Hostname: page-component-848d4c4894-ndmmz Total loading time: 0 Render date: 2024-06-06T09:08:51.969Z Has data issue: false hasContentIssue false

Autophagic mechanisms in longevity intervention: role of natural active compounds

Published online by Cambridge University Press:  30 March 2023

Kevser Taban Akça
Affiliation:
Department of Pharmacognosy, Faculty of Pharmacy, Gazi University, Ankara, Türkiye
İlknur Çınar Ayan
Affiliation:
Department of Medical Biology, Medical Faculty, Necmettin Erbakan University, Meram, Konya, Türkiye
Sümeyra Çetinkaya
Affiliation:
Biotechnology Research Center of Ministry of Agriculture and Forestry, Yenimahalle, Ankara, Türkiye
Ece Miser Salihoğlu
Affiliation:
Biochemistry Department, Faculty of Pharmacy, Gazi University, Ankara, Türkiye
İpek Süntar*
Affiliation:
Department of Pharmacognosy, Faculty of Pharmacy, Gazi University, Ankara, Türkiye
*
Corresponding author: İpek Süntar; Email: ipesin@gazi.edu.tr
Rights & Permissions [Opens in a new window]

Abstract

The term ‘autophagy’ literally translates to ‘self-eating’ and alterations to autophagy have been identified as one of the several molecular changes that occur with aging in a variety of species. Autophagy and aging, have a complicated and multifaceted relationship that has recently come to light thanks to breakthroughs in our understanding of the various substrates of autophagy on tissue homoeostasis. Several studies have been conducted to reveal the relationship between autophagy and age-related diseases. The present review looks at a few new aspects of autophagy and speculates on how they might be connected to both aging and the onset and progression of disease. Additionally, we go over the most recent preclinical data supporting the use of autophagy modulators as age-related illnesses including cancer, cardiovascular and neurodegenerative diseases, and metabolic dysfunction. It is crucial to discover important targets in the autophagy pathway in order to create innovative therapies that effectively target autophagy. Natural products have pharmacological properties that can be therapeutically advantageous for the treatment of several diseases and they also serve as valuable sources of inspiration for the development of possible new small-molecule drugs. Indeed, recent scientific studies have shown that several natural products including alkaloids, terpenoids, steroids, and phenolics, have the ability to alter a number of important autophagic signalling pathways and exert therapeutic effects, thus, a wide range of potential targets in various stages of autophagy have been discovered. In this review, we summarised the naturally occurring active compounds that may control the autophagic signalling pathways.

Type
Review
Creative Commons
Creative Common License - CCCreative Common License - BY
This is an Open Access article, distributed under the terms of the Creative Commons Attribution licence (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted re-use, distribution and reproduction, provided the original article is properly cited.
Copyright
Copyright © The Author(s), 2023. Published by Cambridge University Press

Introduction

Aging is a biological process that causes the organism's quality of life to diminish over time due to time-dependent cellular and functional degradation. Age-related conditions as a group pose a serious socioeconomic burden on the world and a substantial healthcare concern. Finding treatment approaches that slow the deterioration of numerous age-related pathological disorders is therefore crucial (Ref. Reference Aman1).

The incidence of various diseases such as cancer, cardiovascular diseases, neurodegenerative diseases (NDs) and metabolic dysfunction rises with aging (Ref. Reference Menzies2). Above-mentioned illnesses can be brought on by dysregulation of cellular autophagy. Autophagy is a quite preserved catabolic cellular process that expresses lysosomal degradation and recycling of nucleic acids, proteins, lipids and other intracellular components. Autophagy mechanism is associated with homoeostasis, differentiation, development and survival processes, as well as defence against pathogens and conservation of cellular energy. Decreased autophagic activity in aging affects a variety of cellular and molecular processes and results in the accumulation of damaged macromolecules and organelles. Therefore, it has been suggested that maintenance of regular autophagic activity is associated with longevity (Ref. Reference Ruckenstuhl3).

Autophagy functions as both a tumour suppressor and promoter (through protective autophagy) in cancer. Furthermore, it plays a crucial role in preventing cardiac aging and maintaining neuronal homoeostasis. Early regulation of the formation of the mammalian heart, proper maintenance of cardiac structure and function, and the beginning and progression of cardiovascular disorders are all regulated by autophagic activity (Ref. Reference Gatica4). In addition to protein aggregation, NDs have dysregulated autophagy along with mitochondrial dysfunction and reduced lysosomal activity. The prevalence of NDs like Alzheimer's disease (AD), Parkinson's disease (PD), and Huntington's disease (HD) increases with aging and neuronal autophagy dysregulation (Ref. Reference Tan5). Moreover, in metabolic illnesses including obesity and diabetes, autophagy is suppressed or increased (Ref. Reference Jaishy6). Studies have demonstrated the significance of the signalling pathways PI3K/AKT/mTOR, AMP-activated protein kinase (AMPK), MAPK, SIRT1, and FoXO in the control of autophagy, cell aging, and metabolic diseases.

There are many synthetic compounds that have been discovered that modify autophagy and are good candidates for the therapy of cancer, but they come with unfavourable side effects. As a result, several phytochemicals have drawn a lot of interest for their potential to be used as autophagy modulators with minimum adverse effects (Ref. Reference Rahman7). Indeed, naturally occurring compounds including, alkaloids, terpenoids, flavonoids, phenylpropanoids, lignans, phenolic acids, stilbenes, tannins, senevol glycosides, steroids, lectins have been found to be powerful autophagy inducers or inhibitors, opening up new treatment regimens.

In the present review new aspects of autophagy and speculations on how they might be connected to both aging and the onset and progression of age-related diseases are discussed. We also aim to summarise the natural compounds that can be used as autophagy modulators. Eventually, developing practical applications that support long-term health will be made easier with a better understanding of the specific interactions between autophagy and the risk of age-related diseases across organisms.

Autophagy and aging

Three different types of autophagy have been described; macroautophagy; is the basic mechanism of autophagy and occurs when larger particles and organelles are taken into the autophagosome and fused with lysosomes, microautophagy; small particles are taken into the lysosome with the indentation in the membrane and degraded, chaperone-mediated autophagy (CMA); the recognition and transport of cytosolic proteins with a certain peptide sequence by Hsp73, which is complex with molecular chaperones, to lysosomes and this type of degradation pathway does not require vesicular traffic (Refs Reference Majeski8, Reference Kroemer9). On the other hand, selective forms of macroautophagy and microautophagy in which the degradation process occurs due to either the binding of HSC70 to the hydrophobic residues exposed in misfolded or aggregated proteins exists, namely chaperone-assisted selective autophagy (CASA) or the sequence-mediated targeting of proteins by HSC70, namely endosomal microautophagy (eMI) (Refs Reference Tekirdag and Cuervo10, Reference Vitale11).

The morphology of autophagy was first described in mammalian cells in the 1950s, and 31 autophagy-related (ATG) genes involved in the basic mechanism in autophagy were identified (Refs Reference Huang12, Reference Klionsky13). Most yeast genes encoding autophagy proteins are orthologous to the Caenorhabditis elegans genome (ATG2, ATG3, ATG4, ATG5, ATG7, ATG9, ATG10, ATG12, ATG18 genes), but there are two homologues of ATG4, ATG8 and ATG16. The ATG proteins encoded by these genes act at different stages of autophagosome biogenesis such as initiation of autophagy (unc-51/ATG1), vesicle nucleation (bec-1/ATG6, vps-34/VPS34), protein conjugation system (ATG-7/M7.5/ATG7, lgg-1/ATG8, lgg-3/ATG12), uptake and vesicle transformation (ATG-18/F41E6.13/ATG18) (Ref. Reference Kovacs14).

Macroautophagy can be induced in the presence of various stress factors such as hypoxia, starvation, presence of damaged protein or organelles. This type of autophagy involves 5-steps including initiation, nucleation, elongation, fusion and degradation. In the initiation stage, inhibition of mTOR, the master regulator of autophagy, leads to activation of autophagy. The nucleation stage involving the ULK1 complex (ULK1/2, ATG13 and FIP200) and the class III PI3K complex (Beclin 1, ATG14L, p150) participates in phagophore formation. The phagophore membrane is closed via the ATG12 conjugation system (ATG12, ATG5 and ATG16L) and the LC3-II conjugation system in the elongation stage. This structure, called the autophagosome, contains the autophagic cargo. Finally, in the degradation stage, the autophagosome fuses with the lysosomal membrane and the autophagic cargo is sequestered by lysosomal hydrolases (Refs Reference Karampa15, Reference Hu and Reggiori16, Reference Sanchez-Mirasierra17). In macroautophagy and microautophagy (also called endosomal microautophagy in mammals), cargo transport to the lysosome is vesicle-mediated, whereas in CMA it occurs through specific receptors (Refs Reference Scrivo18, Reference McCarty19). In CASA, which is one of the selective macroautophagy types, polyubiquitinated aggregate proteins are transmitted to the autophagosome by helper chaperone Bcl2-related athanogen 3 (Bag3) and cytosolic chaperone heat shock cognate 70 kDa protein (HSC70) without binding to a pentapeptide amino acid motif (KFERQ)-like motif (Ref. Reference Scrivo18). CMA is a variant of autophagy defined only for proteins carrying a specific targeting motif in the KFERQ amino acid sequence. The KFERQ-like motif is recognised by the cytosolic chaperone HSC70 and translocates to the substrate lysosome by the lysosome-associated membrane protein type A (LAMP2A) receptor. CASA is distinct from CMA, by being ubiquitin-independent (Fig. 1) (Refs Reference Massey, Zhang and Cuervo20, Reference Arndt21).

Figure 1. The types of autophagy (Figure created with Biorender.com).

The processes of autophagy are related to aging in terms of biochemical regulation of cells. It has been reported that the decrease in gene expression of ATG5, ATG7, and Beclin-1 with age in humans is a result of decreased stimulation of macroautophagy (Ref. Reference Lipinski22). Activation of macroautophagy as a result of disruption in the Beclin-1-BCL2 complex prolongs the lifespan of mice (Ref. Reference Fernández23). While the impairment of macroautophagy in cells such as neurons, melanocytes, and fibroblasts increases aging (Ref. Reference Moreno-Blas24), chemical regulation of macroautophagy in tendon stem cells decreases senescence (Ref. Reference Nie25). Although the mechanism of microautophagy is well elucidated, the information on the relationship between microautophagy and aging is quite insufficient. One of the limited studies has shown that the accumulation of protein and lipid peroxidation products via microautophagy increases with aging (Ref. Reference Cannizzo26). The accumulation and aggregation of proteins known as CMA substrates are associated with the pathogenesis of aging and its accompanying diseases. Studies have shown that an increase in LAMP2A levels improves the reduction in CMA associated with aging (Ref. Reference Valdor27). Moreover, in previous preclinical studies, impaired CASA was shown to cause Z disc disintegration and progressive muscle weakness in flies, mice, and human, revealing the importance of chaperone-assisted degradation for the preservation of cellular structures of muscle tissues, thereby shedding light on aging (Ref. Reference Arndt21).

Hallmarks of aging

Hallmarks of aging are mainly loss of proteostasis, mitochondrial dysfunction, disordered nutrient perception, genomic instability, telomere attrition, cellular senescence, cellular stemness.

The role of autophagy in the loss of proteostasis

During the aging process, a decrease in the proteostasis activity of many cells and tissues is observed. Loss of proteostasis, a key feature of aging, is characterised by the appearance of misfolded or aggregated proteins. On the other hand, autophagy is capable of detecting proteotoxic stress and developing an appropriate response. Autophagy, which plays an important role in removing misfolded or unfolded protein aggregates, is impaired with age. With this degradation, protein aggregates accumulate and cause proteotoxicity (Ref. Reference Di Domenico28).

Autophagy in mitochondrial dysfunction

The reduced mitophagy (known as clearing damaged mitochondria) associated with aging in mitochondria also leads to decreased energy production. Autophagy plays a key role in cleaning damaged mitochondria. Dysregulation in the autophagy mechanism causes mitochondria accumulation and thus oxidative stress (Ref. Reference Um and Yun29).

Autophagy and cellular stemness

Autophagy plays a major role in the ability of adult stem cells to self-renew and differentiate into specialised cells. The decrease in autophagic activity with age results in a decrease in stem cells (Ref. Reference Boya, Codogno and Rodriguez-Muela30).

Autophagy and senescence

Oxidative stress, DNA damage, telomere shortening lead to cellular senescence. While autophagy over-stimulation induces cell death, its deficiency can trigger cellular senescence (Ref. Reference Rajendran31).

Autophagy and genomic instability

UV light, toxic chemicals, agents such as reactive oxygen species (ROS), mutations in nuclear and mitochondrial genes cause DNA damage. Weakening of the DNA repair mechanism and accumulating DNA damage trigger cellular senescence (Ref. Reference Hewitt and Korolchuk32).

Autophagy and telomere attrition

Damage to telomeres or a decrease in their activity is directly related to the aging process (Ref. Reference Lopez-Otín33). An increase in cytoplasmic vacuoles and autophagy-related ATG5, ATG12 and LC3 proteins was observed in telomere-defected cells (Ref. Reference Cheon34).

