Skip to main content

Autophagy: a double-edged sword in ischemia–reperfusion injury

Abstract

Ischemia–reperfusion (I/R) injury describes the pathological process wherein tissue damage, initially caused by insufficient blood supply (ischemia), is exacerbated upon the restoration of blood flow (reperfusion). This phenomenon can lead to irreversible tissue damage and is commonly observed in contexts such as cardiac surgery and stroke, where blood supply is temporarily obstructed. During ischemic conditions, the anaerobic metabolism of tissues and organs results in compromised enzyme activity. Subsequent reperfusion exacerbates mitochondrial dysfunction, leading to increased oxidative stress and the accumulation of reactive oxygen species (ROS). This cascade ultimately triggers cell death through mechanisms such as autophagy and mitophagy. Autophagy constitutes a crucial catabolic mechanism within eukaryotic cells, facilitating the degradation and recycling of damaged, aged, or superfluous organelles and proteins via the lysosomal pathway. This process is essential for maintaining cellular homeostasis and adapting to diverse stress conditions. As a cellular self-degradation and clearance mechanism, autophagy exhibits a dualistic function: it can confer protection during the initial phases of cellular injury, yet potentially exacerbate damage in the later stages. This paper aims to elucidate the fundamental mechanisms of autophagy in I/R injury, highlighting its dual role in regulation and its effects on both organ-specific and systemic responses. By comprehending the dual mechanisms of autophagy and their implications for organ function, this study seeks to explore the potential for therapeutic interventions through the modulation of autophagy within clinical settings.

Introduction

Ischemia–reperfusion (I/R) injury constitutes a pathological cascade initiated by the reestablishment of blood flow to previously hypoxic tissues, presenting a significant challenge to multiple organ systems [1]. It exacerbates morbidity and mortality across a spectrum of diseases, including myocardial infarction, ischemic stroke, acute kidney injury (AKI), trauma, circulatory failure, sickle cell disease, and sleep apnea [2]. Furthermore, it may precipitate pathological responses such as systemic inflammatory response syndrome (SIRS) and multiple organ dysfunction syndrome (MODS) when organ ischemia results in a disequilibrium between metabolic supply and demand [3, 4]. In the context of cardiac function, the reduction in oxygen supply to ischemic tissues is intricately linked to a decline in mitochondrial oxidative phosphorylation, subsequently causing a transition from aerobic to anaerobic metabolism [5]. Consequently, ischemic injury has emerged as a significant challenge in organ transplantation as well as in cardiothoracic, vascular, and general surgical procedures. The objective of reperfusion is to avert cell death induced by ischemia, sustain cellular metabolism, and facilitate the removal of metabolic waste by reinstating the supply of oxygen and nutrients [6]. Nonetheless, it is crucial to acknowledge that the reintroduction of blood flow following prolonged ischemia can potentially exacerbate tissue damage, manifesting as heightened local inflammation, mitochondrial dysfunction, and an acute surge in reactive oxygen species (ROS) production [6, 7] Furthermore, oxidative stress and the initiation of self-damaging tissue responses are also pivotal mechanisms underlying I/R injury [6]. Over recent decades, oxidative stress has garnered significant attention as a pivotal contributor to cell death and tissue damage in I/R injury, particularly in the context of elucidating molecular mechanisms [8]. The reperfusion of blood following ischemia intensifies both specific and nonspecific immune responses within cells, resulting in the release of cytokines and chemokines, inflammation, and excessive apoptosis. These processes collectively trigger a cascade of detrimental cell death pathways, including the activation of autophagy [9].

