google Scholar]
  40. Moustapha, A.; Pérétout, P.A.; Rainey, N.E.; Sureau, F.; Geze, M.; Petit, J.M.; Dewailly, E.; Slomianny, C.; Petit, P.X. Curcumin induces crosstalk between autophagy and apoptosis mediated by calcium release from the endoplasmic reticulum, lysosomal destabilization and mitochondrial events. Cell Death Discov. 2015, 1, 15017. [Google Scholar] [CrossRef]
  41. Zhu, Y.; Bu, S. Curcumin Induces Autophagy, Apoptosis, and Cell Cycle Arrest in Human Pancreatic Cancer Cells. Evid. Based Complement. Alternat. Med. 2017, 2017, 5787218. [Google Scholar] [CrossRef]
  42. Kim, J.Y.; Cho, T.J.; Woo, B.H.; Choi, K.U.; Lee, C.H.; Ryu, M.H.; Park, H.R. Curcumin-induced autophagy contributes to the decreased survival of oral cancer cells. Arch. Oral Biol. 2012, 57, 1018–1025. [Google Scholar] [CrossRef]
  43. Zhang, J.; Wang, J.; Xu, J.; Lu, Y.; Jiang, J.; Wang, L.; Shen, H.M.; Xia, D. Curcumin targets the TFEB-lysosome pathway for induction of autophagy. Oncotarget 2016, 7, 75659–75671. [Google Scholar] [CrossRef] [PubMed]
  44. Alers, S.; Löffler, A.S.; Wesselborg, S.; Stork, B. Role of AMPK-mTOR-Ulk1/2 in the regulation of autophagy: Cross talk, shortcuts, and feedbacks. Mol Cell Biol 2012, 32, 2–11. [Google Scholar] [CrossRef] [PubMed]
  45. Xu, J.; Du, P.; Liu, X.; Xu, X.; Ge, Y.; Zhang, C. Curcumin supplementation increases longevity and antioxidant capacity in Caenorhabditis elegans. Front. Pharmacol. 2023, 14, 1195490. [Google Scholar] [CrossRef] [PubMed]
  46. Suckow, B.K.; Suckow, M.A. Lifespan extension by the antioxidant curcumin in Drosophila melanogaster. Int. J. Biomed. Sci. 2006, 2, 402–405. [Google Scholar] [CrossRef] [PubMed]
  47. Lee, K.S.; Lee, B.S.; Semnani, S.; Avanesian, A.; Um, C.Y.; Jeon, H.J.; Seong, K.M.; Yu, K.; Min, K.J.; Jafari, M. Curcumin extends life span, improves health span, and modulates the expression of age-associated aging genes in Drosophila melanogaster. Rejuvenation Res. 2010, 13, 561–570. [Google Scholar] [CrossRef]
  48. Stępień, K.; Wojdyła, D.; Nowak, K.; Mołoń, M. Impact of curcumin on replicative and chronological aging in the Saccharomyces cerevisiae yeast. Biogerontology 2020, 21, 109–123. [Google Scholar] [CrossRef]
  49. Kuo, J.J.; Chang, H.H.; Tsai, T.H.; Lee, T.Y. Curcumin ameliorates mitochondrial dysfunction associated with inhibition of gluconeogenesis in free fatty acid-mediated hepatic lipoapoptosis. Int. J. Mol. Med. 2012, 30, 643–649. [Google Scholar] [CrossRef]
  50. Wang, D.; Yang, Y.; Zou, X.; Zheng, Z.; Zhang, J. Curcumin ameliorates CKD-induced mitochondrial dysfunction and oxidative stress through inhibiting GSK-3β activity. J. Nutr. Biochem. 2020, 83, 108404. [Google Scholar] [CrossRef]
  51. Sathyabhama, M.; Priya Dharshini, L.C.; Karthikeyan, A.; Kalaiselvi, S.; Min, T. The Credible Role of Curcumin in Oxidative Stress-Mediated Mitochondrial Dysfunction in Mammals. Biomolecules 2022, 12, 1405. [Google Scholar] [CrossRef]
  52. Sadeghi, S.; Delphan, M.; Shams, M.; Esmaeili, F.; Shanaki-Bavarsad, M.; Shanaki, M. The high-intensity interval training (HIIT) and curcumin supplementation can positively regulate the autophagy pathway in myocardial cells of STZ-induced diabetic rats. BMC Res. Notes 2023, 16, 21. [Google Scholar] [CrossRef]
  53. Yu, W.; Qin, X.; Zhang, Y.; Qiu, P.; Wang, L.; Zha, W.; Ren, J. Curcumin suppresses doxorubicin-induced cardiomyocyte pyroptosis via a PI3K/Akt/mTOR-dependent manner. Cardiovasc. Diagn. Ther. 2020, 10, 752–769. [Google Scholar] [CrossRef] [PubMed]
  54. Yang, L.; Shi, J.; Wang, X.; Zhang, R. Curcumin Alleviates D-Galactose-Induced Cardiomyocyte Senescence by Promoting Autophagy via the SIRT1/AMPK/mTOR Pathway. Evid.-Based Complement. Altern. Med. 2022, 2022, 2990843. [Google Scholar] [CrossRef] [PubMed]
  55. (US), N.C.I. Curcumin (Curcuma, Turmeric) and Cancer (PDQ®): Patient Version. Available online: https://www.ncbi.nlm.nih.gov/books/NBK578436/ (accessed on 28 May 2024).
  56. Thapa, S.B.; Pandey, R.P.; Park, Y.I.; Kyung Sohng, J. Biotechnological Advances in Resveratrol Production and its Chemical Diversity. Molecules 2019, 24, 2571. [Google Scholar] [CrossRef] [PubMed]
  57. Singh, A.K.; Vinayak, M. Resveratrol alleviates inflammatory hyperalgesia by modulation of reactive oxygen species (ROS), antioxidant enzymes and ERK activation. Inflamm. Res. 2017, 66, 911–921. [Google Scholar] [CrossRef]
  58. Inglés, M.; Gambini, J.; Miguel, M.G.; Bonet-Costa, V.; Abdelaziz, K.M.; El Alami, M.; Viña, J.; Borrás, C. PTEN mediates the antioxidant effect of resveratrol at nutritionally relevant concentrations. Biomed. Res. Int. 2014, 2014, 580852. [Google Scholar] [CrossRef]
  59. Meng, T.; Xiao, D.; Muhammed, A.; Deng, J.; Chen, L.; He, J. Anti-Inflammatory Action and Mechanisms of Resveratrol. Molecules 2021, 26, 229. [Google Scholar] [CrossRef]
  60. Song, B.; Wang, W.; Tang, X.; Goh, R.M.W.; Thuya, W.L.; Ho, P.C.L.; Chen, L.; Wang, L. Inhibitory Potential of Resveratrol in Cancer Metastasis: From Biology to Therapy. Cancers 2023, 15, 2758. [Google Scholar] [CrossRef]
  61. Park, D.; Jeong, H.; Lee, M.N.; Koh, A.; Kwon, O.; Yang, Y.R.; Noh, J.; Suh, P.-G.; Park, H.; Ryu, S.H. Resveratrol induces autophagy by directly inhibiting mTOR through ATP competition. Sci. Rep. 2016, 6, 21772. [Google Scholar] [CrossRef]
  62. Mauthe, M.; Jacob, A.; Freiberger, S.; Hentschel, K.; Stierhof, Y.D.; Codogno, P.; Proikas-Cezanne, T. Resveratrol-mediated autophagy requires WIPI-1-regulated LC3 lipidation in the absence of induced phagophore formation. Autophagy 2011, 7, 1448–1461. [Google Scholar] [CrossRef]
  63. Ferretta, A.; Gaballo, A.; Tanzarella, P.; Piccoli, C.; Capitanio, N.; Nico, B.; Annese, T.; Di Paola, M.; Dell’Aquila, C.; De Mari, M.; et al. Effect of resveratrol on mitochondrial function: Implications in parkin-associated familiar Parkinson’s disease. Biochim. Biophys. Acta (BBA)—Mol. Basis Dis. 2014, 1842, 902–915. [Google Scholar] [CrossRef]
  64. Tomou, E.M.; Papakyriakopoulou, P.; Skaltsa, H.; Valsami, G.; Kadoglou, N.P.E. Bio-Actives from Natural Products with Potential Cardioprotective Properties: Isolation, Identification, and Pharmacological Actions of Apigenin, Quercetin, and Silibinin. Molecules 2023, 28, 2387. [Google Scholar] [CrossRef] [PubMed]