Molecular mechanisms of autophagy

Many signalling pathways have been identified to be responsible for regulating longevity and aging. Studies have shown that PI3K/AKT/mTOR, AMP-activated protein kinase (AMPK), MAPK, SIRT1, FoXO, signalling pathways are important in the regulation of autophagy as well as cell aging and metabolic disorders.

The PI3K/AKT/mTOR axis induced by growth factors is an essential signalling pathway in cancer cell growth in nutrient-rich conditions. In nutrient deficient conditions, inhibition of this pathway promotes autophagy (Ref. Reference Fairlie, Tran and Lee35). TOR is an evolutionarily conserved serine/threonine kinase mainly localised in cytoplasm that regulates various cellular functions, including cell metabolism, survival, cytoskeleton, and autophagy (Ref. Reference Ge36). mTOR is encoded by a single gene and there are two types of mTOR complexes as the protein product, functionally and structurally: mTOR complex 1 (mTORC1) sensitive to rapamycin and mTOR complex 2 (mTORC2) insensitive to rapamycin (Ref. Reference Bhaskar and Hay37). mTORC1 consists of mTOR, raptor (regulatory-associated protein of mTOR), mLST8 (mammalian lethal with sec-13 protein 8), PRAS40 (proline-rich AKT substrate 40 kDa), Deptor (the DEP domain containing mTOR-interacting protein), and TTI1/TEL2 (Ref. Reference Papadopoli38). mTORC1 is responsible for negative regulation of cell growth, differentiation and autophagy. Phosphorylation of mTORC1 inactivates ULK/ATG1, which promotes autophagy, increases anabolism, and promotes cell growth. Rapamycin is a well-known inhibitor of mTOR and also an inducer of macroautophagy. The mTORC2 consists of mTOR, mLST8, rictor, mSIN1, protor, and Deptor subunits. mTORC2 is mainly involved in the insulin signalling pathway and indirectly regulates autophagy (Ref. Reference Wang39). The activity of mTORC1 is regulated by the PI3K/AKT/mTOR signalling pathway as well as by the AMPK and Ras-dependent MAPK pathways (Ref. Reference Zada40). It was shown that regulation of autophagy dependent on the AMPK-mTOR signalling pathway can delay cardiomyocyte senescence (Ref. Reference Chen41). It was also determined that overactivation of mTOR leads to impaired autophagy and results in premature aging (Ref. Reference Pan42). ULK1, one of the important autophagy proteins, is a serine/threonine kinase and cellular autophagy is triggered by ULK1 binding to ATG13, FIP200 and ATG101 proteins to form a complex (Ref. Reference Zachari and Ganley43). Under normal energy conditions, mTORC1 phosphorylates ULK1 and AMPK cannot activate ULK1. Cell autophagy is inhibited because it cannot form complexes with inactive ULK1, ATG and FIP200. In the absence of nutrients in cells, AMPK is activated, mTOR is inhibited, and autophagy is activated (Ref. Reference Turco, Fracchiolla and Martens44). Consequently, the activated mTOR pathway inhibits the activation of ULK1 to induce senescence by inhibiting autophagy. When AMPK, the negative regulator of mTOR, is activated, it either activates ULK1 or inhibits mTOR and can delay aging by initiating autophagy.

SIRT1 is the mammalian homologue of Sir2 (silent information regulator) protein, known as stress resistance and longevity factor in yeasts. SIRT1 is the most studied sirtuin of seven human sirtuins (Ref. Reference Kitada, Ogura and Koya45). SIRT1 is a class III protein deacetylase and uses nicotinamide adenine dinucleotide (NAD) as a cofactor to deacetylase lysine residues, thereby silencing genes, which can delay aging, reduce inflammation, improve energy metabolism. It was revealed that SIRT1 can form complexes with autophagy-related ATG5, ATG7, LC3 proteins and regulate autophagy by causing their direct deacetylation (Ref. Reference Lee46). Calorie restriction delays the onset of age-related diseases such as NDs, cardiovascular diseases, and diabetes, therefore it is the only strategy to extend lifespan in many organisms (Ref. Reference Colman47). Although the mechanism of extension of lifespan is unclear, it has also been determined that autophagy is increased during calorie restriction and energy deficiency (Ref. Reference Hansen48). SIRT1 level in humans decreases with age, therefore it causes cellular proteins misfolding and cell damage. SIRT1 and AMPK are positive regulators of each other. NAD expression is increased by AMPK and activates SIRT1 (Ref. Reference Liu49). Calorie restriction also induces activation of SIRT1 with an anti-aging effect. The FoXO family of transcription factors (FoXO1, FoXO3a, FoXO4 and FoXO6) is also an important regulator of cellular metabolism and proliferation (Ref. Reference Fahy50). Recent studies have shown that signalling pathways related to SIRT1 and FoXO3 have an important role in the regulation of autophagy. SIRT1-mediated activation of FoXO1 induces expression of autophagosomes, and Rab7 (GTPase) which is highly important in autophagosome formation (Ref. Reference Ao, Zou and Wu51). Overexpression of Rab7 also triggers autophagy, and activated FoXO3 protein increases the expression of autophagy-related proteins (LC3 and BNIP3) (Fig. 2) (Ref. Reference Salminen and Kaarniranta52).

Figure 2. The molecular signalling mechanisms of autophagy (Figure created with Biorender. com).

Autophagy and age-related diseases

Cellular components that have been implicated in the aging process have been linked to progressively impaired autophagy. In the elderly, dysfunctional autophagy may play a role in age-related disorders such as cancer, cardiovascular and neurological diseases and metabolic syndrome. Thus, restoring defective autophagy to its healthy state may aid in preventing age-related diseases and boosting longevity (Ref. Reference Cheon34).

Autophagy in cancer

Cancer occurs when the balance between cell proliferation and death is lost, and more cells multiply than die or differentiate (Ref. Reference Mak53). Unlike apoptosis, autophagy helps maintain homoeostasis by reversing intracellular molecules in the absence of nutrients or in cellular stress (Ref. Reference Anding and Baehrecke54). Basal autophagy works as a tumour suppressor mechanism during tumorigenesis; excessive autophagy works as a survival pathway in certain cancers (Ref. Reference Yu, Chen and Tooze55). Unlike normal cells, basal levels of autophagy are high in tumour cells, stimulated in hypoxic regions, and provide a constitutive survival advantage (Ref. Reference Ding, Chen and Yin56). Therefore, inhibition of autophagy may prevent the survival of tumour cells and may be a new target for cancer therapy (Ref. Reference Maycotte and Thorburn57) (Table 1).

Table 1. Studies on autophagy in cancer

Autophagy is used to provide components and energy to cells for basal survival in each cell (Ref. Reference Anding and Baehrecke54). Autophagy is controlled by class I PI3K/mTOR and AMPK signalling pathways. During nutrient availability (growth factors and amino acids), mTOR kinase, a target of rapamycin, suppresses autophagy. Mutations in the PI3K/AKT/mTOR pathway, in which autophagy is negatively regulated, may lead to oncogenic transformations by increasing signalling in the pathway (Ref. Reference Alers58).

Beclin-1, which has a central role in autophagy, can function as a tumour suppressor (Ref. Reference Liang59). Beclin-1 is monoallelic deleted in many cancers, such as breast, ovarian, brain, and prostate cancers, and low protein levels have been found in the tumours of these cancers. Studies in mice have shown that heterozygous disruption of beclin-1 increases permeability to spontaneous tumour development (such as lymphomas, lung carcinomas, hepatocellular carcinomas, and mammary precancerous lesions), whereas Beclin-1 restoration in MCF-7 cells reduces proliferation and tumorigenesis (Refs Reference Liang59, Reference Mathew, Karantza-Wadsworth and White60, Reference Miracco61). Bif-1 plays a role in inducing autophagosome formation in autophagy and interacts with Beclin-1 via UVRAG. UVRAG proteins have been observed to be abnormal or deleted in various types of cancer, such as colorectal and gastric cancer. It is also a tumour suppressor candidate because it reduces proliferation and tumour size in cases of excessive UVRAG expression (Ref. Reference Liang62). It has been reported that the MAP1-LC3 gene, which encodes LC3-II located in the inner and outer membrane of the autophagosome and functions in autophagy substrate selection and autophagosome biogenesis, is frequently deleted in the liver, breast, prostate, and ovarian cancers (Ref. Reference Salvi63). Thus, it can be considered that autophagy is both a tumour suppressor and promoter (via protective autophagy) mechanism, tumours with autophagy deficiency grow faster and this situation increases exponentially with defects in apoptosis.

Modulation of autophagy in cancer therapy is a promising potential strategy. Autophagy is also a protective mechanism in cancer cells during anticancer treatment. The common treatment strategy in cancer treatment is chemotherapy, but the development of chemoresistance (such as the protective autophagy mechanism) limits the success rate. Some autophagy regulators, such as the best-known mTOR inhibitors rapamycin and its derivatives (temsirolimus and everolimus), chloroquine (CQ), and hydroxychloroquine (HCQ), are also used in cancer treatment.

Rapamycin, an mTOR inhibitor, was developed to inhibit proliferation signals in the PI3K/AKT/mTOR pathway, which is important for cell growth and proliferation in multiple tumour types (Ref. Reference Huang64). Moreover, rapamycin may sensitise cancer cells to radiation therapy and chemotherapeutic agents such as adriamycin, cisplatin and hormonal therapies (Ref. Reference Rao, Buckner and Sarkaria65). In addition rapamycin is used in rare lung diseases, coronary stents (Ref. Reference Zaytseva66). Temsirolimus was the first mTOR inhibitor class drug to be shown to improve survival in patients with advanced renal cell carcinoma (Refs Reference Motzer67, Reference Jurczak68). Increased PI3K/AKT expression and activation are common in renal cell cancer and this drug is used in the first-line treatment of metastatic renal cell cancer with poor prognosis (Ref. Reference Zaytseva66). It is also used in the treatment of relapsed or refractory mantle cell lymphoma in the European Union. Everolimus is used in combination with exemestane in the treatment of advanced neuroendocrine tumours and breast cancer (Refs Reference Yao69, Reference Zhao70). In preclinical studies in bladder cancer and pancreatic adenocarcinoma, it has been shown that CQ and HCQ can suppress cancer cell growth by inhibiting autophagy. Several studies have demonstrated that these reagents promote apoptosis through suppression of autophagy and enhance the therapeutic effects of chemotherapy by inhibiting autophagy-mediated resistance seen in therapy (Refs Reference Zhao70, Reference Liang, El-Zein and Dave71). Lys05, a water-soluble analogue of HCQ, showed higher anticancer effects than HCQ in low doses of melanoma and colon cancer xenograft models (Ref. Reference Barnard72). 5-Fluorouracil (5-FU), which is used in solid cancers such as colorectal, breast, and pancreatic cancer, has limited efficacy in the treatment because it induces protective autophagy and creates chemoresistance (Refs Reference Malet-Martino, Jolimaitre and Martino73, Reference Liang74). Spautin-1, SAR405 are newly developed autophagy-related anticancer drugs (Ref. Reference Kunimasa75).

Autophagy in cardiovascular diseases

Autophagic activity is implicated in the regulation of mammalian heart development in the early stage, maintenance of cardiac structure and function under normal conditions, in the onset and progression of cardiovascular diseases including ischaemia/reperfusion (I/R) and heart failure (Ref. Reference Gatica4). In addition, autophagy is important in delaying cardiac aging (Ref. Reference Shirakabe77).

Activation of autophagy during myocardial ischaemia ensures the maintenance of energy substrates and removal of damaged mitochondria that may initiate apoptosis by causing oxidative stress (Ref. Reference Zhang78). Autophagy activation during ischaemia relies on AMPK mediated inhibition of the mTORC1 pathway (Ref. Reference Matsui79). Inhibition of autophagy during chronic ischaemia can cause cardiomyocyte death by activating apoptosis. Therefore, induction of autophagy during myocardial ischaemia may be protective.

During reperfusion, tissues exposed to ischaemia are reloaded with nutrients and oxygen. Reperfusion injury is defined as damage to tissues or organs during the re-bleeding period following the ischaemic period. Studies have shown that autophagy is a key regulator with a dual role for I/R and is activated during I/R (Ref. Reference Hao80). AMPK activation is involved in autophagy resulting from ischaemia, whereas activation of Beclin 1 is involved in autophagy during reperfusion. Autophagy plays different roles during ischaemia and reperfusion; while protective during ischaemia, it could be harmful during reperfusion (Ref. Reference Matsui79). (Table 2).

Table 2. Studies on autophagy in cardiovascular diseases

The heart's first response to various stresses, such as hypertension, aortic stenosis, is hypertrophy, and if this situation continues, heart failure may develop (Ref. Reference Dämmrich and Pfeifer81). Furthermore, age-related progressive loss of autophagic activity leads to the development of hypertrophy (Ref. Reference Shirakabe77). It remains unclear whether the altered autophagy observed in cardiac hypertrophy and heart failure is beneficial or harmful. β-adrenergic stimulation, which induces cardiac hypertrophy and heart failure, inhibits autophagy and stimulates apoptosis (Ref. Reference Hao80). The deficiency of ATG5 causes cardiac hypertrophy by inhibition of autophagy with increased ubiquitination levels, while overexpression of ATG5 increases autophagy levels with Beclin-1 function and extends lifespan (Ref. Reference Nakai82). In conclusion, basal autophagy in the heart under normal conditions is a homoeostatic mechanism for maintaining cardiac structure and function.