Autophagy is a crucial cellular cyclic mechanism essential for preserving intracellular homeostasis [10,11,12]. Functioning as the primary intracellular degradation system, it facilitates the entry of cytoplasmic material into the lysosome via the autophagic pathway, where degradation occurs [3]. The objective of autophagy extends beyond mere material elimination; it operates as a dynamic circulatory system that salvages deteriorating cells and supplies new building blocks and energy for cellular repair and homeostasis by degrading and recycling damaged cellular components [3, 12]. For instance, the inhibition of NOD-like receptor protein 3 (NLRP3) inflammasome activity can enhance the occurrence of mitophagy, consequently mitigating apoptosis during AKI and offering a potential novel target for AKI treatment [5]. Mitophagy generally facilitates cellular adaptation and protection through various mechanisms, including the elimination of damaged mitochondria [13, 14]. In certain instances, excessive autophagy can result in cell death, exemplified by the generation of mitochondrial bursts of ROS following I/R. These cell death modalities encompass necrosis, mitochondrial permeability transition-driven necrosis, ferroptosis, pyroptosis, para-apoptosis, cuproptosis, apoptosis, mitophagy, and autophagy, among others [3, 12]. Therefore, autophagy functions as a double-edged sword, serving both as a mechanism for cellular survival and as a potential pathway to cell death [10].

This review examines the dual role of autophagy in I/R injury, positing that it exhibits both protective and deleterious effects. During ischemic conditions, autophagy mitigates cellular damage and preserves cellular integrity by removing dysfunctional organelles and proteins. Conversely, during reperfusion, autophagy can potentially exacerbate damage and contribute to cell death. Through a comprehensive analysis of autophagy and its regulatory mechanisms in the context of I/R injury, this study underscores the critical importance of understanding the regulatory pathways governing autophagy in I/R injury. We anticipate leveraging its protective effects in therapeutic applications while minimizing potential adverse effects, thereby offering targeted intervention strategies to optimize organ protection and recovery processes.