  65. Shah, S.M.A.; Akram, M.; Riaz, M.; Munir, N.; Rasool, G. Cardioprotective Potential of Plant-Derived Molecules: A Scientific and Medicinal Approach. Dose Response 2019, 17, 1559325819852243. [Google Scholar] [CrossRef] [PubMed]
  66. Wu, Z.; Huang, A.; Yan, J.; Liu, B.; Liu, Q.; Zhang, J.; Zhang, X.; Ou, C.; Chen, M. Resveratrol Ameliorates Cardiac Dysfunction by Inhibiting Apoptosis via the PI3K/Akt/FoxO3a Pathway in a Rat Model of Diabetic Cardiomyopathy. J. Cardiovasc. Pharmacol. 2017, 70, 184–193. [Google Scholar] [CrossRef] [PubMed]
  67. Zhou, X.; Yang, J.; Zhou, M.; Zhang, Y.; Liu, Y.; Hou, P.; Zeng, X.; Yi, L.; Mi, M. Resveratrol attenuates endothelial oxidative injury by inducing autophagy via the activation of transcription factor EB. Nutr. Metab. 2019, 16, 42. [Google Scholar] [CrossRef]
  68. Wang, B.; Yang, Q.; Sun, Y.Y.; Xing, Y.F.; Wang, Y.B.; Lu, X.T.; Bai, W.W.; Liu, X.Q.; Zhao, Y.X. Resveratrol-enhanced autophagic flux ameliorates myocardial oxidative stress injury in diabetic mice. J. Cell. Mol. Med. 2014, 18, 1599–1611. [Google Scholar] [CrossRef]
  69. Michala, A.S.; Pritsa, A. Quercetin: A Molecule of Great Biochemical and Clinical Value and Its Beneficial Effect on Diabetes and Cancer. Diseases 2022, 10, 37. [Google Scholar] [CrossRef]
  70. Shabir, I.; Kumar Pandey, V.; Shams, R.; Dar, A.H.; Dash, K.K.; Khan, S.A.; Bashir, I.; Jeevarathinam, G.; Rusu, A.V.; Esatbeyoglu, T.; et al. Promising bioactive properties of quercetin for potential food applications and health benefits: A review. Front. Nutr. 2022, 9, 999752. [Google Scholar] [CrossRef]
  71. Deepika; Maurya, P.K. Health Benefits of Quercetin in Age-Related Diseases. Molecules 2022, 27, 2498. [Google Scholar] [CrossRef]
  72. Rahmani, A.H.; Alsahli, M.A.; Khan, A.A.; Almatroodi, S.A. Quercetin, a Plant Flavonol Attenuates Diabetic Complications, Renal Tissue Damage, Renal Oxidative Stress and Inflammation in Streptozotocin-Induced Diabetic Rats. Metabolites 2023, 13, 130. [Google Scholar] [CrossRef]
  73. Dhanya, R. Quercetin for managing type 2 diabetes and its complications, an insight into multitarget therapy. Biomed. Pharmacother. 2022, 146, 112560. [Google Scholar] [CrossRef]
  74. Shahbaz, M.; Naeem, H.; Momal, U.; Imran, M.; Alsagaby, S.A.; Al Abdulmonem, W.; Waqar, A.B.; El-Ghorab, A.H.; Ghoneim, M.M.; Abdelgawad, M.A.; et al. Anticancer and apoptosis inducing potential of quercetin against a wide range of human malignancies. Int. J. Food Prop. 2023, 26, 2590–2626. [Google Scholar] [CrossRef]
  75. Asgharian, P.; Tazekand, A.P.; Hosseini, K.; Forouhandeh, H.; Ghasemnejad, T.; Ranjbar, M.; Hasan, M.; Kumar, M.; Beirami, S.M.; Tarhriz, V.; et al. Potential mechanisms of quercetin in cancer prevention: Focus on cellular and molecular targets. Cancer Cell Int. 2022, 22, 257. [Google Scholar] [CrossRef] [PubMed]
  76. Xiao, J.; Zhang, B.; Yin, S.; Xie, S.; Huang, K.; Wang, J.; Yang, W.; Liu, H.; Zhang, G.; Liu, X.; et al. Quercetin induces autophagy-associated death in HL-60 cells through CaMKKβ/AMPK/mTOR signal pathway. Acta Biochim. Biophys. Sin. 2022, 54, 1244–1256. [Google Scholar] [PubMed]
  77. Guo, H.; Ding, H.; Tang, X.; Liang, M.; Li, S.; Zhang, J.; Cao, J. Quercetin induces pro-apoptotic autophagy via SIRT1/AMPK signaling pathway in human lung cancer cell lines A549 and H1299 in vitro. Thorac. Cancer 2021, 12, 1415–1422. [Google Scholar] [CrossRef]
  78. Peng, J.; Yang, Z.; Li, H.; Hao, B.; Cui, D.; Shang, R.; Lv, Y.; Liu, Y.; Pu, W.; Zhang, H.; et al. Quercetin Reprograms Immunometabolism of Macrophages via the SIRT1/PGC-1α Signaling Pathway to Ameliorate Lipopolysaccharide-Induced Oxidative Damage. Int. J. Mol. Sci. 2023, 24, 5542. [Google Scholar] [CrossRef]
  79. Chen, Y.F.; Qiu, Q.; Wang, L.; Li, X.R.; Zhou, S.; Wang, H.; Jiang, W.D.; Geng, J.Y.; Qin, G.; Tang, B.; et al. Quercetin Ameliorates Myocardial Injury in Diabetic Rats by Regulating Autophagy and Apoptosis through AMPK/mTOR Signaling Pathway. Am. J. Chin. Med. 2024, 52, 841–864. [Google Scholar] [CrossRef]
  80. Wang, Y.; Zhang, Z.Z.; Wu, Y.; Ke, J.J.; He, X.H.; Wang, Y.L. Quercetin postconditioning attenuates myocardial ischemia/reperfusion injury in rats through the PI3K/Akt pathway. Braz. J. Med. Biol. Res. 2013, 46, 861–867. [Google Scholar] [CrossRef]
  81. Lin, X.; Han, T.; Fan, Y.; Wu, S.; Wang, F.; Wang, C. Quercetin improves vascular endothelial function through promotion of autophagy in hypertensive rats. Life Sci. 2020, 258, 118106. [Google Scholar] [CrossRef]
  82. Hu, J.; Wang, X.; Cui, X.; Kuang, W.; Li, D.; Wang, J. Quercetin prevents isoprenaline-induced myocardial fibrosis by promoting autophagy via regulating miR-223-3p/FOXO3. Cell Cycle 2021, 20, 1253–1269. [Google Scholar] [CrossRef]
  83. Muñoz-Esparza, N.C.; Costa-Catala, J.; Comas-Basté, O.; Toro-Funes, N.; Latorre-Moratalla, M.L.; Veciana-Nogués, M.T.; Vidal-Carou, M.C. Occurrence of Polyamines in Foods and the Influence of Cooking Processes. Foods 2021, 10, 1752. [Google Scholar] [CrossRef]
  84. Bouchereau, A.; Guénot, P.; Larher, F. Analysis of amines in plant materials. Chromatogr. B Biomed. Sci. Appl. 2000, 747, 49–67. [Google Scholar] [CrossRef] [PubMed]
  85. Herbst, E.J.; Tanguay, R.B. The Interaction of Polyamines with Nucleic Acids. In Proceedings of the Research Symposium on Complexes of Biologically Active Substances with Nucleic Acids and Their Modes of Action: Held at the Walter Reed Army Institute of Research Washington, Washington, DC, USA, 16–19 March 1970; Hahn, F.E., Ed.; Springer: Berlin/Heidelberg, Germany, 1971; pp. 166–180. [Google Scholar]
  86. Schuster, I.; Bernhardt, R. Interactions of natural polyamines with mammalian proteins. Biomol. Concepts 2011, 2, 79–94. [Google Scholar] [CrossRef] [PubMed]
  87. Michael, A.J. Biosynthesis of polyamines and polyamine-containing molecules. Biochem. J. 2016, 473, 2315–2329. [Google Scholar] [CrossRef] [PubMed]
  88. Hofer, S.J.; Simon, A.K.; Bergmann, M.; Eisenberg, T.; Kroemer, G.; Madeo, F. Mechanisms of spermidine-induced autophagy and geroprotection. Nat. Aging 2022, 2, 1112–1129. [Google Scholar] [CrossRef]
  89. Wang, J.; Li, S.; Wang, J.; Wu, F.; Chen, Y.; Zhang, H.; Guo, Y.; Lin, Y.; Li, L.; Yu, X.; et al. Spermidine alleviates cardiac aging by improving mitochondrial biogenesis and function. Aging 2020, 12, 650–671. [Google Scholar] [CrossRef]