The prevalence of autophagic activation in cardiac diseases has led to studies in this area for therapeutic targets. In the conjectural applications of cardiovascular diseases, 8-methylchroman-7-ol derivatives and glycosylated anti-tumour ether lipids inhibit autophagy; Rubicon, which binds the class III PI3K/Vps34–Beclin 1 complex, and AP23573, a phosphorus-rapamycin analogue, are used in the regulation of autophagy (Ref. Reference Nemchenko83).

Autophagy in neurological aging

Autophagy is important for the maintenance of neuronal homoeostasis by mediating the clearance of unfolded or misfolded protein aggregates.

Age-related decrease in autophagy may lead to accumulation of intracellular toxic wastes (Ref. Reference Di Domenico28). This condition is characterised by aggregated and abnormally deposited proteins in NDs. Apart from protein aggregation, mitochondrial dysfunction and decreased lysosomal activity accompany dysregulated autophagy in NDs. Aging and neuronal autophagy dysregulation increase the incidence of NDs such as AD, PD, HD (Table 3). In particular, misfolded protein accumulation and disruption of the autophagy pathway due to mTOR are seen in these diseases. With aging, there may be decreases in the ubiquitin-proteasome system and autophagy-lysosomal pathway, which are associated with the clearance of misfolded proteins in the brain (Ref. Reference Hansen, Rubinsztein and Walker84). Measures to eliminate the disorders in the autophagy mechanism may contribute to the prevention of age-related diseases. Gene polymorphisms involved in autophagy mechanisms contribute to aging-related neurodegeneration. Plaques formed as a result of tau protein accumulation in AD, mutant α-synuclein proteins (Lewy bodies) in PD, and mutant Huntingtin protein (mHtt) accumulation in the cytoplasm are seen in HD. Autophagy plays an active role in the clearance of these proteins. There is evidence that overexpression of ATG5, which is involved in autophagy induction, activates autophagy and increases survival. However, it was shown that there was a decrease in the expression of the ATG7 and Beclin-1 genes. In this case, it can be concluded that the decrease in autophagy activation decreases with aging (Ref. Reference Lipinski22).

Table 3. Studies on autophagy in neurodegenerative diseases

AD, Alzheimer's Disease; PD, Parkinson Disease; HD, Huntington's Disease.

Alzheimer's disease

AD is a ND characterised by the accumulation of extracellular β-amyloid (Aβ) plaques and intracellular neurofibrillary tangles containing hyperphosphorylated tau (Ref. Reference Tan5). Only 5% of AD is familial and results from mutations in presenilin 1, presenilin 2, and amyloid precursor protein (APP). Aβ and tau protein are substrates for autophagy. An abundance of autophagic structures containing these substrates is observed in AD. In addition, it has been shown that abnormal conformations of Aβ reduce lysosomal amplification, lead to synaptic defects and increase the course of the disease (Ref. Reference Holtzman, Morris and Goate85). Neurofibrillary tangles caused by mutant tau proteins can block CMA, resulting in decreased autophagy. Beclin-1, an autophagy-related protein, decreases with age in the brains of AD models, and increased levels of Beclin-1 ameliorate amyloid pathology.

Due to the morphological structure of nerve cells, the levels of clearance by autophagosomes may be different compared to other cells. The importance of autophagy in these cells is due to their post-mitotic nature. Autophagy is very important in cell survival as it can remove toxic aggregates in post-mitotic nerve cells. It has been found that autophagosome clearance is impaired in late-stage disease in neuron cells from patients with AD only. In some studies, it has been reported that the levels of some autophagy genes are increased in AD brains (Ref. Reference Lipinski22).

Parkinson disease

PD is a ND caused by selective loss of dopaminergic neurons and decreased dopamine content in the striatum (Ref. Reference Halliday, Lees and Stern86). This loss can be induced by mitochondrial toxins such as 1-methyl-4-phenyl-1,2,3,6 tetrahydropyridine (MPTP) and the complex I inhibitor rotenone. As in AD, mutations that cause PD can directly cause disruption of autophagy. Some of these mutations are α-synuclein (SNCA or PARK1), also known as Lewy bodies, Parkin (PRKN or PARK2), ubiquitin carboxy-terminal hydrolase L1 (UCH-L1), PTEN-induced putative kinase 1 (PINK1), protein deglycase DJ-1 (PARK7) and leucine-rich repeat kinase 2 (LRRK2) (Ref. Reference Senkevich and Gan-Or87). Furthermore, many PD-related gene mutations can cause loss of function in the autophagy-lysosome pathway (ALP). Because these genes play a role in mitophagy, mutations in these genes cause insufficient mitochondrial quality control (Ref. Reference Pickrell and Youle88). Two major mutations in α-synuclein (A53T and A30P) are associated with PD (Ref. Reference Recchia89). Mutant α-synuclein causes disruption of CMA activity, inhibition of autophagosome formation and disruption of lysosomal degradation (Ref. Reference Winslow90). Increased expression of mTOR, one of the important autophagy pathways, has been reported after α-synuclein accumulation in patients with PD. Apart from aging and genetic mutations, PD may also develop due to dopaminergic neuron-specific toxins such as 6-hydroxydopamine (6-OHDA), 1-methyl-4-phenyl-1,2,3,6 tetrahydropyridine (MPTP) and rotenone.

Huntington's disease

HD is an autosomal dominant ND caused by long CAG trinucleotide expansion (over 37 repeats) in the Htt gene (Ref. Reference McColgan and Tabrizi91). In patients with HD, an abnormally long polyglutamine-encoding CAG trinucleotide expansion produces perinuclear cytoplasmic aggregates and intranuclear inclusions. This results in mutant Htt, an autophagic substrate, ingestion of defective autophagic substrate into autophagosomes. In addition, this expansion of polyglutamine causes misfolded proteins or accumulation of protein aggregates. Deletion of polyglutamine in the Htt gene has been shown to improve disease symptoms and increase autophagosome formation (Ref. Reference Jeong92). In addition, the decrease in Beclin-1 expression with aging may lead to the accumulation of mutant Htt (Ref. Reference Wu93). It has also been reported that the mTOR pathway plays a role in HD pathology. In a recent study, they identified the Homeodomain Interacting Protein Kinase 3 (HIPK3) gene as a negative modulator of autophagy and a positive regulator of mHtt expression levels in HD cells. It is considered that modulation of mHtt by HIPK3 can be a therapeutic target for HD (Ref. Reference Fu, Sun and Lu94). Stimulation of autophagy usually occurs by inhibition of mTOR. However, since mTOR is involved in many cellular processes, its inhibition can lead to unexpected effects. Walter et al. showed that AMPK activation causes increased expression of LC3-II and p62, and induced autophagy in an mTOR-independent manner. This resulted in decreased aggregates containing mHtt and cell viability (Ref. Reference Walter95).

Autophagy in obesity and diabetes

Obesity, which is associated with excessive calorie intake, is a chronic disease that occurs as a result of many genetic and environmental factors, affecting the metabolism and physiology of organs (Ref. Reference Zhang, Sowers and Ren115). Since triglycerides are stored in adipose tissue and metabolised by the liver, adipose tissue is at the centre of obesity and metabolic diseases (Ref. Reference Oga and Eseyin116). Autophagy, on the other hand, is a physiological process that removes damaged organelles, misfolded proteins, and lipids in case of excessive calorie intake. Autophagy is a highly sensitive mechanism to excessive calorie intake. Suppression or increase of autophagy is seen in metabolic diseases such as obesity and diabetes (Ref. Reference Jaishy6).

Insulin resistance, which develops due to the increase in calorie intake, suppresses the activity of the mTOR pathway; this initiates the autophagy process in adipose tissues. However, it has been determined that calorie restriction increases autophagy activity and, accordingly, insulin sensitivity in obese individuals. In addition, an increase in the LC3 level and the number of autophagosomes in the fat cells of obese patients was observed. In another study, it was determined that the mRNA expression level of the autophagy gene ATG5 was higher in patients with higher BMI and larger visceral adipose tissue (Ref. Reference Kovsan117). Obesity was shown to stimulate inflammation by causing hypothalamic resistance to insulin and leptin hormones through an inhibitor of the Nuclear Factor kappa B (NF-kB) pathway and induces hypothalamic dysfunction (Ref. Reference Zhang118). Therefore, hypothalamic dysfunction can be considered to play a role in the pathophysiology of obesity and diabetes. In studies, hypothalamic inhibition of autophagy via siRNA-mediated ATG7 suppression was achieved in mice fed a high-fat diet, resulting in increased energy consumption. In other words, feeding with HFD in mice caused disruption of lipolysis and insulin resistance. It has also been observed that hypothalamic autophagy is impaired in HFD-induced obesity (Ref. Reference Kaushik119).

Diabetes is a metabolic disease identified as chronic hyperglycaemia. There are two main clinical types, insulin-dependent (Type 1) and insulin-independent (Type 2) (Ref. Reference Gonzalez120). Type 1 diabetes mellitus (T1DM) is a disease that develops by autoimmune destruction of pancreatic beta cells. It is known that insulin reduces tau phosphorylation and autophagy by inhibiting the PI3K/AKT signalling pathway-mediated Glycogen synthase kinase-3 (GSK-3). Phosphorylation of tau protein is increased in T1DM with insulin deficiency (Ref. Reference Santos121). On the other hand, insulin resistance, hyperglycaemia and relative insulin deficiency determine the development of Type 2 diabetes mellitus (T2DM). The increase of adipose tissue in T2DM causes insulin resistance by activating the mTOR pathway. mTORC1 is activated by high amounts of glucose, fatty acids and amino acids. Moreover, insulin is a hormone that inhibits autophagy (Ref. Reference Brännmark122). Insulin resistance causes autophagy activation in adipose tissues of obese patients. In the previous studies, it was demonstrated that insulin resistance was induced and autophagy was inhibited by decreasing the expression levels of autophagy-related LC3, ULK2, ATG12 proteins in livers of mice fed a high-fat diet (Ref. Reference Ueno and Komatsu123). Studies on autophagy in obesity and diabetes are summarised in Table 4.

Table 4. Studies on autophagy in obesity and diabetes

Natural active compounds in longevity intervention

Age is the greatest risk factor for all major age-related pathologies. In recent years, it has been reported that the molecular mechanisms underlying aging are associated with cancer, cardiovascular diseases and neurodegeneration. It can also affect the same pathway, autophagy, as these diseases. Therefore, autophagy is gaining importance in the discovery of therapeutic interventions that promote healthy aging and increase longevity (Ref. Reference Niso-Santano142).

Natural products were widely used to treat different medical conditions before the development of modern pharmaceuticals. Today they still serve as an important pool for the identification of new drug precursors against various diseases including cancer and chronic diseases, which become widespread with increasing age. The structural complexity and diversity of natural products provide a valuable resource for future drug discovery.

In the following section, we summarise the autophagic mechanisms of natural compounds in cancer, cardiac diseases, neurological aging, diabetes and obesity.

Autophagic mechanisms of natural compounds in cancer

Natural compounds have been identified as potent inducers or inhibitors of autophagy to exhibit antitumour mechanisms, opening up new therapeutic regimens against cancer. Some studies suggest that autophagy exerts a tumour-suppressing role, while others suggest a tumour-promoting role. This complexity of autophagy in cancer biology may arise from different contexts such as stress types, tumour staging and cancer types. In vitro studies on different groups of natural compounds modulating autophagy and molecular target mechanisms were summarised in Table 5 and Figure 3.

Figure 3. Autophagy-related pathways regulated by natural compounds in cancer (Figure created with Biorender.com).