The fundamental mechanism of autophagy

Autophagy is an intricate intracellular self-degradation mechanism meticulously governed by a multitude of regulators and signaling pathways, which collectively dictate the initiation, progression, and termination of the process [15]. The most widely recognized form of autophagy involves the extensive processing of cytoplasmic components via the autophagosome-dependent lysosomal pathway, commonly referred to as macroautophagy [16]. In summary, the macroautophagy process comprises several distinct stages: initially, the formation of the phagophore occurs, which is succeeded by the expansion of the autophagosome membrane. This is followed by the fusion of autophagosomes with lysosomes, culminating in the degradation of the sequestered components within the autophagosome [17]. Beyond macroautophagy, the autophagic process also encompasses chaperone-mediated autophagy (CMA) and microautophagy [18]. In the course of the CMA process, the specific degraded protein associates with the chaperone heat shock cognate (HSC) protein 70 via a distinct amino acid sequence known as the KFERQ motif, which facilitates protein degradation within the CMA pathway. This interaction subsequently enables the protein’s translocation into the lysosome through its interaction with lysosome-associated membrane protein (LAMP)2A [18]. In contrast, microautophagy entails the direct engulfment of cytoplasmic components by lysosomes or the invagination of the endoplasmic reticulum [19]. Macroautophagy constitutes a sophisticated intracellular degradation pathway characterized by a multistep mechanism involving numerous critical proteins, which is crucial for preserving cellular homeostasis [20]. The initiation of autophagosome formation in macroautophagy is triggered by the activation of the Unc-51-like autophagy-activating kinase 1 (ULK1) complex, a pivotal component in the autophagy initiation phase, subsequently leading to the development of double-membrane vesicles that encapsulate cellular debris [19]. Adenosine monophosphate (AMP)-activated protein kinase (AMPK) functions as a critical cellular energy sensor and regulator, responding to fluctuations in the intracellular AMP to adenosine triphosphate (ATP) ratio by modulating metabolic pathways to accommodate variations in energy availability. During the initiation of autophagy, AMPK inhibits the mammalian target of rapamycin complex 1 (mTORC1), a principal regulator of cell growth and proliferation, and facilitates the formation of autophagic vesicles by alleviating the inhibition of the ULK1 complex [19, 21]. Subsequently, the ULK1 complex translocates to the endoplasmic reticulum, where phosphatidylinositol 3-kinase (PI3K) III, also referred to as vacuolar protein sorting (Vps) 34, serves as a pivotal enzyme in the regulation of mammalian endocytosis, lysogenesis, autophagy, and intracellular trafficking. Notably, Vps34 is essential during both the initiation and maturation phases of autophagy, facilitating the formation of autophagosomes through the production of phosphatidylinositol 3-phosphate (PI3P). This process recruits autophagy-related proteins such as Beclin-1, WIPI2, and DFCP1ULK1 [22]. The Beclin-1/Vps34 complex facilitates the expansion of autophagic vesicles. This process is initiated by the phosphorylation of B-cell lymphoma 2 (BCL-2) and BCL-2-interacting cell death mediator (BIM) by activated JNK kinases, leading to the release of Beclin-1 and the subsequent dissociation of the Beclin-1/BCL-2 and BIM complexes. The liberated Beclin-1 subsequently activates Vps34, forming a complex that produces PI3P, thereby promoting the elongation of autophagic vesicles [23]. Currently, autophagy-related genes (ATGs) and protein complexes, such as ATG5–ATG12, which involve ATG7, ATG3, and ATG5–ATG12, facilitate autophagy via a ubiquitin-like covalent binding mechanism. This process subsequently enhances ATG8/microtubule-associated protein 1 light chain 3 (LC3) binding, thereby promoting the expansion and closure of autophagosomes [24]. Furthermore, the ATG12–ATG5 complex can associate with ATG16L to form a polymer complex essential for autophagosome assembly [25]. During the elongation phase, the Beclin-1 and PI3K complexes play a crucial role in coordinating the nucleation of the autophagosomal membrane [9]. LC3 plays a pivotal role in the autophagy pathway, wherein it transitions from its cytosolic form (LC3-I) to a membrane-bound form (LC3-II), a process crucial for the formation and maturation of autophagosomes. Initially, the cysteine protease ATG4 cleaves LC3 to generate LC3-I, which is then processed by ATG3, ATG7, and phosphatidylethanolamine to form LC3-II. Following this, LC3-II is amplified, the ESCRT complex facilitates the completion of the closure phase, and LC3-II becomes integrated into the autophagosomal membrane [24]. In the terminal phase, autophagosomes merge with lysosomes to form autophagolysosomes, a process predominantly facilitated by the soluble N-ethylmaleimide-sensitive factor attachment protein receptors (SNARE) complex. This complex plays a crucial role in regulating endomembrane fusion events, particularly in the formation of proteins within the secretory pathway and during endocytosis. STX17 (a t-SNARE protein), VAMP8 (a v-SNARE protein), and SNAP29 (a member of the SNAP family) interact to facilitate the trafficking of complexes to the autophagosomal membrane and the subsequent fusion of lysosomes and autophagosomes. This process culminates in the completion of autophagy through the degradation and recycling of cellular components [10, 23]. As discussed above, during this process, the autophagosome recruits lysosomal fusion proteins while the ATG proteins on its outer membrane are sequentially removed. During this process, STX17 undergoes deacetylation, resulting in the embedding of its C-terminal hairpin-like structure within the autophagosome membrane. This structural configuration facilitates interactions with SNAP29 and the HOPS complex, a substantial protein assembly consisting of six core subunits, thereby promoting the fusion of autophagosomes with lysosomes. Consequently, the damaged organelle components are degraded into smaller molecules within the fused autophagosome-lysosome structures and subsequently recycled [10, 22, 26]. To date, macroautophagy has effectively facilitated the lysosomal degradation of target substrates, encompassing protein aggregates, damaged organelles such as mitochondria and peroxisomes, carbohydrates, lipids, nucleic acids, and pathogens [27]. During this process, lysosomes break down complex molecules and release amino acids, fatty acids, and nucleotides, underscoring the significance of autophagy as a mechanism integral to the metabolic precursor cycle [11].