  90. Fairley, L.H.; Lejri, I.; Grimm, A.; Eckert, A. Spermidine Rescues Bioenergetic and Mitophagy Deficits Induced by Disease-Associated Tau Protein. Int. J. Mol. Sci. 2023, 24, 5297. [Google Scholar] [CrossRef]
  91. Zhang, H.; Alsaleh, G.; Feltham, J.; Sun, Y.; Napolitano, G.; Riffelmacher, T.; Charles, P.; Frau, L.; Hublitz, P.; Yu, Z.; et al. Polyamines Control eIF5A Hypusination, TFEB Translation, and Autophagy to Reverse B Cell Senescence. Mol. Cell 2019, 76, 110–125.e9. [Google Scholar] [CrossRef]
  92. Yang, Y.; Chen, S.; Zhang, Y.; Lin, X.; Song, Y.; Xue, Z.; Qian, H.; Wang, S.; Wan, G.; Zheng, X.; et al. Induction of autophagy by spermidine is neuroprotective via inhibition of caspase 3-mediated Beclin 1 cleavage. Cell Death Dis. 2017, 8, e2738. [Google Scholar] [CrossRef]
  93. Zhang, H.; Wang, J.; Li, L.; Chai, N.; Chen, Y.; Wu, F.; Zhang, W.; Wang, L.; Shi, S.; Zhang, L.; et al. Spermine and spermidine reversed age-related cardiac deterioration in rats. Oncotarget 2017, 8, 64793–64808. [Google Scholar] [CrossRef]
  94. Michiels, C.F.; Kurdi, A.; Timmermans, J.P.; De Meyer, G.R.Y.; Martinet, W. Spermidine reduces lipid accumulation and necrotic core formation in atherosclerotic plaques via induction of autophagy. Atherosclerosis 2016, 251, 319–327. [Google Scholar] [CrossRef]
  95. Yan, J.; Yan, J.Y.; Wang, Y.X.; Ling, Y.N.; Song, X.D.; Wang, S.Y.; Liu, H.Q.; Liu, Q.C.; Zhang, Y.; Yang, P.Z.; et al. Spermidine-enhanced autophagic flux improves cardiac dysfunction following myocardial infarction by targeting the AMPK/mTOR signalling pathway. Br. J. Pharmacol. 2019, 176, 3126–3142. [Google Scholar] [CrossRef] [PubMed]
  96. Hou, Y.; Dan, X.; Babbar, M.; Wei, Y.; Hasselbalch, S.G.; Croteau, D.L.; Bohr, V.A. Ageing as a risk factor for neurodegenerative disease. Nat. Rev. Neurol. 2019, 15, 565–581. [Google Scholar] [CrossRef] [PubMed]
  97. Leidal, A.M.; Levine, B.; Debnath, J. Autophagy and the cell biology of age-related disease. Nat. Cell Biol. 2018, 20, 1338–1348. [Google Scholar] [CrossRef] [PubMed]
  98. Lynch-Day, M.A.; Mao, K.; Wang, K.; Zhao, M.; Klionsky, D.J. The role of autophagy in Parkinson’s disease. Cold Spring Harb. Perspect. Med. 2012, 2, a009357. [Google Scholar] [CrossRef]
  99. Deng, Z.; Zhou, X.; Lu, J.H.; Yue, Z. Autophagy deficiency in neurodevelopmental disorders. Cell Biosci. 2021, 11, 214. [Google Scholar] [CrossRef]
  100. Uddin, M.S.; Stachowiak, A.; Mamun, A.A.; Tzvetkov, N.T.; Takeda, S.; Atanasov, A.G.; Bergantin, L.B.; Abdel-Daim, M.M.; Stankiewicz, A.M. Autophagy and Alzheimer’s Disease: From Molecular Mechanisms to Therapeutic Implications. Front. Aging Neurosci. 2018, 10, 4. [Google Scholar] [CrossRef]
  101. François, A.; Rioux Bilan, A.; Quellard, N.; Fernandez, B.; Janet, T.; Chassaing, D.; Paccalin, M.; Terro, F.; Page, G. Longitudinal follow-up of autophagy and inflammation in brain of APPswePS1dE9 transgenic mice. J. Neuroinflamm. 2014, 11, 139. [Google Scholar] [CrossRef]
  102. Tan, C.; Ai, J.; Zhu, Y. mTORC1-Dependent Protein and Parkinson’s Disease: A Mendelian Randomization Study. Brain Sci. 2023, 13, 536. [Google Scholar] [CrossRef]
  103. Batiha, G.E.; Al-Kuraishy, H.M.; Al-Gareeb, A.I.; Elekhnawy, E. SIRT1 pathway in Parkinson’s disease: A faraway snapshot but so close. Inflammopharmacology 2023, 31, 37–56. [Google Scholar] [CrossRef]
  104. Mehramiz, M.; Porter, T.; O’Brien, E.K.; Rainey-Smith, S.R.; Laws, S.M. A Potential Role for Sirtuin-1 in Alzheimer’s Disease: Reviewing the Biological and Environmental Evidence. J. Alzheimers Dis. Rep. 2023, 7, 823–843. [Google Scholar] [CrossRef]
  105. Maruyama, H.; Ooizumi, T.; Kawakami, F.; Lwin, T.-T.; Akita, H.; Kunii, T.; Shirai, R.; Takeda, T. Long-term oral administration of curcumin is effective in preventing short-term memory deterioration and prolonging lifespan in a mouse model of Alzheimer’s disease. Adv. Tradit. Med. 2024, 24, 373–385. [Google Scholar] [CrossRef]
  106. Yi, L.-T.; Dong, S.-Q.; Wang, S.-S.; Chen, M.; Li, C.-F.; Geng, D.; Zhu, J.-X.; Liu, Q.; Cheng, J. Curcumin attenuates cognitive impairment by enhancing autophagy in chemotherapy. Neurobiol. Dis. 2020, 136, 104715. [Google Scholar] [CrossRef] [PubMed]
  107. Rathore, A.S.; Singh, S.S.; Birla, H.; Zahra, W.; Keshri, P.K.; Dilnashin, H.; Singh, R.; Singh, S.; Singh, S.P. Curcumin Modulates p62–Keap1–Nrf2-Mediated Autophagy in Rotenone-Induced Parkinson’s Disease Mouse Models. ACS Chem. Neurosci. 2023, 14, 1412–1423. [Google Scholar] [CrossRef] [PubMed]
  108. Kou, X.; Chen, N. Resveratrol as a Natural Autophagy Regulator for Prevention and Treatment of Alzheimer’s Disease. Nutrients 2017, 9, 927. [Google Scholar] [CrossRef]
  109. Wang, N.; He, J.; Pan, C.; Wang, J.; Ma, M.; Shi, X.; Xu, Z. Resveratrol Activates Autophagy via the AKT/mTOR Signaling Pathway to Improve Cognitive Dysfunction in Rats with Chronic Cerebral Hypoperfusion. Front. Neurosci. 2019, 13, 859. [Google Scholar] [CrossRef]
  110. Xu, J.; Jackson, C.W.; Khoury, N.; Escobar, I.; Perez-Pinzon, M.A. Brain SIRT1 Mediates Metabolic Homeostasis and Neuroprotection. Front. Endocrinol. 2018, 9, 702. [Google Scholar] [CrossRef]
  111. Baeken, M.W. Sirtuins and their influence on autophagy. J. Cell Biochem. 2023. [Google Scholar] [CrossRef]
  112. Hao, Y.; Shao, L.; Hou, J.; Zhang, Y.; Ma, Y.; Liu, J.; Xu, C.; Chen, F.; Cao, L.H.; Ping, Y. Resveratrol and Sir2 Reverse Sleep and Memory Defects Induced by Amyloid Precursor Protein. Neurosci. Bull. 2023, 39, 1117–1130. [Google Scholar] [CrossRef]
  113. Turner, R.S.; Thomas, R.G.; Craft, S.; van Dyck, C.H.; Mintzer, J.; Reynolds, B.A.; Brewer, J.B.; Rissman, R.A.; Raman, R.; Aisen, P.S. A randomized, double-blind, placebo-controlled trial of resveratrol for Alzheimer disease. Neurology 2015, 85, 1383–1391. [Google Scholar] [CrossRef]
  114. Zhang, L.F.; Yu, X.L.; Ji, M.; Liu, S.Y.; Wu, X.L.; Wang, Y.J.; Liu, R.T. Resveratrol alleviates motor and cognitive deficits and neuropathology in the A53T α-synuclein mouse model of Parkinson’s disease. Food Funct. 2018, 9, 6414–6426. [Google Scholar] [CrossRef]
  115. Illes-Toth, E.; Rempel, D.L.; Gross, M.L. Exploration of Resveratrol as a Potent Modulator of α-Synuclein Fibril Formation. ACS Chem. Neurosci. 2024, 15, 503–516. [Google Scholar] [CrossRef] [PubMed]