Table 5. In vitro studies on natural compounds modulating autophagy in various cancers signalling pathways

Alkaloids are a group of secondary metabolites that are extensively found in nature and have strong pharmacological activities. They can target the autophagy process in different cancer types including breast, lung, cervical, colon, liver and pancreatic cancer. Among the alkaloids, berbamine, cepharanthine, dauricine, daurisolin, liensinine inhibited the autophagy via different molecular target mechanism while matrine induced autophagy by p53/AMPK pathway (Refs Reference Fu143, Reference Tang144, Reference Wu145, Reference Zhou146, Reference Xie, He and Yao147). Natural cyclopeptide RA-XII suppresses protective autophagy through AMPK/mTOR/P70S6 K pathways in HepG2 cells (Ref. Reference Song148). Oridanin, concanavalin and ferulic acid induced autophagy and displayed an anticancer effect against cervical cancer (Refs Reference Cui149, Reference Roy150, Reference Gao151). Kaempferol and quercetin are natural flavonoids that are found in many fruits, vegetables, and medicinal plants. They mediated autophagy in gastric and pancreatic cancer cell lines via inhibition of HDAC/G9a and mTOR activity (Refs Reference Kim152, Reference Lan153). An indole derivative of 3,3′-diindolylmethane also plays a regulatory role in gastric cancer and inhibits miR-30e leading to ATG5 activation (Ref. Reference Ye154). Honokiol is a lignan isolated from the genus Magnolia L. (Magnoliaceae) that inhibits melanoma stem cells by targeting notch signalling and induced in autophagic cell death (Ref. Reference Kaushik155). Paeoniflorigenone regulated autophagy to induce anticancer bioactivities in human head and neck squamous cell carcinomas. This compound inactivated PI3K/AKT/mTOR/p70S6K in YD-10B cells (Ref. Reference Park156). β-lapachone and plumbagin, the active naphthoquinone, showed potent anticancer effects through autophagy induction (Refs Reference Park, Choi and Kwon157, Reference Li158). p-Coumaric acid, an ubiquitous plant phenolic acid, had cytotoxicity on neuroblastoma via generation of ROS that enhanced autophagy-induced mitochondria dysfunction (Ref. Reference Shailasree159). Elaiophylin, a natural autophagy inhibitor, exerted antitumour activity in multiple myeloma and ovarian cancer (Refs Reference Zhao70, Reference Wang160). Polyphenols are a large group of natural compounds that played an important role in modulating autophagy. They had significant antioxidant and anti-inflammatory properties as well as autophagic regulation in cancer cells. Curcumin is a natural polyphenol derived from rhizomes of Curcuma longa L. (Zingiberaceae) commonly known as turmeric. Numerous studies indicated that curcumin was able to modulate autophagy against various cancer including prostate, colon, oral, pancreatic cancers and brain tumours (Refs Reference Aoki161, Reference Teiten162, Reference Lee163, Reference Kim164, Reference Zhu and Bu165, Reference Liu166). Withaferin A had a steroidal lactone structure and inhibited lysosomal activity to block autophagic flux in breast cancer cells (Ref. Reference Muniraj167). Periplocin is a natural active steroid isolated from Periploca forrestii Schltr. (Apocynaceae). This compound promoted autophagy in pancreatic cancer cells via regulating the AMPK/mTOR pathway (Ref. Reference Zhang168). Resveratrol (3,5,4-trihydroxystilbene) is a stilbenoid found mainly in red grape, cranberry, mulberry, and peanut. It has gained more attention over the past two decades because of its ability to prevent and treat various cancers (Ref. Reference Tian169). A link between resveratrol and autophagy regulation by different moleculer mechanism was reported in breast, colon and ovarian cancers (Refs Reference Fu170, Reference Miki171, Reference Ferraresi172). Corilagin and punicalagin, members of the tannin group, induced autophagy in gastric cancer and glioblastoma cell lines, respectively (Refs Reference Xu173, Reference Wang174). Lucidone, a terpeneoid, inhibited autophagy via HMGB1/RAGE/PI3K/Akt signalling pathway in pancreatic cancer cells (Ref. Reference Chen175). Celastrol induced autophagy by targeting AR/miR-101 and served as epigenetic regulator in prostate cancer cell lines (Ref. Reference Guo176). Two natural triterpenoids, toosendanin and isotoosendanin, suppressed triple-negative breast cancer growth through inducing autophagy (Ref. Reference Zhang177).

Autophagic mechanisms of natural compounds in cardiac diseases

Despite recent advances in therapeutic regimens, cardiac disease is still a leading cause of morbidity and mortality and remains one of the greatest threats to public health. Dysregulation of autophagy in cardiomyocytes is associated with myocardial infarction, cardiac hypertrophy, heart failure and diabetic cardiomyopathy. In this context, autophagy appears to be an important therapeutic target and delays cardiac aging (Ref. Reference Wu178). Different groups of natural compounds including berberine, tanshinone IIA, oridanin, hesperidin, icariin, luteolin, nobiletin, puerarin, melatonin, hinokitiol, thymoquinone, gallic acid, spermidine, curcumin, allicin, resveratrol, ginsenoside Rg3, cucurbitacin B were reported to display cardioprotective effect in experimental animal models and in vitro studies via autophagy modulation (Refs Reference Huang179, Reference Zeng180, Reference Wang181, Reference Xu182, Reference Li183, Reference Hu184, Reference Hu185, Reference Wu186, Reference Liu187, Reference Chen188, Reference Xie189, Reference Xiao190, Reference Liu191, Reference Yan192, Reference Eisenberg193, Reference Yan194, Reference Liu195, Reference Ba196, Reference Gu197, Reference Xu198, Reference Sun199, Reference Xiao200). Autophagy regulating natural compounds in cardiac diseases and molecular target mechanisms were summarised in Table 6 and Figure 4.

Figure 4. Autophagy-related pathways regulated by natural compounds in cardiac disease (Figure created with Biorender.com).

Table 6. Autophagy regulating natural compounds in cardiac diseases

Autophagic mechanisms of natural compounds in neurological aging

Many studies indicated that natural compounds had therapeutic benefits in NDs via different mechanisms by targeting autophagy. Autophagy regulating natural compounds in neurological aging and molecular target mechanisms were given in Table 7 and Figure 5. Berberine, conophylline, curcumin, ginsenoside-Rg2, and celastrol improved cognitive impairment via autophagy induction in mice models of AD (Refs Reference Huang201, Reference Umezawa202, Reference Wang203, Reference Fan204, Reference Yang205). Caffeine is a well-known natural compound commonly used to increase alertness and energy. It prevented human prion protein-mediated neurotoxicity via autophagy induction (Ref. Reference Moon206). Dendrobine, an alkaloid, induced autophagy flux in hippocampus primary neurons of rats (Ref. Reference Li207). Abnormalities in neuron axonal transport-related proteins inhibit autophagosome maturation in AD. Curcumin increased autophagic flux by inducing interactions among autophagic axonal transport-related proteins and promoting lysosome-autophagosome fusion in N2a/APP695swe cells (Ref. Reference Liang208). Curcumin also showed a protective effect on amyloid-β-42 induced cytotoxicity in HT-22 cells (Ref. Reference Zhang209). β-asarone and cubeben inhibited amyloid-β, and PI3K-AKT, respectively via promoting autophagy in AD model with neuronal cells (Refs Reference Wang210, Reference Li, Song and Dong211). Wogonin, natural active flavonoid, increased β-amyloid clearance and inhibited mTOR in SH-SHY5Y cells (Ref. Reference Zhu and Wang212). Baicalein inhibited caspase-3 activity and exhibited beneficial effect in PD model with SH-SY5Y (Ref. Reference Kuang, Cao and Lu213). Calycosin, an phytoestrogen, showed protective effect against paraquat (PQ)-induced neurodegeneration by reducing pS6K and p4EBP1 levels (Ref. Reference Chaouhan214). Quercetin, a flavonoid known for its neuroprotective effects, acted as an autophagy enhancer and upregulated Beclin-1 in PD rat model (Ref. Reference El-Horany215). n-Butylidenephthalide induced autophagic down-regulation in motor neurons and prolonged the survival of amyotrophic lateral sclerosis (ALS) mice (Ref. Reference Hsueh216). Oleuropein is the active constituent of olive leaves and fruits and known as antioxidant, neuroprotective and autophagy-regulating properties. These polyphenolic compounds showed therapeutic benefits against PD in neuronal PC12 cells and TgCRND8 mouse model (Refs Reference Achour217, Reference Rigacci218). Resveratrol was reported to have neuroprotective potential in HD. It protected mutant SH-SY5Y cells from dopamine toxicity via rescuing ATG4-mediated mediated autophagosome formation (Ref. Reference Vidoni219). Also, resveratrol showed beneficial effects by inducing autophagy in both in vitro and in vivo AD and PD models (Refs Reference Lin220, Reference Deng and Mi221, Reference Guo222). D-limonene is a fragrance agent and belongs to the terpene group. D-limonene increased LC3-II and reduced p62 levels by inducing autophagy in SH-SY5Y cells (Ref. Reference Russo223). Cucurbitacin E is a tetracyclic triterpenoid isolated from Cucurbitaceae plants, decreased autophagic flux and neuronal death in a postmitotic cellular model of PD (Ref. Reference Arel-Dubeau224). Geraniol also diminished autophagy flux in an in vitro model of PD (Ref. Reference Rekha and Sivakamasundari225). Cannabidiol, a natural compound from Cannabis sativa L. (Cannabaceae) extended lifespan and improved neuronal health in C. elegans by induction of autophagy (Ref. Reference Wang226).

Figure 5. Autophagy-related pathways regulated by natural compounds in neurological aging (Figure created with Biorender.com).

Table 7. Autophagy regulating natural compounds in neurological aging

Autophagic mechanisms of natural compounds in obesity and diabetes

Autophagy has a modulating role in the process of adipocyte conversion. It is also an important signalling pathway for T2DM. Autophagy regulating natural compounds in obesity and diabetes and molecular target mechanisms were given in Table 8 and Figure 6. Berberine, an alkaloid, inhibited basal autophagy in adipocytes in mice fed a high-fat diet by decreasing Beclin-1 (Ref. Reference Deng227). Resveratrol increased AMPK and improved health and survival of high-calorie diet induced mice (Ref. Reference Baur228). Resveratrol activated SIRT1/FoXO1/Rab7 and ameliorated myocardial oxidative stress injury in diabetic mice (Ref. Reference Wang229). It alleviated I/R injury of diabetic myocardium by promoting autophagy (Ref. Reference Qu230). Apart from resveratrol, epigallocatechin-3-gallate, trehalose, dihydromyricetin, puerarin, melatonin, ferulic acid, arjunolic acid, and curcumin were reported to effective in experimental animal models and in vitro studies of diabetes via autophagy modulation (Refs Reference Yan231, Reference Xu232, Reference Shi233, Reference Xu234, Reference Zhang235, Reference Ma236, Reference Zhang237, Reference Yao238, Reference Zhang239).

Figure 6. Autophagy-related pathways regulated by natural compounds in obesity and diabetes (Figure created with Biorender.com).

Table 8. Autophagy regulating natural compounds in obesity and diabetes

Conclusion and future perspectives

Aging, an irreversible biological process, is an independent risk factor for cancer, neurodegeneration, cardiovascular diseases, obesity and diabetes. Under both healthy and pathological circumstances, autophagy is a crucial mechanism. Studies have revealed that the dysregulation of autophagy plays a role in the pathogenesis of illnesses associated with aging and have proposed potential treatments involving the control of the autophagy system. It is thought that a number of bioactive substances generated from medicinal plants have the ability to regulate autophagy by concentrating on autophagic pathways. The targeted control of autophagy is thought to offer a method of treating age-related disorders, hence regulation of the autophagic process is increasingly seen as an intriguing drug development technique. The regulating function of natural substances on autophagy to delay or treat age-related illnesses in vitro and in animal models was the main focus of this review. However, there is broad agreement about the effectiveness of natural pleiotropic substances capable of enhancing or reestablishing deficient autophagy in aggregate-prone proteins. According to literature data, different groups of phytochemicals have attracted attention as promising autophagy modulators. Curcumin and resveratrol stand out in terms of effectiveness and popularity and these two compounds can be identified as potent autophagy regulating compounds in cancer, cardiovascular and NDs, and metabolic dysfunction. Keep in mind that autophagy can be a two-way process. To successfully combat age-related illnesses, significant improvements in our understanding of their modes of action, pharmacokinetics, and nonspecific effects are required.