The selective phagocytosis of cytoplasmic material by autophagosomes is governed by highly specific and genetically regulated mechanisms, collectively referred to as selective autophagy; an example of this is the targeted autophagic degradation of mitochondria [16, 24]. Mitochondria, characterized by their double-membrane structure, primarily facilitate ATP production and regulate cellular energy metabolism [28]. Beyond their role as energy producers, mitochondria also participate in diverse physiological processes, including the mediation of Ca2+ signaling in most cells [29]. In cardiomyocytes, mitochondria can account for over 30% of the cell volume to satisfy their consistently elevated energy demands. Nonetheless, mitochondria are vulnerable to cellular stressors, including hypoxia, which can result in the generation of ROS and the release of pro-apoptotic proteins. These processes may ultimately culminate in mitochondrial damage and potentially lead to cell death [28]. Mitochondrial quality control represents a critical mechanism in the regulation of mitochondrial size, quantity, morphology, quality, and biological activity [30], playing a pivotal role in sustaining cellular homeostasis and survival. This process encompasses mitochondrial biogenesis, fusion, fission, and mitophagy [25, 28]. In response to cellular stress-induced mitochondrial damage, cells initially preserve their structural integrity and composition through mechanisms including antioxidative defense, DNA repair, protein folding, and degradation [31, 32]. Should the initial defense mechanisms prove inadequate, an extensive quality control system encompassing mitochondrial biogenesis, fusion, fission, and mitophagy is subsequently activated [31, 32]. In instances where damaged mitochondria are irreparable, mitophagy serves as the final defense mechanism to eliminate compromised mitochondria and preserve cellular viability before the onset of apoptosis and necrosis [8]. Thus, the processes of mitochondrial biogenesis, clearance, dynamics, and their interactions collectively form a robust quality control system that responds to pathological stress and sustains mitochondrial function [28]. Currently, mitophagy pathways encompass both the PINK1-Parkin-mediated and the PINK1-Parkin-independent mitochondrial autophagy pathways [33]. These pathways influence the dual role of I/R injury in the precise regulation of the autophagic process [34]. The specific mechanisms underlying these pathways will be elaborated upon in the subsequent sections. The role of gender in autophagy represents a complex and significant area of research. Notably, studies have indicated that female patients with Alzheimer’s disease exhibit a more pronounced accumulation of autophagosomes, autophagic degradation, and mitophagy compared with their male counterparts. This suggests that, owing to greater mitochondrial or protein damage, female patients may engage in compensatory autophagy, potentially accelerating the pathogenesis of Alzheimer’s disease [35]. Furthermore, it is crucial to address not only the differential impact of diseases across genders but also the underrepresentation of women and gender minorities in scientific research. These groups require increased support and opportunities within the scientific community [36]. Further investigation is essential to elucidate the mechanisms driving gender differences in autophagy and to develop personalized therapeutic strategies targeting these pathways.

Additionally, during ischemia–reperfusion, autophagy may serve as a protective mechanism in early ischemic conditions by eliminating damaged organelles and proteins, thereby preventing the accumulation of toxic substances. However, during reperfusion, excessive autophagic activity may result in the accumulation of undigested materials within autophagic lysosomes, potentially leading to cellular damage. Currently, there exists a significant gap in research concerning the translation of mitochondrial autophagy mechanisms into effective targeted pharmacological interventions. The majority of mitochondrial autophagy inducers currently available are primarily mitochondrial uncoupling agents or mitochondrial toxins, which present numerous limitations. Furthermore, the clinical efficacy of mitophagy modulators remains to be thoroughly validated [37]. A comprehensive understanding of the molecular mechanisms that govern the transition of autophagy from protective to deleterious effects is essential for the development of targeted therapeutic strategies. Such strategies aim to modulate autophagy in the context of I/R injury to promote cellular recovery rather than destruction. A schematic diagram of the basic mechanistic pathway of autophagy is shown in Fig. 1.