  116. Guo, Y.J.; Dong, S.Y.; Cui, X.X.; Feng, Y.; Liu, T.; Yin, M.; Kuo, S.H.; Tan, E.K.; Zhao, W.J.; Wu, Y.C. Resveratrol alleviates MPTP-induced motor impairments and pathological changes by autophagic degradation of α-synuclein via SIRT1-deacetylated LC3. Mol. Nutr. Food Res. 2016, 60, 2161–2175. [Google Scholar] [CrossRef]
  117. Shen, D.F.; Qi, H.P.; Zhang, W.N.; Sang, W.X. Resveratrol Promotes Autophagy to Improve neuronal Injury in Parkinson’s Disease by Regulating SNHG1/miR-128-3p/SNCA Axis. Brain Sci. 2023, 13, 1124. [Google Scholar] [CrossRef] [PubMed]
  118. Ning, B.; Hang, S.; Zhang, W.; Mao, C.; Li, D. An update on the bridging factors connecting autophagy and Nrf2 antioxidant pathway. Front. Cell Dev. Biol. 2023, 11, 1232241. [Google Scholar] [CrossRef] [PubMed]
  119. Cheng, Z. The FoxO-Autophagy Axis in Health and Disease. Trends Endocrinol. Metab. 2019, 30, 658–671. [Google Scholar] [CrossRef]
  120. Collier, J.J.; Suomi, F.; Oláhová, M.; McWilliams, T.G.; Taylor, R.W. Emerging roles of ATG7 in human health and disease. EMBO Mol. Med. 2021, 13, e14824. [Google Scholar] [CrossRef]
  121. Sabarathinam, S. Unraveling the therapeutic potential of quercetin and quercetin-3-O-glucuronide in Alzheimer’s disease through network pharmacology, molecular docking, and dynamic simulations. Sci. Rep. 2024, 14, 14852. [Google Scholar] [CrossRef]
  122. León, R.; Gutiérrez, D.A.; Pinto, C.; Morales, C.; de la Fuente, C.; Riquelme, C.; Cortés, B.I.; González-Martin, A.; Chamorro, D.; Espinosa, N.; et al. c-Abl tyrosine kinase down-regulation as target for memory improvement in Alzheimer’s disease. Front. Aging Neurosci. 2023, 15, 1180987. [Google Scholar] [CrossRef]
  123. Schiavi, A.; Cirotti, C.; Gerber, L.-S.; Di Lauro, G.; Maglioni, S.; Shibao, P.Y.T.; Montresor, S.; Kirstein, J.; Petzsch, P.; Köhrer, K.; et al. Abl depletion via autophagy mediates the beneficial effects of quercetin against Alzheimer pathology across species. Cell Death Discov. 2023, 9, 376. [Google Scholar] [CrossRef]
  124. Liu, Y.; Zhou, H.; Yin, T.; Gong, Y.; Yuan, G.; Chen, L.; Liu, J. Quercetin-modified gold-palladium nanoparticles as a potential autophagy inducer for the treatment of Alzheimer’s disease. J. Colloid Interface Sci. 2019, 552, 388–400. [Google Scholar] [CrossRef]
  125. Kong, Y.; Li, K.; Fu, T.; Wan, C.; Zhang, D.; Song, H.; Zhang, Y.; Liu, N.; Gan, Z.; Yuan, L. Quercetin ameliorates Aβ toxicity in Drosophila AD model by modulating cell cycle-related protein expression. Oncotarget 2016, 7, 67716–67731. [Google Scholar] [CrossRef] [PubMed]
  126. Yu, X.; Li, Y.; Mu, X. Effect of Quercetin on PC12 Alzheimer’s Disease Cell Model Induced by Aβ25-35 and Its Mechanism Based on Sirtuin1/Nrf2/HO-1 Pathway. BioMed Res. Int. 2020, 2020, 8210578. [Google Scholar] [CrossRef] [PubMed]
  127. Pakrashi, S.; Chakraborty, J.; Bandyopadhyay, J. Neuroprotective Role of Quercetin on Rotenone-Induced Toxicity in SH-SY5Y Cell Line Through Modulation of Apoptotic and Autophagic Pathways. Neurochem. Res. 2020, 45, 1962–1973. [Google Scholar] [CrossRef] [PubMed]
  128. Wang, W.W.; Han, R.; He, H.J.; Li, J.; Chen, S.Y.; Gu, Y.; Xie, C. Administration of quercetin improves mitochondria quality control and protects the neurons in 6-OHDA-lesioned Parkinson’s disease models. Aging (Albany NY) 2021, 13, 11738–11751. [Google Scholar] [CrossRef]
  129. El-Horany, H.E.; El-Latif, R.N.; ElBatsh, M.M.; Emam, M.N. Ameliorative Effect of Quercetin on Neurochemical and Behavioral Deficits in Rotenone Rat Model of Parkinson’s Disease: Modulating Autophagy (Quercetin on Experimental Parkinson’s Disease). J. Biochem. Mol. Toxicol. 2016, 30, 360–369. [Google Scholar] [CrossRef]
  130. Jimenez Gutierrez, G.E.; Borbolla Jiménez, F.V.; Muñoz, L.G.; Tapia Guerrero, Y.S.; Murillo Melo, N.M.; Cristóbal-Luna, J.M.; Leyva Garcia, N.; Cordero-Martínez, J.; Magaña, J.J. The Molecular Role of Polyamines in Age-Related Diseases: An Update. Int. J. Mol. Sci. 2023, 24, 16469. [Google Scholar] [CrossRef]
  131. Williams, K.; Romano, C.; Dichter, M.A.; Molinoff, P.B. Modulation of the NMDA receptor by polyamines. Life Sci. 1991, 48, 469–498. [Google Scholar] [CrossRef]
  132. Morrison, L.D.; Kish, S.J. Brain polyamine levels are altered in Alzheimer’s disease. Neurosci. Lett. 1995, 197, 5–8. [Google Scholar] [CrossRef]
  133. Sandusky-Beltran, L.A.; Kovalenko, A.; Ma, C.; Calahatian, J.I.T.; Placides, D.S.; Watler, M.D.; Hunt, J.B.; Darling, A.L.; Baker, J.D.; Blair, L.J.; et al. Spermidine/spermine-N(1)-acetyltransferase ablation impacts tauopathy-induced polyamine stress response. Alzheimers Res. Ther. 2019, 11, 58. [Google Scholar] [CrossRef]
  134. Xu, T.T.; Li, H.; Dai, Z.; Lau, G.K.; Li, B.Y.; Zhu, W.L.; Liu, X.Q.; Liu, H.F.; Cai, W.W.; Huang, S.Q.; et al. Spermidine and spermine delay brain aging by inducing autophagy in SAMP8 mice. Aging 2020, 12, 6401–6414. [Google Scholar] [CrossRef]
  135. Lumkwana, D.; Peddie, C.; Kriel, J.; Michie, L.L.; Heathcote, N.; Collinson, L.; Kinnear, C.; Loos, B. Investigating the Role of Spermidine in a Model System of Alzheimer’s Disease Using Correlative Microscopy and Super-resolution Techniques. Front. Cell Dev. Biol. 2022, 10, 819571. [Google Scholar] [CrossRef] [PubMed]
  136. Mackenbrock, L.H.B.; Labuz, G.; Baur, I.D.; Yildirim, T.M.; Auffarth, G.U.; Khoramnia, R. Cataract Classification Systems: A Review. Klin. Monbl. Augenheilkd. 2024, 241, 75–83. [Google Scholar] [CrossRef] [PubMed]
  137. Pesudovs, K.; Lansingh, V.C.; Kempen, J.H.; Tapply, I.; Fernandes, A.G.; Cicinelli, M.V.; Arrigo, A.; Leveziel, N.; Briant, P.S.; Vos, T.; et al. Global estimates on the number of people blind or visually impaired by cataract: A meta-analysis from 2000 to 2020. Eye 2024, 38, 2156–2172. [Google Scholar]
  138. Fernández-Albarral, J.A.; de Julián-López, E.; Soler-Domínguez, C.; de Hoz, R.; López-Cuenca, I.; Salobrar-García, E.; Ramírez, J.M.; Pinazo-Durán, M.D.; Salazar, J.J.; Ramírez, A.I. The Role of Autophagy in Eye Diseases. Life 2021, 11, 189. [Google Scholar] [CrossRef]
  139. Costello, M.J.; Brennan, L.A.; Basu, S.; Chauss, D.; Mohamed, A.; Gilliland, K.O.; Johnsen, S.; Menko, S.; Kantorow, M. Autophagy and mitophagy participate in ocular lens organelle degradation. Exp. Eye Res. 2013, 116, 141–150. [Google Scholar] [CrossRef]