References

Aman, Y et al. (2021) Autophagy in healthy aging and disease. Nature Aging 1, 634650.CrossRefGoogle ScholarPubMed
Menzies, FM et al. (2017) Autophagy and neurodegeneration: pathogenic mechanisms and therapeutic opportunities. Neuron 93, 10151034.CrossRefGoogle ScholarPubMed
Ruckenstuhl, C et al. (2014) Lifespan extension by methionine restriction requires autophagy-dependent vacuolar acidification. PLoS Genetics 10, e1004347.CrossRefGoogle ScholarPubMed
Gatica, D et al. (2015) Molecular mechanisms of autophagy in the cardiovascular system. Circulation Research 116, 456467.CrossRefGoogle ScholarPubMed
Tan, CC et al. (2014) Autophagy in aging and neurodegenerative diseases: implications for pathogenesis and therapy. Neurobiology of Aging 35, 941957.CrossRefGoogle ScholarPubMed
Jaishy, B et al. (2015) Lipid-induced NOX2 activation inhibits autophagic flux by impairing lysosomal enzyme activity. Journal of Lipid Research 56, 546561.CrossRefGoogle ScholarPubMed
Rahman, MA et al. (2020) Molecular insights into the multifunctional role of natural compounds: autophagy modulation and cancer prevention. Biomedicines 8, 517.CrossRefGoogle ScholarPubMed
Majeski, AE et al. (2004) Mechanisms of chaperone-mediated autophagy. International Journal of Biochemistry & Cell Biology 36, 24352444.CrossRefGoogle ScholarPubMed
Kroemer, G et al. (2010) Autophagy and integrated stress response. Molecular Cell 40, 280293.CrossRefGoogle ScholarPubMed
Tekirdag, K and Cuervo, AM (2018) Chaperone-mediated autophagy and endosomal microautophagy: jointed by a chaperone. Journal of Biological Chemistry 293, 54145424.CrossRefGoogle Scholar
Vitale, E et al. (2022) Apelin-13 increases functional connexin-43 through autophagy inhibition via AKT/mTOR pathway in the non-myocytic cell population of the heart. International Journal of Molecular Sciences 23, 13073.CrossRefGoogle ScholarPubMed
Huang, J et al. (2007) Autophagy and human disease. Cell Cycle 6, 18371849.CrossRefGoogle ScholarPubMed
Klionsky, DJ et al. (2003) A unified nomenclature for yeast autophagy-related genes. Developmental 5, 539545.Google ScholarPubMed
Kovacs, AL et al. (2010) Role of autophagy in Caenorhabditis elegans. FEBS Letters 584, 13351341.CrossRefGoogle ScholarPubMed
Karampa, AD et al. (2022) The role of macroautophagy and chaperone-mediated autophagy in the pathogenesis and management of hepatocellular carcinoma. Cancers 14, 760.CrossRefGoogle ScholarPubMed
Hu, Y and Reggiori, F (2022) Molecular regulation of autophagosome formation. Biochemical Society transactions 50, 5569.CrossRefGoogle ScholarPubMed
Sanchez-Mirasierra, I et al. (2022) Targeting macroautophagy as a therapeutic opportunity to treat Parkinson's disease. Frontiers in Cell and Developmental Biology 10, 921314.CrossRefGoogle ScholarPubMed
Scrivo, A et al. (2018) Selective autophagy as a potential therapeutic target for neurodegenerative disorders. The Lancet. Neurology 17, 802815.CrossRefGoogle ScholarPubMed
McCarty, MF (2022) Nutraceutical and dietary strategies for up-regulating macroautophagy. International journal of molecular sciences 23, 2054.CrossRefGoogle ScholarPubMed
Massey, AC, Zhang, C and Cuervo, AM (2006) Chaperone-mediated autophagy in aging and disease. Current Topics in Developmental Biology 73, 205235.CrossRefGoogle ScholarPubMed
Arndt, V (2010) Chaperone-assisted selective autophagy is essential for muscle maintenance. Current Biology 20, 143148.CrossRefGoogle ScholarPubMed
Lipinski, MM et al. (2010) Genome-wide analysis reveals mechanisms modulating autophagy in normal brain aging and in Alzheimer's disease. Proceedings of the National Academy of Sciences of the United States of America 107, 14164–9.CrossRefGoogle ScholarPubMed
Fernández, ÁF et al. (2018) Disruption of the beclin 1-BCL2 autophagy regulatory complex promotes longevity in mice. Nature 558, 136140.CrossRefGoogle ScholarPubMed
Moreno-Blas, D et al. (2019) Cortical neurons develop a senescence-like phenotype promoted by dysfunctional autophagy. Aging (Albany NY) 11, 61756198.CrossRefGoogle ScholarPubMed
Nie, D et al. (2021) Rapamycin treatment of tendon stem/progenitor cells reduces cellular senescence by upregulating autophagy. Stem Cells International 2021, 6638249.CrossRefGoogle ScholarPubMed
Cannizzo, ES et al. (2012) Age-related oxidative stress compromises endosomal proteostasis. Cell Reports 2, 136149.CrossRefGoogle ScholarPubMed
Valdor, R et al. (2014) Chaperone-mediated autophagy regulates T cell responses through targeted degradation of negative regulators of T cell activation. Nature Immunology 15, 10461054.CrossRefGoogle ScholarPubMed
Di Domenico, F et al. (2014) Oxidative stress and proteostasis network: Culprit and casualty of Alzheimer's-like neurodegeneration. Advances in Geriatrics 2014, 14.CrossRefGoogle Scholar
Um, JH and Yun, J (2017) Emerging role of mitophagy in human diseases and physiology. BMB Reports 50, 299307.CrossRefGoogle ScholarPubMed
Boya, P, Codogno, P and Rodriguez-Muela, N (2018) Autophagy in stem cells: repair, remodelling and metabolic reprogramming. Development (Cambridge, England) 145, 114.CrossRefGoogle ScholarPubMed
Rajendran, P et al. (2019) Autophagy and senescence: a new insight in selected human diseases. Journal of Cellular Physiology 234, 2148521492.CrossRefGoogle ScholarPubMed
Hewitt, G and Korolchuk, VI (2017) Repair, reuse, recycle: the expanding role of autophagy in genome maintenance. Trends in Cell Biology 27, 340351.CrossRefGoogle ScholarPubMed
Lopez-Otín, C et al. (2013) The hallmarks of aging. Cell 153, 11941217.CrossRefGoogle ScholarPubMed
Cheon, SY et al. (2019) Autophagy, cellular aging and age-related human diseases. Experimental Neurobiology 28, 643657.CrossRefGoogle ScholarPubMed
Fairlie, WD, Tran, S and Lee, EF (2020) Crosstalk between apoptosis and autophagy signaling pathways. International Review of Cell and Molecular Biology 352, 115158.CrossRefGoogle ScholarPubMed
Ge, Y et al. (2022) Role of AMPK mediated pathways in autophagy and aging. Biochimie 195, 100113.CrossRefGoogle ScholarPubMed
Bhaskar, PT and Hay, N (2007) The two TORCs and Akt. Developmental Cell 12, 487502.CrossRefGoogle ScholarPubMed
Papadopoli, D et al. (2019) mTOR as a central regulator of lifespan and aging. F1000Research 8, F1000 Faculty Rev-998, 121.CrossRefGoogle ScholarPubMed
Wang, X et al. (2020) AMPK and Akt/mTOR signalling pathways participate in glucose-mediated regulation of hepatitis B virus replication and cellular autophagy. Cellular Microbiology 22, e13131.CrossRefGoogle ScholarPubMed
Zada, S et al. (2021) Cross talk between autophagy and oncogenic signaling pathways and implications for cancer therapy. Biochimica et Biophysica Acta. Reviews on Cancer 1876, 188565.CrossRefGoogle ScholarPubMed
Chen, Z et al. (2019) Carvedilol exerts myocardial protection via regulation of AMPK-mTOR-dependent autophagy? Biomedicine & Pharmacotherapy = Biomedecine & Pharmacotherapie 118, 109283.CrossRefGoogle ScholarPubMed
Pan, X et al. (2020) Accumulation of prelamin A induces premature aging through mTOR overactivation. FASEB Journal: Official Publication of the Federation of American Societies for Experimental Biology 34, 79057914.CrossRefGoogle ScholarPubMed
Zachari, M and Ganley, IG (2017) The mammalian ULK1 complex and autophagy initiation. Essays in Biochemistry 61, 585596.Google ScholarPubMed
Turco, E, Fracchiolla, D and Martens, S (2020) Recruitment and activation of the ULK1/Atg1 kinase complex in selective autophagy. Journal of Molecular Biology 432, 123134.CrossRefGoogle ScholarPubMed
Kitada, M, Ogura, Y and Koya, D (2016) Role of Sirt1 as a Regulator of Autophagy, Autophagy: Cancer, Other Pathologies, Inflammation, Immunity, Infection, and Aging. Amsterdam, Netherlands: Academic Press, pp. 89100.Google Scholar
Lee, IH et al. (2008) A role for the NAD-dependent deacetylase Sirt1 in the regulation of autophagy. Proceedings of the National Academy of Sciences of the United States of America 105, 33743379.CrossRefGoogle ScholarPubMed
Colman, RJ et al. (2009) Caloric restriction delays disease onset and mortality in Rhesus monkeys. Science (New York, N.Y.) 325, 201204.CrossRefGoogle ScholarPubMed
Hansen, M et al. (2008) A role for autophagy in the extension of lifespan by dietary restriction in C. elegans. PLoS Genetics 4, e24.CrossRefGoogle ScholarPubMed
Liu, D et al. (2019) PGC1α Activation by pterostilbene ameliorates acute doxorubicin cardiotoxicity by reducing oxidative stress via enhancing AMPK and SIRT1 cascades. Aging 11, 1006110073.CrossRefGoogle ScholarPubMed
Fahy, GM et al. (2019) Reversal of epigenetic aging and immunosenescent trends in humans. Aging cell 18, e13028.CrossRefGoogle ScholarPubMed
Ao, X, Zou, L and Wu, Y (2014) Regulation of autophagy by the Rab GTPase network. Cell Death and Differentiation 21, 348358.CrossRefGoogle ScholarPubMed
Salminen, A and Kaarniranta, K (2009) Regulation of the aging process by autophagy. Trends in Molecular Medicine 15, 217224.CrossRefGoogle ScholarPubMed
Mak, T (2003) The E. Donnall Thomas lecture-apoptosis: “Tis death that makes life live”. Biology of Blood and Marrow Transplantation 9, 483488.CrossRefGoogle Scholar
Anding, AL and Baehrecke, EH (2015) Autophagy in cell life and cell death. Current Topics in Developmental Biology 114, 6791.CrossRefGoogle ScholarPubMed
Yu, L, Chen, Y and Tooze, SA (2018) Autophagy pathway: cellular and molecular mechanisms. Autophagy 14, 207215.CrossRefGoogle ScholarPubMed
Ding, WX, Chen, X and Yin, XM (2012) Tumor cells can evade dependence on autophagy through adaptation. Biochemical and Biophysical Research Communications 425, 684688.CrossRefGoogle ScholarPubMed
Maycotte, P and Thorburn, A (2011) Autophagy and cancer therapy. Cancer Biology & Therapy 11, 127137.CrossRefGoogle ScholarPubMed
Alers, S et al. (2012) Role of AMPK-mTOR-Ulk1/2 in the regulation of autophagy: cross talk, shortcuts, and feedbacks. Molecular and Cellular Biology 32, 211.CrossRefGoogle ScholarPubMed
Liang, XH et al. (1999) Induction of autophagy and inhibition of tumorigenesis by beclin 1. Nature 402, 672676.CrossRefGoogle ScholarPubMed
Mathew, R, Karantza-Wadsworth, V and White, E (2007) Role of autophagy in cancer. Nature Reviews Cancer 7, 961967.CrossRefGoogle ScholarPubMed
Miracco, C et al. (2007) Protein and mRNA expression of autophagy gene Beclin 1 in human brain tumours. International Journal of Oncology 30, 429436.Google ScholarPubMed
Liang, C et al. (2006) Autophagic and tumour suppressor activity of a novel Beclin1-binding protein UVRAG. Nature Cell Biology 8, 688699.CrossRefGoogle ScholarPubMed
Salvi, A et al. (2022) PHY34 inhibits autophagy through V-ATPase V0A2 subunit inhibition and CAS/CSE1L nuclear cargo trafficking in high grade serous ovarian cancer. Cell Death and Disease 13, 45.CrossRefGoogle ScholarPubMed
Huang, W et al. (2022) UBE2T is upregulated, predicts poor prognosis, and promotes cell proliferation and invasion by promoting epithelial-mesenchymal transition via inhibiting autophagy in an AKT/mTOR dependent manner in ovarian cancer. Cell Cycle 21, 780791.CrossRefGoogle Scholar
Rao, RD, Buckner, JC and Sarkaria, JN (2004) Mammalian target of rapamycin (mTOR) inhibitors as anticancer agents. Current Cancer Drug Targets 4, 621635.CrossRefGoogle Scholar
Zaytseva, YY et al. (2012) mTOR inhibitors in cancer therapy. Cancer Letters 319, 17.CrossRefGoogle ScholarPubMed
Motzer, RJ et al. (2008) RECORD-1 Study group. Efficacy of everolimus in advanced renal cell carcinoma: a double-blind, randomised, placebo-controlled phase III trial. Lancet 372, 449456.CrossRefGoogle Scholar
Jurczak, W et al. (2018) Comparison of two doses of intravenous temsirolimus in patients with relapsed/refractory mantle cell lymphoma. Leukemia & Lymphoma 59, 670678.CrossRefGoogle ScholarPubMed
Yao, JC et al. (2008) Efficacy of RAD001 (everolimus) and octreotide LAR in advanced low- to intermediate-grade neuroendocrine tumors: results of a phase II study. Journal of Clinical Oncology 10, 43114318.CrossRefGoogle Scholar
Zhao, X et al. (2015) Elaiophylin, a novel autophagy inhibitor, exerts antitumor activity as a single agent in ovarian cancer cells. Autophagy 11, 18491863.CrossRefGoogle ScholarPubMed
Liang, DH, El-Zein, R and Dave, B (2015) Autophagy inhibition to increase radiosensitization in breast cancer. Journal of Nuclear Medicine & Radiation Therapy 6, 254.CrossRefGoogle ScholarPubMed
Barnard, RA et al. (2014) Phase I clinical trial and pharmacodynamic evaluation of combination hydroxychloroquine and doxorubicin treatment in pet dogs treated for spontaneously occurring lymphoma. Autophagy 10, 14151425.CrossRefGoogle ScholarPubMed
Malet-Martino, M, Jolimaitre, P and Martino, R (2022) The prodrugs of 5-fluorouracil. Current Medicinal Chemistry – Anti-Cancer Agents 2, 267310.CrossRefGoogle Scholar
Liang, X et al. (2014) Suppression of autophagy by chloroquine sensitizes 5-fluorouracil-mediated cell death in gallbladder carcinoma cells. Cell & Bioscience 4, 10.CrossRefGoogle ScholarPubMed
Kunimasa, K et al. (2022) Spautin-1 inhibits mitochondrial complex I and leads to suppression of the unfolded protein response and cell survival during glucose starvation. Scientific Reports 12, 11533.CrossRefGoogle ScholarPubMed
Zhao, XG et al. (2015) Chloroquine-enhanced efficacy of cisplatin in the treatment of hypopharyngeal carcinoma in xenograft mice. PLoS ONE 10, e0126147.CrossRefGoogle ScholarPubMed
Shirakabe, A et al. (2016) Aging and autophagy in the heart. Circulation Research 118, 15631576.CrossRefGoogle ScholarPubMed
Zhang, H et al. (2008) Mitochondrial autophagy is an HIF-1-dependent adaptive metabolic response to hypoxia. Journal of Biological Chemistry 283, 1089210903.CrossRefGoogle ScholarPubMed
Matsui, Y et al. (2007) Distinct roles of autophagy in the heart during ischemia and reperfusion: roles of AMP-activated protein kinase and Beclin 1 in mediating autophagy. Circulation Research 30, 914922.CrossRefGoogle Scholar
Hao, M et al. (2017) Myocardial ischemic postconditioning promotes autophagy against ischemia reperfusion injury via the activation of the nNOS/AMPK/mTOR pathway. International Journal of Molecular Sciences 18, 614.CrossRefGoogle ScholarPubMed
Dämmrich, J and Pfeifer, U (1983) Cardiac hypertrophy in rats after supravalvular aortic constriction. II. Inhibition of cellular autophagy in hypertrophying cardiomyocytes. Virchows Archiv. B, Cell Pathology Including Molecular Pathology 43, 287307.CrossRefGoogle ScholarPubMed
Nakai, A et al. (2007) The of autophagy in cardiomyocytes in the basal state and in response to hemodynamic stress. Nature Medicine 3, 619624.CrossRefGoogle Scholar
Nemchenko, A et al. (2011) Autophagy as a therapeutic target in cardiovascular disease. Journal of Molecular and Cellular Cardiology 51, 584593.CrossRefGoogle ScholarPubMed
Hansen, M, Rubinsztein, DC and Walker, DW (2018) Autophagy as a promoter of longevity: insights from model organisms. Nature Reviews. Molecular Cell Biology 19, 579593.CrossRefGoogle ScholarPubMed
Holtzman, DM, Morris, JC and Goate, AM (2011) Alzheimer's disease: the challenge of the second century. Science Translational Medicine 3, 135.CrossRefGoogle ScholarPubMed
Halliday, G, Lees, A and Stern, M (2011) Milestones in Parkinson's disease-clinical and pathologic features. Movement Disorders: Official Journal of the Movement Disorder Society 26, 10151021.CrossRefGoogle ScholarPubMed