Fig. 1
figure 1

The basic mechanism of autophagy. Autophagy is a cellular process responsible for the degradation and recycling of intracellular components, encompassing three primary forms: macroautophagy, microautophagy, and CMA. Macroautophagy, in particular, serves as an intracellular self-digestion mechanism characterized by a complex sequence of events involving the coordinated action of multiple key proteins. This process can be delineated into four distinct stages: initiation of the autophagosome, elongation of the autophagosome, maturation or blocking of the autophagosome, and the fusion of autophagosomes with lysosomes. (Created using Adobe Illustrator)

The protective function of autophagy in I/R injury

Autophagy potentially serves a protective function in preserving cellular integrity during I/R injury, particularly during the ischemic phase [3, 12]. It functions as an intracellular “cleaner” by eliminating dysfunctional organelles and misfolded proteins, thereby removing debris that could otherwise result in cell death [31]. For instance, during the early stages of tumorigenesis, autophagy contributes to an antitumor response by engaging in oxidative stress management and eliminating dysregulated cells, which helps maintain genomic stability and inhibit tissue damage and inflammation [38]. This clearance mechanism holds significant importance within the central nervous system, as ischemic injury swiftly disturbs the intricate equilibrium of the neuronal environment [33]. During the reperfusion phase, autophagy assumes a crucial role in preserving homeostasis within the intracellular milieu as oxygen and nutrients are reintroduced [10, 11]. It facilitates the recycling of cellular components into usable substrates, thereby supporting ATP production and promoting cellular recovery [20]. This metabolic reorganization represents not merely a response to energy expenditure but also an active strategy that equips cells to address the challenges associated with reperfusion, including oxidative stress and inflammation [27]. Clinical research has demonstrated that intravascular reperfusion therapies, such as intravenous thrombolysis or mechanical thrombus extraction, administered within a defined time window, constitute a relatively safe and restorative intervention for patients experiencing acute ischemic stroke (AIS) [39,40,41]. Increased mitochondrial fragmentation and fission activity have been documented during the ischemic phase in both brain tissue and cardiomyocytes, as well as throughout I/R injury [42,43,44,45]. The neuroprotective effect of autophagy is attributed to its capacity to mitigate the accumulation of toxic proteins and damaged mitochondria, thereby preserving neuronal viability and function [33, 46]. The mechanism of ischemic injury in patients with acute stroke is illustrated in Fig. 2.

Fig. 2
figure 2

The mechanism of ischemic injury in acute stroke. In the context of acute ischemic stroke (AIS), the obstruction of cerebral blood flow results in damage to cerebral vascular endothelial cells and increased permeability of the blood–brain barrier (BBB). This compromise of the BBB leads to vasogenic cerebral edema, elevated intracranial pressure, and an imbalance in the neuronal microenvironment. Within the framework of AIS, injured neurons may release damage-associated molecular patterns (DAMPs), which subsequently stimulate the production of pro-inflammatory cytokines, including IL-8 and tumor necrosis factor-alpha (TNF-α). The release of cytokines exacerbates cerebral injury by promoting inflammatory responses and neuronal apoptosis. I/R therapy remains a crucial strategy in the management of AIS, as it alleviates both ischemic and reperfusion injuries, thereby protecting cerebral tissue and facilitating neurological recovery. (Created using Adobe Illustrator)