  140. Morishita, H. Role of autophagy in the eye: From physiology to disease. Curr. Opin. Physiol. 2022, 30, 100592. [Google Scholar] [CrossRef]
  141. Chai, P.; Ni, H.; Zhang, H.; Fan, X. The Evolving Functions of Autophagy in Ocular Health: A Double-edged Sword. Int. J. Biol. Sci. 2016, 12, 1332–1340. [Google Scholar] [CrossRef]
  142. Sidjanin, D.J.; Park, A.K.; Ronchetti, A.; Martins, J.; Jackson, W.T. TBC1D20 mediates autophagy as a key regulator of autophagosome maturation. Autophagy 2016, 12, 1759–1775. [Google Scholar] [CrossRef]
  143. Cai, Y.; Liu, K.; Wu, P.; Yuan, R.; He, F.; Zou, J. Association of mTORC1-dependent circulating protein levels with cataract formation: A mendelian randomization study. BMC Genom. 2022, 23, 719. [Google Scholar] [CrossRef]
  144. Shukal, D.K.; Malaviya, P.B.; Sharma, T. Role of the AMPK signalling pathway in the aetiopathogenesis of ocular diseases. Hum. Exp. Toxicol. 2022, 41, 9603271211063165. [Google Scholar] [CrossRef]
  145. Zhou, M.; Luo, J.; Zhang, H. Role of Sirtuin 1 in the pathogenesis of ocular disease (Review). Int. J. Mol. Med. 2018, 42, 13–20. [Google Scholar] [CrossRef] [PubMed]
  146. Radomska-Leśniewska, D.M.; Osiecka-Iwan, A.; Hyc, A.; Góźdź, A.; Dąbrowska, A.M.; Skopiński, P. Therapeutic potential of curcumin in eye diseases. Cent. Eur. J. Immunol. 2019, 44, 181–189. [Google Scholar] [CrossRef] [PubMed]
  147. Liu, X.F.; Hao, J.L.; Xie, T.; Mukhtar, N.J.; Zhang, W.; Malik, T.H.; Lu, C.W.; Zhou, D.D. Curcumin, A Potential Therapeutic Candidate for Anterior Segment Eye Diseases: A Review. Front. Pharmacol. 2017, 8, 66. [Google Scholar] [CrossRef] [PubMed]
  148. Pinelli, R.; Ferrucci, M.; Biagioni, F.; Bumah, V.; Scaffidi, E.; Puglisi-Allegra, S.; Fornai, F. Curcumin as a Perspective Protection for Retinal Pigment Epithelium during Autophagy Inhibition in the Course of Retinal Degeneration. Curr. Neuropharmacol. 2023, 21, 2227–2232. [Google Scholar] [CrossRef]
  149. Sun, Y.; Wang, X.; Chen, B.; Huang, M.; Ma, P.; Xiong, L.; Huang, J.; Chen, J.; Huang, S.; Liu, Y. TFEB-Mediated Lysosomal Restoration Alleviates High Glucose-Induced Cataracts via Attenuating Oxidative Stress. Investig. Ophthalmol. Vis. Sci. 2022, 63, 26. [Google Scholar] [CrossRef]
  150. Josifovska, N.; Albert, R.; Nagymihály, R.; Lytvynchuk, L.; Moe, M.C.; Kaarniranta, K.; Veréb, Z.J.; Petrovski, G. Resveratrol as Inducer of Autophagy, Pro-Survival, and Anti-Inflammatory Stimuli in Cultured Human RPE Cells. Int. J. Mol. Sci. 2020, 21, 813. [Google Scholar] [CrossRef]
  151. Subirada, P.V.; Vaglienti, M.V.; Joray, M.B.; Paz, M.C.; Barcelona, P.F.; Sánchez, M.C. Rapamycin and Resveratrol Modulate the Gliotic and Pro-Angiogenic Response in Müller Glial Cells Under Hypoxia. Front. Cell Dev. Biol. 2022, 10, 855178. [Google Scholar] [CrossRef]
  152. Ye, M.J.; Meng, N. Resveratrol acts via the mitogen-activated protein kinase (MAPK) pathway to protect retinal ganglion cells from apoptosis induced by hydrogen peroxide. Bioengineered 2021, 12, 4878–4886. [Google Scholar] [CrossRef]
  153. Chen, P.; Yao, Z.; He, Z. Resveratrol protects against high glucose-induced oxidative damage in human lens epithelial cells by activating autophagy. Exp. Ther. Med. 2021, 21, 440. [Google Scholar] [CrossRef]
  154. Petrovski, G.; Berenyi, E.; Albert, R.; Moe, M.; Fesus, L.; Berta, A. Resveratrol, rapamycin and MG-132 as inducers of autophagy in ARPE-19 cells. Acta Ophthalmol. 2011, 89. [Google Scholar] [CrossRef]
  155. Wang, S.; Wang, Z.; Yang, S.; Yin, T.; Zhang, Y.; Qin, Y.; Weinreb, R.N.; Sun, X. Tissue Distribution of trans-Resveratrol and Its Metabolites after Oral Administration in Human Eyes. J. Ophthalmol. 2017, 2017, 4052094. [Google Scholar] [CrossRef] [PubMed]
  156. Hsu, M.Y.; Hsiao, Y.P.; Lin, Y.T.; Chen, C.; Lee, C.M.; Liao, W.C.; Tsou, S.C.; Lin, H.W.; Chang, Y.Y. Quercetin Alleviates the Accumulation of Superoxide in Sodium Iodate-Induced Retinal Autophagy by Regulating Mitochondrial Reactive Oxygen Species Homeostasis through Enhanced Deacetyl-SOD2 via the Nrf2-PGC-1α-Sirt1 Pathway. Antioxidants 2021, 10, 1125. [Google Scholar] [CrossRef] [PubMed]
  157. Zhao, L.; Wang, H.; Du, X. The therapeutic use of quercetin in ophthalmology: Recent applications. Biomed. Pharmacother. 2021, 137, 111371. [Google Scholar] [CrossRef] [PubMed]
  158. Sanderson, J.; McLauchlan, W.R.; Williamson, G. Quercetin inhibits hydrogen peroxide-induced oxidation of the rat lens. Free. Radic. Biol. Med. 1999, 26, 639–645. [Google Scholar] [CrossRef]
  159. Cornish, K.M.; Williamson, G.; Sanderson, J. Quercetin metabolism in the lens: Role in inhibition of hydrogen peroxide induced cataract. Free. Radic. Biol. Med. 2002, 33, 63–70. [Google Scholar] [CrossRef]
  160. Ferlemi, A.V.; Makri, O.E.; Mermigki, P.G.; Lamari, F.N.; Georgakopoulos, C.D. Quercetin glycosides and chlorogenic acid in highbush blueberry leaf decoction prevent cataractogenesis in vivo and in vitro: Investigation of the effect on calpains, antioxidant and metal chelating properties. Exp. Eye Res. 2016, 145, 258–268. [Google Scholar] [CrossRef]
  161. Bungau, S.; Abdel-Daim, M.M.; Tit, D.M.; Ghanem, E.; Sato, S.; Maruyama-Inoue, M.; Yamane, S.; Kadonosono, K. Health Benefits of Polyphenols and Carotenoids in Age-Related Eye Diseases. Oxid. Med. Cell Longev. 2019, 2019, 9783429. [Google Scholar] [CrossRef]
  162. Liu, X.; Chen, A.; Liang, Q.; Yang, X.; Dong, Q.; Fu, M.; Wang, S.; Li, Y.; Ye, Y.; Lan, Z.; et al. Spermidine inhibits vascular calcification in chronic kidney disease through modulation of SIRT1 signaling pathway. Aging Cell 2021, 20, e13377. [Google Scholar] [CrossRef]
  163. Ağaoğlu, N.B.; Varol, N.; Yıldız, S.H.; Karaosmanoğlu, C.; Duman, R.; Özdemir Erdoğan, M.; Solak, M. Relationship between SIRT1 gene expression level and disease in age-related cataract cases. Turk. J. Med. Sci. 2019, 49, 1068–1072. [Google Scholar] [CrossRef]
  164. Wu, F.; Xia, X.; Lei, T.; Du, H.; Hua, H.; Liu, W.; Xu, B.; Yang, T. Inhibition of SIRT1 promotes ultraviolet B induced cataract via downregulation of the KEAP1/NFE2L2 signaling pathway. J. Photochem. Photobiol. B Biol. 2023, 245, 112753. [Google Scholar] [CrossRef]