Senkevich, K and Gan-Or, Z (2020) Autophagy lysosomal pathway dysfunction in Parkinson's disease; evidence from human genetics. Parkinsonism & Related Disorders 73, 6071.CrossRefGoogle ScholarPubMed
Pickrell, AM and Youle, RJ (2015) The roles of PINK1, Parkin and mitochondrial fidelity in Parkinson's disease. Neuron 85, 257273.CrossRefGoogle ScholarPubMed
Recchia, A et al. (2004) α-Synuclein and Parkinson's disease. The FASEB Journal 18, 617626.CrossRefGoogle ScholarPubMed
Winslow, AR et al. (2010) α-Synuclein impairs macroautophagy: implications for Parkinson's disease. The Journal of Cell Biology 190, 10231037.CrossRefGoogle ScholarPubMed
McColgan, P and Tabrizi, SJ (2018) Huntington's disease: a clinical review. European Journal of Neurology 25, 2434.CrossRefGoogle ScholarPubMed
Jeong, H et al. (2009) Acetylation targets mutant huntingtin to autophagosomes for degradation. Cell 137, 72.CrossRefGoogle ScholarPubMed
Wu, JC et al. (2012) The regulation of N-terminal Huntingtin (Htt552) accumulation by Beclin1. Acta Pharmacologica Sinica 33, 751.CrossRefGoogle ScholarPubMed
Fu, Y, Sun, X and Lu, B (2018) HIPK3 modulates autophagy and HTT protein levels in neuronal and mouse models of Huntington disease. Autophagy 14, 169170.CrossRefGoogle ScholarPubMed
Walter, C et al. (2016) Activation of AMPK-induced autophagy ameliorates Huntington disease pathology in vitro. Neuropharmacology 108, 2438.CrossRefGoogle ScholarPubMed
Pickford, F et al. (2008) The autophagy-related protein beclin 1 shows reduced expression in early Alzheimer disease and regulates amyloid beta accumulation in mice. The Journal of Clinical Investigation 118, 21902199.Google ScholarPubMed
Khandelwal, PJ et al. (2011) Parkin mediates beclin-dependent autophagic clearance of defective mitochondria and ubiquitinated Abeta in AD models. Human Molecular Genetics 20, 20912102.CrossRefGoogle ScholarPubMed
Rocchi, A et al. (2017) A Becn1 mutation mediates hyperactive autophagic sequestration of amyloid oligomers and improved cognition in Alzheimer's disease. PLoS Genetics 13, e1006962.CrossRefGoogle ScholarPubMed
Ma, Q et al. (2011) Age-related autophagy alterations in the brain of senescence accelerated mouse prone 8 (SAMP8) mice. Experimental Gerontology 46, 533541.CrossRefGoogle ScholarPubMed
Caccamo, A et al. (2013) mTOR regulates tau phosphorylation and degradation: implications for Alzheimer's disease and other tauopathies. Aging Cell 12, 370380.CrossRefGoogle ScholarPubMed
Caccamo, A et al. (2011) Naturally secreted amyloid-beta increases mammalian target of rapamycin (mTOR) activity via a PRAS40-mediated mechanism. The Journal of Biological Chemistry 286, 89248932.CrossRefGoogle Scholar
Albanese, F et al. (2021) Constitutive silencing of LRRK2 kinase activity leads to early glucocerebrosidase deregulation and late impairment of autophagy in vivo. Neurobiology of Disease 159, 105487.CrossRefGoogle ScholarPubMed
Zuo, L et al. (2021) 7,8-Dihydroxyflavone Ameliorates motor deficits via regulating autophagy in MPTP-induced mouse model of Parkinson's disease. Cell Death Discovery 7, 254.CrossRefGoogle ScholarPubMed
Kang, SY et al. (2017) Autophagic modulation by rosuvastatin prevents rotenone-induced neurotoxicity in an in vitro model of Parkinson's disease. Neuroscience Letters 642, 2026.CrossRefGoogle Scholar
Mammadova, N et al. (2019) Accelerated accumulation of retinal α-synuclein (pSer129) and tau, neuroinflammation, and autophagic dysregulation in a seeded mouse model of Parkinson's disease. Neurobiology of Disease 121, 116.CrossRefGoogle Scholar
Zhang, Y et al. (2019) Caffeic acid reduces A53T α-synuclein by activating JNK/Bcl-2-mediated autophagy in vitro and improves behaviour and protects dopaminergic neurons in a mouse model of Parkinson's disease. Pharmacological Research 150, 104538.CrossRefGoogle Scholar
Corrochano, S et al. (2012) α-Synuclein levels affect autophagosome numbers in vivo and modulate Huntington disease pathology. Autophagy 8, 431432.CrossRefGoogle ScholarPubMed
Tomás-Zapico, C et al. (2012) α-Synuclein accumulates in huntingtin inclusions but forms independent filaments and its deficiency attenuates early phenotype in a mouse model of Huntington's disease. Human Molecular Genetics 21, 495510.CrossRefGoogle Scholar
Ehrnhoefer, DE et al. (2018) Preventing mutant huntingtin proteolysis and intermittent fasting promote autophagy in models of Huntington disease. Acta Neuropathologica Communications 6, 16.CrossRefGoogle ScholarPubMed
Fox, LM et al. (2020) Huntington's disease pathogenesis is modified in vivo by Alfy/Wdfy3 and selective macroautophagy. Neuron 105, 813821, e6.CrossRefGoogle ScholarPubMed
Fu, H, Hardy, J and Duff, KE (2018) Selective vulnerability in neurodegenerative diseases. Nature Neuroscience 21, 13501358.CrossRefGoogle ScholarPubMed
Hegde, RN et al. (2020) TBK1 phosphorylates mutant Huntingtin and suppresses its aggregation and toxicity in Huntington's disease models. The EMBO journal 39, e104671.CrossRefGoogle ScholarPubMed
Pinho, BR et al. (2020) The interplay between redox signaling and proteostasis in neurodegeneration: in vivo effects of a mitochondria-targeted antioxidant in Huntington's disease mice. Free Radical Biology & Medicine 146, 372382.CrossRefGoogle ScholarPubMed
Wold, MS et al. (2016) ULK1-mediated phosphorylation of ATG14 promotes autophagy and is impaired in Huntington's disease models. Molecular Neurodegeneration 11, 76.CrossRefGoogle ScholarPubMed
Zhang, Y, Sowers, JR and Ren, J (2018) Targeting autophagy in obesity: from pathophysiology to management. Nature reviews. Endocrinology 14, 356376.CrossRefGoogle ScholarPubMed
Oga, EA and Eseyin, OR (2016) The obesity paradox and heart failure: a systematic review of a decade of evidence. Journal of Obesity 2016, 9040248.CrossRefGoogle ScholarPubMed
Kovsan, J et al. (2011) Altered autophagy in human adipose tissues in obesity. The Journal of Clinical Endocrinology and Metabolism 96, E268E277.CrossRefGoogle ScholarPubMed
Zhang, X et al. (2008) Hypothalamic IKKbeta/NF-kappaB and ER stress link overnutrition to energy imbalance and obesity. Cell 135, 6173.CrossRefGoogle ScholarPubMed
Kaushik, S et al. (2012) Loss of autophagy in hypothalamic POMC neurons impairs lipolysis. EMBO reports 13, 258265.CrossRefGoogle ScholarPubMed
Gonzalez, CD et al. (2011) The emerging role of autophagy in the pathophysiology of diabetes mellitus. Autophagy 7, 211.CrossRefGoogle ScholarPubMed
Santos, RX et al. (2014) Insulin therapy modulates mitochondrial dynamics and biogenesis, autophagy and tau protein phosphorylation in the brain of type 1 diabetic rats. Biochimica et Biophysica Acta 1842, 11541166.CrossRefGoogle ScholarPubMed
Brännmark, C et al. (2013) Insulin signaling in type 2 diabetes: experimental and modeling analyses reveal mechanisms of insulin resistance in human adipocytes. The Journal of Biological Chemistry 288, 98679880.CrossRefGoogle ScholarPubMed
Ueno, T and Komatsu, M (2017) Autophagy in the liver: functions in health and disease. Nature Reviews Gastroenterology & Hepatology 14, 170184.CrossRefGoogle ScholarPubMed
Huang, SS et al. (2017) Resveratrol protects podocytes against apoptosis via stimulation of autophagy in a mouse model of diabetic nephropathy. Scientific Reports 7, 45692.CrossRefGoogle Scholar
Zheng, J et al. (2022) cPKCγ Deficiency exacerbates autophagy impairment and hyperphosphorylated Tau buildup through the AMPK/mTOR pathway in mice with type 1 diabetes mellitus. Neuroscience Bulletin 38, 11531169.CrossRefGoogle ScholarPubMed
Jung, HS et al. (2008) Loss of autophagy diminishes pancreatic beta cell mass and function with resultant hyperglycemia. Cell Metabolism 8, 318324.CrossRefGoogle ScholarPubMed
Ebato, C et al. (2008) Autophagy is important in islet homeostasis and compensatory increase of beta cell mass in response to high-fat diet. Cell Metabolism 8, 325332.CrossRefGoogle ScholarPubMed
Jung, HS and Lee, MS (2009) Macroautophagy in homeostasis of pancreatic beta-cell. Autophagy 5, 241243.CrossRefGoogle ScholarPubMed
Marsh, BJ et al. (2007) Regulated autophagy controls hormone content in secretory-deficient pancreatic endocrine beta-cells. Molecular Endocrinology (Baltimore, Md.) 21, 22552269.CrossRefGoogle ScholarPubMed
Quan, W et al. (2012) Autophagy deficiency in beta cells leads to compromised unfolded protein response and progression from obesity to diabetes in mice. Diabetologia 55, 392403.CrossRefGoogle ScholarPubMed
Kaniuk, NA et al. (2007) Ubiquitinated-protein aggregates form in pancreatic beta-cells during diabetes-induced oxidative stress and are regulated by autophagy. Diabetes 56, 930939.CrossRefGoogle ScholarPubMed
Shigihara, N et al. (2014) Human IAPP-induced pancreatic β cell toxicity and its regulation by autophagy. The Journal of Clinical Investigation 124, 36343644.CrossRefGoogle ScholarPubMed
Jansen, HJ et al. (2012) Autophagy activity is up-regulated in adipose tissue of obese individuals and modulates proinflammatory cytokine expression. Endocrinology 153, 58665874.CrossRefGoogle ScholarPubMed
Kosacka, J et al. (2015) Autophagy in adipose tissue of patients with obesity and type 2 diabetes. Molecular and Cellular Endocrinology 409, 2132.CrossRefGoogle ScholarPubMed
López-Vicario, C et al. (2015) Inhibition of soluble epoxide hydrolase modulates inflammation and autophagy in obese adipose tissue and liver: role for omega-3 epoxides. Proceedings of the National Academy of Sciences of the United States of America 112, 536541.CrossRefGoogle ScholarPubMed
Yan, H, Gao, Y and Zhang, Y (2017) Inhibition of JNK suppresses autophagy and attenuates insulin resistance in a rat model of nonalcoholic fatty liver disease. Molecular Medicine Reports 15, 180186.CrossRefGoogle Scholar
Chang, E et al. (2015) Ezetimibe improves hepatic steatosis in relation to autophagy in obese and diabetic rats. World Journal of Gastroenterology 21, 77547763.CrossRefGoogle ScholarPubMed
Yang, L et al. (2010) Defective hepatic autophagy in obesity promotes ER stress and causes insulin resistance. Cell Metabolism 11, 467478.CrossRefGoogle ScholarPubMed
Komiya, K et al. (2010) Free fatty acids stimulate autophagy in pancreatic β-cells via JNK pathway. Biochemical and Biophysical Research Communications 401, 561567.CrossRefGoogle ScholarPubMed
He, Q et al. (2016) GLP-1 analogue improves hepatic lipid accumulation by inducing autophagy via AMPK/mTOR pathway. Biochemical and Biophysical Research Communications 476, 196203.CrossRefGoogle ScholarPubMed
Lai, CH et al. (2016) Multi-strain probiotics inhibit cardiac myopathies and autophagy to prevent heart injury in high-fat diet-fed rats. International Journal of Medical Sciences 13, 27285.CrossRefGoogle ScholarPubMed
Niso-Santano, M et al. (2019) Natural products in the promotion of healthspan and longevity. Clinical Pharmacology and Translational Medicine 3, 149.Google ScholarPubMed
Fu, R et al. (2018) A novel autophagy inhibitor berbamine blocks SNARE-mediated autophagosome-lysosome fusion through upregulation of BNIP3. Cell Death & Disease 9, 115.CrossRefGoogle ScholarPubMed
Tang, ZH et al. (2018) Identification of a novel autophagic inhibitor cepharanthine to enhance the anti-cancer property of dacomitinib in non-small cell lung cancer. Cancer Letters 412, 19.CrossRefGoogle ScholarPubMed
Wu, MY et al. (2017) Natural autophagy blockers, dauricine (DAC) and daurisoline (DAS), sensitize cancer cells to camptothecin-induced toxicity. Oncotarget 8, 77673.CrossRefGoogle ScholarPubMed
Zhou, J et al. (2015) A novel autophagy/mitophagy inhibitor liensinine sensitizes breast cancer cells to chemotherapy through DNM1L-mediated mitochondrial fission. Autophagy 11, 12591279.CrossRefGoogle ScholarPubMed
Xie, SB, He, XX and Yao, SK (2015) Matrine-induced autophagy regulated by p53 through AMP-activated protein kinase in human hepatoma cells. International Journal of Oncology 47, 517526.CrossRefGoogle ScholarPubMed
Song, L et al. (2017) Natural cyclopeptide RA-XII, a new autophagy inhibitor, suppresses protective autophagy for enhancing apoptosis through AMPK/mTOR/P70S6K pathways in HepG2 cells. Molecules 22, 1934.CrossRefGoogle ScholarPubMed
Cui, Q et al. (2007) Oridonin induced autophagy in human cervical carcinoma HeLa cells through Ras, JNK, and P38 regulation. Journal of Pharmacological Sciences 105, 317325.CrossRefGoogle ScholarPubMed
Roy, B et al. (2014) Role of PI3K/Akt/mTOR and MEK/ERK pathway in Concanavalin A induced autophagy in HeLa cells. Chemico-Biological Interactions 210, 96102.CrossRefGoogle ScholarPubMed
Gao, J et al. (2018) The anticancer effects of ferulic acid is associated with induction of cell cycle arrest and autophagy in cervical cancer cells. Cancer Cell International 18, 19.CrossRefGoogle ScholarPubMed
Kim, TW et al. (2018) Kaempferol induces autophagic cell death via IRE1-JNK-CHOP pathway and inhibition of G9a in gastric cancer cells. Cell Death & Disease 9, 114.CrossRefGoogle ScholarPubMed
Lan, CY et al. (2019) Quercetin facilitates cell death and chemosensitivity through RAGE/PI3K/AKT/mTOR axis in human pancreatic cancer cells. Journal of Food and Drug Analysis 27, 887896.CrossRefGoogle ScholarPubMed
Ye, Y et al. (2016) 3, 3′-Diindolylmethane induces anti-human gastric cancer cells by the miR-30e-ATG5 modulating autophagy. Biochemical Pharmacology 115, 7784.CrossRefGoogle ScholarPubMed
Kaushik, G et al. (2015) Honokiol inhibits melanoma stem cells by targeting notch signaling. Molecular Carcinogenesis 54, 17101721.CrossRefGoogle ScholarPubMed
Park, KR et al. (2022) Paeoniflorigenone regulates apoptosis, autophagy, and necroptosis to induce anti-cancer bioactivities in human head and neck squamous cell carcinomas. Journal of Ethnopharmacology 288, 115000.CrossRefGoogle ScholarPubMed
Park, EJ, Choi, KS and Kwon, TK (2011) β-Lapachone-induced reactive oxygen species (ROS) generation mediates autophagic cell death in glioma U87 MG cells. Chemico-Biological Interactions 189, 3744.CrossRefGoogle ScholarPubMed
Li, YC et al. (2014) Plumbagin induces apoptotic and autophagic cell death through inhibition of the PI3K/Akt/mTOR pathway in human non-small cell lung cancer cells. Cancer Letters 344, 239259.CrossRefGoogle ScholarPubMed
Shailasree, S et al. (2015) Cytotoxic effect of p-coumaric acid on neuroblastoma, N2a cell via generation of reactive oxygen species leading to dysfunction of mitochondria inducing apoptosis and autophagy. Molecular Neurobiology 51, 119130.CrossRefGoogle ScholarPubMed
Wang, G et al. (2017) The novel autophagy inhibitor elaiophylin exerts antitumor activity against multiple myeloma with mutant TP53 in part through endoplasmic reticulum stress-induced apoptosis. Cancer Biology & Therapy 18, 584595.CrossRefGoogle ScholarPubMed
Aoki, H et al. (2007) Evidence that curcumin suppresses the growth of malignant gliomas in vitro and in vivo through induction of autophagy: role of Akt and extracellular signal-regulated kinase signaling pathways. Molecular Pharmacology 72, 2939.CrossRefGoogle ScholarPubMed
Teiten, MH et al. (2011) Anti-proliferative potential of curcumin in androgen-dependent prostate cancer cells occurs through modulation of the wingless signaling pathway. International Journal of Oncology 38, 603611.Google ScholarPubMed
Lee, YJ et al. (2011) Involvement of ROS in curcumin-induced autophagic cell death. The Korean Journal of Physiology & Pharmacology 15, 17.CrossRefGoogle ScholarPubMed
Kim, JY et al. (2012) Curcumin-induced autophagy contributes to the decreased survival of oral cancer cells. Archives of Oral Biology 57, 10181025.CrossRefGoogle Scholar