  165. Han, W.; Li, H.; Chen, B. Research Progress and Potential Applications of Spermidine in Ocular Diseases. Pharmaceutics 2022, 14, 1500. [Google Scholar] [CrossRef] [PubMed]
  166. Maekawa, S.; Hibasami, H.; Uji, Y.; Nakashima, K. Active transport and metabolic characteristics of polyamines in the rat lens. Biochim. Biophys. Acta (BBA)—Gen. Subj. 1989, 993, 199–203. [Google Scholar] [CrossRef]
  167. Kremzner, L.T.; Roy, D.; Spector, A. Polyamines in normal and cataractous human lenses: Evidence for post-translational modification. Exp. Eye Res. 1983, 37, 649–659. [Google Scholar] [CrossRef] [PubMed]
  168. Maekawa, S.; Hibasami, H.; Tsukada, T.; Furusako, S.; Nakashima, K.; Yokoyama, M. Induction of spermidine/spermine N1-acetyltransferase in needle-punctured rat lens as a model of traumatic cataract. Biochim. Biophys. Acta 1986, 883, 501–505. [Google Scholar] [CrossRef]
  169. Mischiati, C.; Feriotto, G.; Tabolacci, C.; Domenici, F.; Melino, S.; Borromeo, I.; Forni, C.; De Martino, A.; Beninati, S. Polyamine Oxidase Is Involved in Spermidine Reduction of Transglutaminase Type 2-Catalyzed βH-Crystallins Polymerization in Calcium-Induced Experimental Cataract. Int. J. Mol. Sci. 2020, 21, 5427. [Google Scholar] [CrossRef]
  170. Lentini, A.; Tabolacci, C.; Mattioli, P.; Provenzano, B.; Beninati, S. Spermidine delays eye lens opacification in vitro by suppressing transglutaminase-catalyzed crystallin cross-linking. Protein J. 2011, 30, 109–114. [Google Scholar] [CrossRef]
  171. Rao, Y.L.; Ganaraja, B.; Suresh, P.K.; Joy, T.; Ullal, S.D.; Manjrekar, P.A.; Murlimanju, B.V.; Sharma, B.G. Effect of resveratrol and combination of resveratrol and donepezil on the expression of microglial cells and astrocytes in Wistar albino rats of colchicine-induced Alzheimer’s disease. 3 Biotech 2023, 13, 319. [Google Scholar] [CrossRef]
  172. Yao, J.; Liu, X.; Sun, Y.; Dong, X.; Liu, L.; Gu, H. Curcumin-Alleviated Osteoarthritic Progression in Rats Fed a High-Fat Diet by Inhibiting Apoptosis and Activating Autophagy via Modulation of MicroRNA-34a. J. Inflamm. Res. 2021, 14, 2317–2331. [Google Scholar] [CrossRef]
  173. Jin, Z.; Chang, B.; Wei, Y.; Yang, Y.; Zhang, H.; Liu, J.; Piao, L.; Bai, L. Curcumin exerts chondroprotective effects against osteoarthritis by promoting AMPK/PINK1/Parkin-mediated mitophagy. Biomed. Pharmacother. 2022, 151, 113092. [Google Scholar] [CrossRef]
  174. Li, X.; Feng, K.; Li, J.; Yu, D.; Fan, Q.; Tang, T.; Yao, X.; Wang, X. Curcumin Inhibits Apoptosis of Chondrocytes through Activation ERK1/2 Signaling Pathways Induced Autophagy. Nutrients 2017, 9, 414. [Google Scholar] [CrossRef]
  175. Qin, N.; Wei, L.; Li, W.; Yang, W.; Cai, L.; Qian, Z.; Wu, S. Local intra-articular injection of resveratrol delays cartilage degeneration in C57BL/6 mice by inducing autophagy via AMPK/mTOR pathway. J. Pharmacol. Sci. 2017, 134, 166–174. [Google Scholar] [CrossRef] [PubMed]
  176. Darwish, M.A.; Abdel-Bakky, M.S.; Messiha, B.A.S.; Abo-Saif, A.A.; Abo-Youssef, A.M. Resveratrol mitigates pancreatic TF activation and autophagy-mediated beta cell death via inhibition of CXCL16/ox-LDL pathway: A novel protective mechanism against type 1 diabetes mellitus in mice. Eur. J. Pharmacol. 2021, 901, 174059. [Google Scholar] [CrossRef] [PubMed]
  177. Sacitharan, P.K.; Lwin, S.; Gharios, G.B.; Edwards, J.R. Spermidine restores dysregulated autophagy and polyamine synthesis in aged and osteoarthritic chondrocytes via EP300. Exp. Mol. Med. 2018, 50, 1–10. [Google Scholar] [CrossRef] [PubMed]
  178. D’Adamo, S.; Cetrullo, S.; Guidotti, S.; Silvestri, Y.; Minguzzi, M.; Santi, S.; Cattini, L.; Filardo, G.; Flamigni, F.; Borzì, R.M. Spermidine rescues the deregulated autophagic response to oxidative stress of osteoarthritic chondrocytes. Free Radic. Biol. Med. 2020, 153, 159–172. [Google Scholar] [CrossRef]
  179. Hosoda, R.; Nakashima, R.; Yano, M.; Iwahara, N.; Asakura, S.; Nojima, I.; Saga, Y.; Kunimoto, R.; Horio, Y.; Kuno, A. Resveratrol, a SIRT1 activator, attenuates aging-associated alterations in skeletal muscle and heart in mice. J. Pharmacol. Sci. 2023, 152, 112–122. [Google Scholar] [CrossRef]
  180. Fan, J.; Yang, X.; Li, J.; Shu, Z.; Dai, J.; Liu, X.; Li, B.; Jia, S.; Kou, X.; Yang, Y.; et al. Spermidine coupled with exercise rescues skeletal muscle atrophy from D-gal-induced aging rats through enhanced autophagy and reduced apoptosis via AMPK-FOXO3a signal pathway. Oncotarget 2017, 8, 17475–17490. [Google Scholar] [CrossRef]
  181. Khater, S.I.; Dowidar, M.F.; Abdel-Aziz, A.E.; Khamis, T.; Dahran, N.; Alqahtani, L.S.; Metwally, M.M.M.; Al-Hady Abd-Elrahamn, A.S.; Alsieni, M.; Alosaimi, M.E.; et al. β-Cell Autophagy Pathway and Endoplasmic Reticulum Stress Regulating-Role of Liposomal Curcumin in Experimental Diabetes Mellitus: A Molecular and Morphometric Study. Antioxidants 2022, 11, 2400. [Google Scholar] [CrossRef]
  182. Zhang, P.; Fang, J.; Zhang, J.; Ding, S.; Gan, D. Curcumin Inhibited Podocyte Cell Apoptosis and Accelerated Cell Autophagy in Diabetic Nephropathy via Regulating Beclin1/UVRAG/Bcl2. Diabetes Metab. Syndr. Obes. 2020, 13, 641–652. [Google Scholar] [CrossRef]
  183. Zhang, X.; Zhang, L.; Chen, Z.; Li, S.; Che, B.; Wang, N.; Chen, J.; Xu, C.; Wei, C. Exogenous spermine attenuates diabetic kidney injury in rats by inhibiting AMPK/mTOR signaling pathway. Int. J. Mol. Med. 2021, 47, 27. [Google Scholar] [CrossRef]
  184. Aman, Y.; Schmauck-Medina, T.; Hansen, M.; Morimoto, R.I.; Simon, A.K.; Bjedov, I.; Palikaras, K.; Simonsen, A.; Johansen, T.; Tavernarakis, N.; et al. Autophagy in healthy aging and disease. Nat. Aging 2021, 1, 634–650. [Google Scholar] [CrossRef]
  185. Pavlova, J.A.; Guseva, E.A.; Dontsova, O.A.; Sergiev, P.V. Natural Activators of Autophagy. Biochemistry 2024, 89, 1–26. [Google Scholar] [PubMed]
  186. Tabibzadeh, S. Role of autophagy in aging: The good, the bad, and the ugly. Aging Cell 2023, 22, e13753. [Google Scholar] [CrossRef] [PubMed]
  187. Qin, S.; Tang, H.; Li, W.; Gong, Y.; Li, S.; Huang, J.; Fang, Y.; Yuan, W.; Liu, Y.; Wang, S.; et al. AMPK and its Activator Berberine in the Treatment of Neurodegenerative Diseases. Curr. Pharm Des. 2020, 26, 5054–5066. [Google Scholar] [CrossRef] [PubMed]
  188. DiNicolantonio, J.J.; McCarty, M.F.; Assanga, S.I.; Lujan, L.L.; O’Keefe, J.H. Ferulic acid and berberine, via Sirt1 and AMPK, may act as cell cleansing promoters of healthy longevity. Open Heart 2022, 9, e001801. [Google Scholar] [CrossRef]
  189. Ren, G.; Guo, J.H.; Qian, Y.Z.; Kong, W.J.; Jiang, J.D. Berberine Improves Glucose and Lipid Metabolism in HepG2 Cells Through AMPKα1 Activation. Front. Pharmacol. 2020, 11, 647. [Google Scholar] [CrossRef]
  190. Su, X.; Yang, D.; Hu, Y.; Yuan, Y.; Song, L. Berberine suppressed sarcopenia insulin resistance through SIRT1-mediated mitophagy. Open Life Sci. 2023, 18, 20220648. [Google Scholar] [CrossRef]
  191. Dang, Y.; An, Y.; He, J.; Huang, B.; Zhu, J.; Gao, M.; Zhang, S.; Wang, X.; Yang, B.; Xie, Z. Berberine ameliorates cellular senescence and extends the lifespan of mice via regulating p16 and cyclin protein expression. Aging Cell 2020, 19, e13060. [Google Scholar] [CrossRef]
  192. Cai, Y.; Xin, Q.; Lu, J.; Miao, Y.; Lin, Q.; Cong, W.; Chen, K. A New Therapeutic Candidate for Cardiovascular Diseases: Berberine. Front. Pharmacol. 2021, 12, 631100. [Google Scholar] [CrossRef]
  193. FDA Curcumin GRN No. 822. Available online: https://www.cfsanappsexternal.fda.gov/scripts/fdcc/index.cfm?set=GRASNotices&id=822&sort=GRN_No&order=DESC&startrow=1&type=basic&search=822 (accessed on 20 September 2024).
  194. FDA Quercetin GRN No. 341. Available online: https://www.cfsanappsexternal.fda.gov/scripts/fdcc/?set=GRASNotices&id=341&sort=GRN_No&order=DESC&startrow=1&type=basic&search=quercetin (accessed on 20 September 2024).
  195. Barber-Chamoux, N.; Milenkovic, D.; Verny, M.A.; Habauzit, V.; Pereira, B.; Lambert, C.; Richard, D.; Boby, C.; Mazur, A.; Lusson, J.R.; et al. Substantial Variability Across Individuals in the Vascular and Nutrigenomic Response to an Acute Intake of Curcumin: A Randomized Controlled Trial. Mol. Nutr. Food Res. 2018, 62, 1700418. [Google Scholar] [CrossRef]
  196. Mirza, M.A.; Mahmood, S.; Hilles, A.R.; Ali, A.; Khan, M.Z.; Zaidi, S.A.A.; Iqbal, Z.; Ge, Y. Quercetin as a Therapeutic Product: Evaluation of Its Pharmacological Action and Clinical Applications-A Review. Pharmaceuticals 2023, 16, 1631. [Google Scholar] [CrossRef]
  197. Tomé-Carneiro, J.; Gonzálvez, M.; Larrosa, M.; Yáñez-Gascón, M.J.; García-Almagro, F.J.; Ruiz-Ros, J.A.; Tomás-Barberán, F.A.; García-Conesa, M.T.; Espín, J.C. Grape resveratrol increases serum adiponectin and downregulates inflammatory genes in peripheral blood mononuclear cells: A triple-blind, placebo-controlled, one-year clinical trial in patients with stable coronary artery disease. Cardiovasc. Drugs Ther. 2013, 27, 37–48. [Google Scholar] [CrossRef] [PubMed]
  198. Schwarz, C.; Stekovic, S.; Wirth, M.; Benson, G.; Royer, P.; Sigrist, S.J.; Pieber, T.; Dammbrueck, C.; Magnes, C.; Eisenberg, T.; et al. Safety and tolerability of spermidine supplementation in mice and older adults with subjective cognitive decline. Aging (Albany NY) 2018, 10, 19–33. [Google Scholar] [CrossRef] [PubMed]
  199. Quagliariello, V.; Iaffaioli, R.V.; Armenia, E.; Clemente, O.; Barbarisi, M.; Nasti, G.; Berretta, M.; Ottaiano, A.; Barbarisi, A. Hyaluronic Acid Nanohydrogel Loaded With Quercetin Alone or in Combination to a Macrolide Derivative of Rapamycin RAD001 (Everolimus) as a New Treatment for Hormone-Responsive Human Breast Cancer. J. Cell Physiol. 2017, 232, 2063–2074. [Google Scholar] [CrossRef] [PubMed]
  200. Alayev, A.; Berger, S.M.; Kramer, M.Y.; Schwartz, N.S.; Holz, M.K. The combination of rapamycin and resveratrol blocks autophagy and induces apoptosis in breast cancer cells. J. Cell Biochem. 2015, 116, 450–457. [Google Scholar] [CrossRef]
  201. Jin, H.G.; Wu, G.Z.; Wu, G.H.; Bao, Y.G. Combining the mammalian target of rapamycin inhibitor, rapamycin, with resveratrol has a synergistic effect in multiple myeloma. Oncol. Lett. 2018, 15, 6257–6264. [Google Scholar] [CrossRef]
  202. Shui, L.; Wang, W.; Xie, M.; Ye, B.; Li, X.; Liu, Y.; Zheng, M. Isoquercitrin induces apoptosis and autophagy in hepatocellular carcinoma cells via AMPK/mTOR/p70S6K signaling pathway. Aging 2020, 12, 24318–24332. [Google Scholar] [CrossRef]
  203. Dehzad, M.J.; Ghalandari, H.; Nouri, M.; Askarpour, M. Antioxidant and anti-inflammatory effects of curcumin/turmeric supplementation in adults: A GRADE-assessed systematic review and dose-response meta-analysis of randomized controlled trials. Cytokine 2023, 164, 156144. [Google Scholar] [CrossRef]
  204. Li, F.; Liu, J.; Tang, S.; Yan, J.; Chen, H.; Li, D.; Yan, X. Quercetin regulates inflammation, oxidative stress, apoptosis, and mitochondrial structure and function in H9C2 cells by promoting PVT1 expression. Acta Histochem. 2021, 123, 151819. [Google Scholar] [CrossRef]
  205. Hussain, T.; Tan, B.; Ren, W.; Rahu, N.; Dad, R.; Kalhoro, D.H.; Yin, Y. Polyamines: Therapeutic perspectives in oxidative stress and inflammatory diseases. Amino Acids 2017, 49, 1457–1468. [Google Scholar] [CrossRef]
Figure 1. The autophagy process. The initiation stage involves the participation of protein complexes such as ULK-Atg1 (PDBID: 6QAS), Class III PI3-K complex ATG9, Rab1, TRAPPIII, and ubiquitin-like molecules (LC3A-D) (PDBID: 2Z0E). In the nucleation stage, the membrane begins to expand by recruiting proteins and lipids, transforming into a primary double membrane sequestering compartment structure called a phagophore. In this stage, the most important event is the recruiting of the proteins for the ATG14-Class III PI3-K complex (PDBID: 8SOR). During the expansion and elongation process, the membrane of the phagophore expands and closes around its cargo to form the autophagosome; also, the objective of this stage is to determine the site of lipidation of LC3. The closure and fusion stage is characterized by the maturation of the autophagosomes and their fusion with mature lysosomes to become autolysosomes, regulated by the SNARE complex (PDBID: 1KIL). Finally, in the degradation stage, macromolecules contained in the autolysosome are degraded by lysosomal hydrolases into monomeric units and returned to the cytoplasm to be recycled; in addition, the process is modulated by activation of signal pathways as AMPK (PDBID: 4CFF), which in turn is modulated by Sirt1 (PDBID: 5BTR).