Zhu, Y and Bu, S (2017) Curcumin induces autophagy, apoptosis, and cell cycle arrest in human pancreatic cancer cells. Evidence-Based Complementary and Alternative Medicine 2017, 5787218.CrossRefGoogle ScholarPubMed
Liu, J et al. (2017) Curcumin sensitizes prostate cancer cells to radiation partly via epigenetic activation of miR-143 and miR-143 mediated autophagy inhibition. Journal of Drug Targeting 25, 645652.CrossRefGoogle ScholarPubMed
Muniraj, N et al. (2019) Withaferin A inhibits lysosomal activity to block autophagic flux and induces apoptosis via energetic impairment in breast cancer cells. Carcinogenesis 40, 11101120.Google Scholar
Zhang, H et al. (2022) Periplocin induces apoptosis of pancreatic cancer cells through autophagy via the AMPK/mTOR pathway. Journal of Oncology 2022, 8055004.Google ScholarPubMed
Tian, Y et al. (2019) Resveratrol as a natural regulator of autophagy for prevention and treatment of cancer. OncoTargets and Therapy 17, 86018609.CrossRefGoogle Scholar
Fu, Y et al. (2014) Resveratrol inhibits breast cancer stem-like cells and induces autophagy via suppressing Wnt/β-catenin signaling pathway. PLoS ONE 9, e102535.CrossRefGoogle ScholarPubMed
Miki, H et al. (2012) Resveratrol induces apoptosis via ROS-triggered autophagy in human colon cancer cells. International Journal of Oncology 40, 10201028.CrossRefGoogle ScholarPubMed
Ferraresi, A et al. (2017) Resveratrol inhibits IL-6-induced ovarian cancer cell migration through epigenetic up-regulation of autophagy. Molecular Carcinogenesis 56, 11641181.CrossRefGoogle ScholarPubMed
Xu, J et al. (2019) Corilagin induces apoptosis, autophagy and ROS generation in gastric cancer cells in vitro. International Journal of Molecular Medicine 43, 967979.Google ScholarPubMed
Wang, SG et al. (2013) Punicalagin induces apoptotic and autophagic cell death in human U87MG glioma cells. Acta Pharmacologica Sinica 34, 14111419.CrossRefGoogle ScholarPubMed
Chen, SY et al. (2022) Lucidone inhibits autophagy and MDR1 via HMGB1/RAGE/PI3K/Akt signaling pathway in pancreatic cancer cells. Phytotherapy Research 36, 16641677.CrossRefGoogle ScholarPubMed
Guo, J et al. (2015) Celastrol induces autophagy by targeting AR/miR-101 in prostate cancer cells. PLoS ONE 10, e0140745.CrossRefGoogle ScholarPubMed
Zhang, J et al. (2022) Toosendanin and isotoosendanin suppress triple-negative breast cancer growth via inducing necrosis, apoptosis and autophagy. Chemico-Biological Interactions 351, 109739.CrossRefGoogle ScholarPubMed
Wu, X et al. (2021) Autophagy and cardiac diseases: therapeutic potential of natural products. Medicinal Research Reviews 41, 314341.CrossRefGoogle ScholarPubMed
Huang, Z et al. (2015) Berberine alleviates cardiac ischemia/reperfusion injury by inhibiting excessive autophagy in cardiomyocytes. European Journal of Pharmacology 762, 110.CrossRefGoogle ScholarPubMed
Zeng, Z et al. (2019) Myocardial hypertrophy is improved with berberine treatment via long non-coding RNA MIAT-mediated autophagy. Journal of Pharmacy and Pharmacology 71, 18221831.CrossRefGoogle ScholarPubMed
Wang, X et al. (2019) Tanshinone IIA restores dynamic balance of autophagosome/autolysosome in doxorubicin-induced cardiotoxicity via targeting beclin1/LAMP1. Cancers 11, 910.CrossRefGoogle ScholarPubMed
Xu, M et al. (2019) Oridonin protects against cardiac hypertrophy by promoting P21-related autophagy. Cell Death & Disease 10, 116.CrossRefGoogle ScholarPubMed
Li, X et al. (2018) Inhibition of autophagy via activation of PI3K/Akt/mTOR pathway contributes to the protection of hesperidin against myocardial ischemia/reperfusion injury. International Journal of Molecular Medicine 42, 19171924.Google Scholar
Hu, L et al. (2022) Icariin inhibits isoproterenol-induced cardiomyocyte hypertropic injury through activating autophagy via the AMPK/mTOR signaling pathway. Biochemical and Biophysical Research Communications 593, 6572.CrossRefGoogle ScholarPubMed
Hu, J et al. (2016) Luteolin alleviates post-infarction cardiac dysfunction by up-regulating autophagy through Mst1 inhibition. Journal of Cellular and Molecular Medicine 20, 147156.CrossRefGoogle ScholarPubMed
Wu, X et al. (2017) Nobiletin attenuates adverse cardiac remodeling after acute myocardial infarction in rats via restoring autophagy flux. Biochemical and Biophysical Research Communications 492, 262268.CrossRefGoogle ScholarPubMed
Liu, B et al. (2015) Puerarin prevents cardiac hypertrophy induced by pressure overload through activation of autophagy. Biochemical and Biophysical Research Communications 464, 908915.CrossRefGoogle ScholarPubMed
Chen, WR et al. (2018) Melatonin attenuates myocardial ischemia/reperfusion injury by inhibiting autophagy via an AMPK/mTOR signaling pathway. Cellular Physiology and Biochemistry : International Journal of Experimental Cellular Physiology, Biochemistry, and Pharmacology 47, 20672076.Google Scholar
Xie, S et al. (2015) Melatonin protects against chronic intermittent hypoxia-induced cardiac hypertrophy by modulating autophagy through the 5′ adenosine monophosphate-activated protein kinase pathway. Biochemical and Biophysical Research Communications 464, 975981.CrossRefGoogle ScholarPubMed
Xiao, H et al. (2022) Hinokitiol protects cardiomyocyte from oxidative damage by inhibiting GSK3β-mediated autophagy. Oxidative Medicine and Cellular Longevity 2022, 113.Google ScholarPubMed
Liu, H et al. (2019) Role of thymoquinone in cardiac damage caused by sepsis from BALB/c mice. Inflammation 42, 516525.CrossRefGoogle ScholarPubMed
Yan, X et al. (2019) Gallic acid suppresses cardiac hypertrophic remodeling and heart failure. Molecular Nutrition & Food Research 63, 1800807.CrossRefGoogle ScholarPubMed
Eisenberg, T et al. (2016) Cardioprotection and lifespan extension by the natural polyamine spermidine. Nature Medicine 22, 14281438.CrossRefGoogle ScholarPubMed
Yan, J et al. (2019) Spermidine-enhanced autophagic flux improves cardiac dysfunction following myocardial infarction by targeting the AMPK/mTOR signalling pathway. British Journal of Pharmacology 176, 31263142.Google ScholarPubMed
Liu, R et al. (2018) Curcumin alleviates isoproterenol-induced cardiac hypertrophy and fibrosis through inhibition of autophagy and activation of mTOR. European Review for Medical and Pharmacological Sciences 22, 75007508.Google ScholarPubMed
Ba, L et al. (2019) Allicin attenuates pathological cardiac hypertrophy by inhibiting autophagy via activation of PI3K/Akt/mTOR and MAPK/ERK/mTOR signaling pathways. Phytomedicine: International Journal of Phytotherapy and Phytopharmacology 58, 152765.CrossRefGoogle ScholarPubMed
Gu, J et al. (2018) Resveratrol suppresses doxorubicin-induced cardiotoxicity by disrupting E2F1 mediated autophagy inhibition and apoptosis promotion. Biochemical Pharmacology 150, 202213.CrossRefGoogle ScholarPubMed
Xu, X et al. (2012) Resveratrol attenuates doxorubicin-induced cardiomyocyte death via inhibition of p70 S6 kinase 1-mediated autophagy. Journal of Pharmacology and Experimental Therapeutics 341, 183195.CrossRefGoogle ScholarPubMed
Sun, GZ et al. (2020) Ginsenoside Rg3 protects heart against isoproterenol-induced myocardial infarction by activating AMPK mediated autophagy. Cardiovascular Diagnosis and Therapy 10, 153160.CrossRefGoogle ScholarPubMed
Xiao, Y et al. (2017) Cucurbitacin B protects against pressure overload induced cardiac hypertrophy. Journal of Cellular Biochemistry 118, 38993910.CrossRefGoogle ScholarPubMed
Huang, M et al. (2017) Berberine improves cognitive impairment by promoting autophagic clearance and inhibiting production of β-amyloid in APP/tau/PS1 mouse model of Alzheimer's disease. Experimental Gerontology 91, 2533.CrossRefGoogle ScholarPubMed
Umezawa, K (2018) Therapeutic activity of plant-derived alkaloid conophylline on metabolic syndrome and neurodegenerative disease models. Human Cell 31, 95101.CrossRefGoogle ScholarPubMed
Wang, C et al. (2014) Downregulation of PI3K/Akt/mTOR signaling pathway in curcumin-induced autophagy in APP/PS1 double transgenic mice. European Journal of Pharmacology 740, 312320.CrossRefGoogle ScholarPubMed
Fan, Y (2017) Identification of natural products with neuronal and metabolic benefits through autophagy induction. Autophagy 13, 4156.CrossRefGoogle ScholarPubMed
Yang, C et al. (2022) Celastrol enhances transcription factor EB (TFEB)-mediated autophagy and mitigates Tau pathology: implications for Alzheimer's disease therapy. Acta Pharmaceutica Sinica B 12, 17071722.CrossRefGoogle ScholarPubMed
Moon, J et al. (2014) Caffeine prevents human prion-protein-mediated neurotoxicity through the induction of autophagy. International Journal of Molecular Medicine 34, 553558.CrossRefGoogle ScholarPubMed
Li, LS et al. (2017) Dendrobium nobile Lindl alkaloid, a novel autophagy inducer, protects against axonal degeneration induced by Aβ25-35 in hippocampus neurons in vitro. CNS Neuroscience & Therapeutics 23, 329340.CrossRefGoogle ScholarPubMed
Liang, J et al. (2019) Enhancing the retrograde axonal transport by curcumin promotes autophagic flux in N2a/APP695swe cells. Aging (Albany NY 11, 70367050.Google ScholarPubMed
Zhang, L et al. (2018) The potential protective effect of curcumin on amyloid-β-42 induced cytotoxicity in HT-22 cells. BioMed Research International 2018, 8134902.Google ScholarPubMed
Wang, N (2020) β-Asarone inhibits amyloid-β by promoting autophagy in a cell model of Alzheimer's disease. Frontiers in Pharmacology 10, 1529.CrossRefGoogle Scholar
Li, X, Song, J and Dong, R (2019) Cubeben induces autophagy via PI3K-AKT-mTOR pathway to protect primary neurons against amyloid beta in Alzheimer's disease. Cytotechnology 71, 679686.CrossRefGoogle ScholarPubMed
Zhu, Y and Wang, J (2015) Wogonin increases β-amyloid clearance and inhibits tau phosphorylation via inhibition of mammalian target of rapamycin: potential drug to treat Alzheimer's disease. Neurological Sciences 36, 11811188.CrossRefGoogle ScholarPubMed
Kuang, L, Cao, X and Lu, Z (2017) Baicalein protects against rotenone-induced neurotoxicity through induction of autophagy. Biological and Pharmaceutical Bulletin 40, 15371543.CrossRefGoogle ScholarPubMed
Chaouhan, HS et al. (2022) Calycosin alleviates paraquat-induced neurodegeneration by improving mitochondrial functions and regulating autophagy in a drosophila model of Parkinson's disease. Antioxidants 11, 222.CrossRefGoogle Scholar
El-Horany, HE et al. (2016) 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). Journal of Biochemical and Molecular Toxicology 30, 360369.CrossRefGoogle ScholarPubMed
Hsueh, KW et al. (2016) Autophagic down-regulation in motor neurons remarkably prolongs the survival of ALS mice. Neuropharmacology 108, 152160.CrossRefGoogle ScholarPubMed
Achour, I et al. (2016) Oleuropein prevents neuronal death, mitigates mitochondrial superoxide production and modulates autophagy in a dopaminergic cellular model. International Journal of Molecular Sciences 17, 1293.CrossRefGoogle Scholar
Rigacci, S et al. (2015) Oleuropein aglycone induces autophagy via the AMPK/mTOR signalling pathway: a mechanistic insight. Oncotarget 6, 35344.CrossRefGoogle ScholarPubMed
Vidoni, C et al. (2018) Resveratrol protects neuronal-like cells expressing mutant Huntingtin from dopamine toxicity by rescuing ATG4-mediated autophagosome formation. Neurochemistry International 117, 174187.CrossRefGoogle ScholarPubMed
Lin, TK et al. (2014) Resveratrol partially prevents rotenone-induced neurotoxicity in dopaminergic SH-SY5Y cells through induction of heme oxygenase-1 dependent autophagy. International Journal of Molecular Sciences 15, 16251646.CrossRefGoogle ScholarPubMed
Deng, H and Mi, MT (2016) Resveratrol attenuates Aβ25–35 caused neurotoxicity by inducing autophagy through the TyrRS-PARP1-SIRT1 signaling pathway. Neurochemical Research 41, 23672379.CrossRefGoogle ScholarPubMed
Guo, YJ et al. (2016) Resveratrol alleviates MPTP-induced motor impairments and pathological changes by autophagic degradation of α-synuclein via SIRT1-deacetylated LC3. Molecular Nutrition & Food Research 60, 21612175.CrossRefGoogle ScholarPubMed
Russo, R et al. (2014) Role of D-Limonene in autophagy induced by bergamot essential oil in SH-SY5Y neuroblastoma cells. PLoS ONE 9, e113682.CrossRefGoogle ScholarPubMed
Arel-Dubeau, AM (2014) Cucurbitacin E has neuroprotective properties and autophagic modulating activities on dopaminergic neurons. Oxidative Medicine and Cellular Longevity 2014, 115.CrossRefGoogle ScholarPubMed
Rekha, K and Sivakamasundari, R (2018) Geraniol protects against the protein and oxidative stress induced by rotenone in an in vitro model of Parkinson's disease. Neurochemical Research 43, 19471962.CrossRefGoogle Scholar
Wang, Z et al. (2022) Cannabidiol induces autophagy and improves neuronal health associated with SIRT1 mediated longevity. GeroScience 44, 15051524.CrossRefGoogle ScholarPubMed
Deng, Y et al. (2014) Berberine attenuates autophagy in adipocytes by targeting BECN1. Autophagy 10, 17761786.CrossRefGoogle ScholarPubMed
Baur, JA et al. (2006) Resveratrol improves health and survival of mice on a high-calorie diet. Nature 444, 337342.CrossRefGoogle ScholarPubMed
Wang, B et al. (2014) Resveratrol-enhanced autophagic flux ameliorates myocardial oxidative stress injury in diabetic mice. Journal of Cellular and Molecular Medicine 18, 15991611.CrossRefGoogle ScholarPubMed
Qu, X et al. (2019) Resveratrol alleviates ischemia/reperfusion injury of diabetic myocardium via inducing autophagy. Experimental and Therapeutic Medicine 18, 27192725.Google ScholarPubMed
Yan, J et al. (2012) Enhanced autophagy plays a cardinal role in mitochondrial dysfunction in type 2 diabetic Goto–Kakizaki (GK) rats: ameliorating effects of (−)-epigallocatechin-3-gallate. The Journal of Nutritional Biochemistry 23, 716724.CrossRefGoogle Scholar
Xu, C et al. (2019) Trehalose restores functional autophagy suppressed by high glucose. Reproductive Toxicology 85, 5158.CrossRefGoogle ScholarPubMed
Shi, L et al. (2015) Dihydromyricetin improves skeletal muscle insulin sensitivity by inducing autophagy via the AMPK-PGC-1α-Sirt3 signaling pathway. Endocrine 50, 378389.CrossRefGoogle ScholarPubMed
Xu, X et al. (2020) The effects of puerarin on autophagy through regulating of the PERK/eIF2α/ATF4 signaling pathway influences renal function in diabetic nephropathy. Diabetes, Metabolic Syndrome and Obesity: Targets and Therapy 13, 25832592.CrossRefGoogle ScholarPubMed
Zhang, M et al. (2017) Melatonin protects against diabetic cardiomyopathy through Mst1/Sirt3 signaling. Journal of Pineal Research 63, e12418.CrossRefGoogle ScholarPubMed
Ma, R et al. (2022) Ferulic acid ameliorates renal injury via improving autophagy to inhibit inflammation in diabetic nephropathy mice. Biomedicine & Pharmacotherapy 153, 113424.CrossRefGoogle ScholarPubMed
Zhang, XX et al. (2022) Arjunolic acid from Cyclocarya paliurus ameliorates diabetic retinopathy through AMPK/mTOR/HO-1 regulated autophagy pathway. Journal of Ethnopharmacology 284, 114772.CrossRefGoogle ScholarPubMed
Yao, Q et al. (2018) Curcumin protects against diabetic cardiomyopathy by promoting autophagy and alleviating apoptosis. Journal of Molecular and Cellular Cardiology 124, 2634.CrossRefGoogle ScholarPubMed
Zhang, P et al. (2020) Curcumin inhibited podocyte cell apoptosis and accelerated cell autophagy in diabetic nephropathy via regulating Beclin1/UVRAG/Bcl2. Diabetes, Metabolic Syndrome and Obesity: Targets and Therapy 13, 641.CrossRefGoogle ScholarPubMed
Figure 0

Figure 1. The types of autophagy (Figure created with Biorender.com).

Figure 1

Figure 2. The molecular signalling mechanisms of autophagy (Figure created with Biorender. com).

Figure 2

Table 1. Studies on autophagy in cancer

Figure 3

Table 2. Studies on autophagy in cardiovascular diseases

Figure 4

Table 3. Studies on autophagy in neurodegenerative diseases

Figure 5

Table 4. Studies on autophagy in obesity and diabetes

Figure 6

Figure 3. Autophagy-related pathways regulated by natural compounds in cancer (Figure created with Biorender.com).

Figure 7

Table 5. In vitro studies on natural compounds modulating autophagy in various cancers signalling pathways

Figure 8

Figure 4. Autophagy-related pathways regulated by natural compounds in cardiac disease (Figure created with Biorender.com).

Figure 9

Table 6. Autophagy regulating natural compounds in cardiac diseases

Figure 10

Figure 5. Autophagy-related pathways regulated by natural compounds in neurological aging (Figure created with Biorender.com).

Figure 11

Table 7. Autophagy regulating natural compounds in neurological aging

Figure 12

Figure 6. Autophagy-related pathways regulated by natural compounds in obesity and diabetes (Figure created with Biorender.com).

Figure 13

Table 8. Autophagy regulating natural compounds in obesity and diabetes