Figure 1. The autophagy process. The initiation stage involves the participation of protein complexes such as ULK-Atg1 (PDBID: 6QAS), Class III PI3-K complex ATG9, Rab1, TRAPPIII, and ubiquitin-like molecules (LC3A-D) (PDBID: 2Z0E). In the nucleation stage, the membrane begins to expand by recruiting proteins and lipids, transforming into a primary double membrane sequestering compartment structure called a phagophore. In this stage, the most important event is the recruiting of the proteins for the ATG14-Class III PI3-K complex (PDBID: 8SOR). During the expansion and elongation process, the membrane of the phagophore expands and closes around its cargo to form the autophagosome; also, the objective of this stage is to determine the site of lipidation of LC3. The closure and fusion stage is characterized by the maturation of the autophagosomes and their fusion with mature lysosomes to become autolysosomes, regulated by the SNARE complex (PDBID: 1KIL). Finally, in the degradation stage, macromolecules contained in the autolysosome are degraded by lysosomal hydrolases into monomeric units and returned to the cytoplasm to be recycled; in addition, the process is modulated by activation of signal pathways as AMPK (PDBID: 4CFF), which in turn is modulated by Sirt1 (PDBID: 5BTR).
Cells 13 01611 g001
Figure 2. Natural autophagy activators in age-related diseases. On the left: natural compounds (curcumin, resveratrol, quercetin, and polyamines such as spermidine) that can induce autophagy in the context of age-related diseases. On the right: main regulatory targets of autophagy that, when activated, enhance autophagic flux, which is often diminished in cardiovascular diseases, neurodegenerative disorders, and cataracts.
Figure 2. Natural autophagy activators in age-related diseases. On the left: natural compounds (curcumin, resveratrol, quercetin, and polyamines such as spermidine) that can induce autophagy in the context of age-related diseases. On the right: main regulatory targets of autophagy that, when activated, enhance autophagic flux, which is often diminished in cardiovascular diseases, neurodegenerative disorders, and cataracts.
Cells 13 01611 g002
Table 1. Summary table of natural compounds and their autophagic effect on ARDs.
Table 1. Summary table of natural compounds and their autophagic effect on ARDs.
Age-Related DiseasesNatural CompoundAutophagic EffectRef.
CardiopathiesCurcumin Attenuation of oxidative stress and activation of autophagy via the AKT/mTOR regulation in diabetic rats with myocardial damage.[52]
Restoration of Beclin-1 and LC3 I/II ratio in mice with doxorubicin-induced cardiomyopathy.[54]
ResveratrolAmelioration of cardiac dysfunction of neonatal rat ventricular myocytes via the negative regulation of the PI3K/Akt pathway and decreasing of apoptosis. [66]
Activation of TFEB for autophagy activation. [67]
Increased the expression of LC3 and Beclin-1 via Sirt1on endothelial oxidative damage of cardiovascular models. [68]
QuercetinActivation of the AMPK pathway that leads to increased LC3 and Beclin-1 protein levels to ameliorate myocardial injury.[79]
Suppression of the PI3K/Akt pathway to attenuate myocardial ischemia.[80]
Prevention of myocardial fibrosis via the regulation of the miR-223/FOXO3 that leads to increasing LC3 II/I ratio. [81,82]
PolyaminesPrevention of lipidic plaque formation via increased expression of LC3 I/II ratio.[94]
Amelioration of myocardial dysfunction by promoting AMPK/mTOR pathway.[95]
Alzheimer’s diseaseCurcumin Reverse cognitive disorders in an AD mice model induced by cisplatin by the activation of the AMPK-JNK signaling pathway to inhibit mTOR and Bcl-2, which modulates autophagy by the overexpression of LC3 and downregulation of p62.[106]
ResveratrolEnhance cognitive function in rats by stimulating the AKT/mTOR pathway.[109]
Activation of the Sirt1 pathway, leading to increased mitophagy and subsequent neuroprotection. [171]
Improvements in sleep and memory dependent on the ortholog Sir2, a sirtuin ortholog that stimulates autophagy by activating HSP70 and ATG4 while it also interacts with LC3 and p62. [111,112]
QuercetinActivates Sirt1 pathway, influencing the expression of FOXO1 and NRF2. [118,119]
Reduction of the Ab1 kinase expression, preventing tau phosphorylation and neuronal apoptosis triggered by Aβ.[122,123]
Research on a PC12 cell model showed quercetin improves cell survival and proliferation, mitigates Aβ toxicity, and enhances the expression of Sirt1. [125,126]
PolyaminesElevates LC3 and Beclin-1 levels compared to control in SY5Y cells stably expressing a mutant form of human tau protein. [90]
Spermidines induce autophagy and clear amyloid precursor protein clusters. [135]
Parkison’s diseaseCurcuminIncreases the expression of LC3-II, facilitating enhanced clearance of α-synuclein proteins in mice models.[107]
ResveratrolIn mice model, ameliorates the deficit in cognitive and motor function in a dose-dependent manner along with a decrease in α-synuclein aggregation. [114]
In mice model, autophagic degradation of α-synuclein was promoted via the activation of the Sirt1 pathway. [116]
In two cell models, resveratrol promoted autophagy by increasing beclin-1 and the LC3-II/LC3-I ratio, and the reduction in p62.[117]
QuercetinAmeliorates neuronal death, mitochondrial dysfunction, and α-synuclein accumulation in animal and cell models by activating autophagy. [127]
Increase of ULK1(Ser555)/ULK1, p-TBK1(Ser172)/TBK1, and LC3 levels. [128]
Increase autophagy and attenuate behavioral impairments. [129]
PolyaminesAutophagy stimulation[90]
Ocular disordersCurcumin* Activation of TFEB to promote lysosomal biogenesis on rats with glucose-induced cataracts.[149]
ResveratrolIncreased expression of LC3 and Beclin-1 in high glucose treated HLEC cells.[153]
PolyaminesDownregulation of Sirt1 in age-related cataracts may be indirectly linked to altered polyamine (PA) levels in the human lens. Spermidine, known to positively regulate Sirt1, could play a role in this process.[163,164]
* Curcumin analog C1.
Table 2. Natural autophagy activators in other ARDs.
Table 2. Natural autophagy activators in other ARDs.
Age-Related DiseaseNatural CompoundAutophagic EffectRef.
OsteoarthritisCurcumin





Resveratrol

Polyamines (Spermidine)
Reduction of miR-34a expression and probably through the Akt/mTOR pathway[172]
mTOR via the PI3K-Akt pathway[172]
Mitophagy AMPK/PINK1/Parkin pathway[173]
Activation of MAPK/ERK1/2 pathway[174]
AMPK/mTOR pathway[175]
Inhibition of CXCL16/ox-LDL pathway[176]
Overexpression of EP300 acetyltransferase[177]
Overexpression of BECN-1, LC3-II and p62[178]
Sarcopenia Resveratrol
Polyamines (Spermidine)
SIRT1[179]
Probably AMPK-FOXO3a[180]
DiabetesCurcumin



Polyamines (Spermidine)
In β cells miR-29b and miR-137 overexpression[181]
In podocyte via regulating Beclin1/UVRAG/Bcl2[182]
In podocyte regulate the AMPK/mTOR pathway[183]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Mundo Rivera, V.M.; Tlacuahuac Juárez, J.R.; Murillo Melo, N.M.; Leyva Garcia, N.; Magaña, J.J.; Cordero Martínez, J.; Jiménez Gutierrez, G.E. Natural Autophagy Activators to Fight Age-Related Diseases. Cells 2024, 13, 1611. https://doi.org/10.3390/cells13191611

AMA Style

Mundo Rivera VM, Tlacuahuac Juárez JR, Murillo Melo NM, Leyva Garcia N, Magaña JJ, Cordero Martínez J, Jiménez Gutierrez GE. Natural Autophagy Activators to Fight Age-Related Diseases. Cells. 2024; 13(19):1611. https://doi.org/10.3390/cells13191611

Chicago/Turabian Style

Mundo Rivera, Vianey M., José Roberto Tlacuahuac Juárez, Nadia Mireya Murillo Melo, Norberto Leyva Garcia, Jonathan J. Magaña, Joaquín Cordero Martínez, and Guadalupe Elizabeth Jiménez Gutierrez. 2024. "Natural Autophagy Activators to Fight Age-Related Diseases" Cells 13, no. 19: 1611. https://doi.org/10.3390/cells13191611

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

Article Metrics

Back to TopTop