The association between mitochondrial injury and the restoration of blood flow represents a critical pathophysiological mechanism following I/R events [47]. Mitochondria that are functionally impaired, along with ROS generated through oxidative stress within these organelles, serve as substrates that can trigger intracellular autophagy [48]. This process facilitates the recovery or degradation of proteins and damaged organelles across various diseases [49]. Mitophagy, a specialized form of autophagy, specifically targets and degrades damaged mitochondria, enabling the recycling of their components [28]. Mitophagy plays a critical role in cellular homeostasis by selectively eliminating and degrading damaged or superfluous mitochondria, thereby preventing the accumulation of mitochondrial DNA mutations and facilitating the reprogramming of cellular metabolism [47]. The PINK1-PRKN/Parkin axis is considered the principal regulator of the PINK1-Parkin-mediated pathway, one of the two mitophagic autophagy pathways discussed above. This pathway initiates selective autophagy by marking damaged mitochondria with ubiquitin chains [50, 51]. In this process, PINK1 is hypothesized to function as a sensor for mitochondrial damage, Parkin as a signal amplifier, and the ubiquitinated chain as a crucial effector for signal transmission [37]. Nevertheless, the ubiquitinated chain does not directly interact with the free autophagic membrane or the associated ATG8 family proteins, indicating that ubiquitinated entities must be anchored to the autophagic membrane via specific molecular pathways [52]. Autophagy aptamers are characterized as proteins possessing mitochondrial ubiquitin-binding domains (UBDs), which facilitate the recognition of ubiquitin tags, as well as LC3 interaction regions (LIRs) that engage with ATG8 family proteins. These proteins include sequestosome 1 (P62/SQSTM1) [24], neighbor of BRCA1 gene 1 (NBR1) [53], nuclear dot protein 52 (NDP52/CALCOCO2), TAX1BP1 [54], and optineurin (OPTN) [51]. As receptors, they function to recognize ubiquitin chains on the mitochondrial surface and also bind to LC3B on phagocytic cell membranes [50]. Conversely, PINK1 may facilitate mitophagy through mechanisms independent of Parkin. For instance, the mitochondrial E3 ubiquitin ligase (MUL1), also known as MITA, can be activated via phosphorylation by PINK1, thereby contributing to the process of mitophagy [55]. Furthermore, the autophagy receptors involved in mitophagy include BNIP3, NIX, FUNDC1, MCL-1, cardiolipin (CL), among others [56]. PINK1 is pivotal in mitophagy, facilitating the removal of damaged mitochondria through the activation of Parkin as well as via Parkin-independent pathways. The process of mitophagy is also associated with the fusion of nascent mitochondria. Optic atrophy 1 (Opa1) is an inner mitochondrial membrane protein crucial for preserving mitochondrial structure and function, and it plays a significant role in regulating mitochondrial fusion and fission [42]. Research indicates that Opa1 undergoes apoptosis-associated modifications, shifting the equilibrium of mitochondrial dynamics toward fission by suppressing fusion [57]. While I/R injury primarily results in mitochondrial dysfunction, causing disruptions in oxidative stress regulation, calcium homeostasis, and apoptosis, targeting mitophagy-related pathways with specific molecules may offer therapeutic benefits for certain patients experiencing ischemic stroke. Certain regulators of mitophagy have demonstrated significant promise in clinical applications, particularly during extended recovery periods, where mitophagy may offer critical neuroprotective benefits and result in improved outcomes [58].

To comprehensively harness the potential of mitophagy in clinical treatment, it is imperative to further investigate and identify therapeutic targets capable of modulating the mitophagy pathway, alongside the development of corresponding pharmacological interventions. By conducting an in-depth analysis of the mechanisms through which autophagy protects cells, we can elucidate the specific pathways involved in mitigating I/R injury. This understanding will establish a theoretical foundation for the development of targeted intervention strategies. Such insights will enhance our ability to artificially sustain cell survival while minimizing the risk of inducing excessive cellular activation. Consequently, this research will offer scientific guidance for the development of therapeutic approaches that effectively harness the protective benefits of autophagy.

The adverse function of autophagy in I/R injury

Autophagy has been identified as a crucial cellular survival mechanism, facilitating the degradation and recycling of damaged organelles and proteins into nutrients that support cell viability. By regulating the autophagic pathway, cells are effectively maintained during states of homeostasis, stress, and infection. Consequently, autophagy was initially perceived primarily as a self-protective strategy enabling cells to mitigate damage induced by external stimuli [59, 60]. Nevertheless, although autophagy serves as a survival mechanism, its dysregulation may initiate a cascade of events culminating in excessive autophagy and potentially resulting in cell death [33]. The activation process of autophagy is intricate and modulated by environmental factors, with interactions at multiple levels exerting a significant influence on the ultimate outcome [61]. Thus, the protective function of autophagy in I/R injury is not unequivocal [28, 30], and disturbances in intracellular homeostasis beyond a certain threshold of time or magnitude can lead to deleterious effects [62,63,64]. For instance, in the management of ischemic stroke, while reperfusion strategies such as thrombolysis and thrombectomy are crucial for re-establishing blood flow and enhancing patient outcomes, they may also result in reperfusion injury, which can cause mitochondrial DNA damage and disrupt calcium homeostasis within the cytoplasm and mitochondria [33].

Furthermore, the interplay between autophagy and cell death serves as a critical determinant of cellular fate in I/R injury [10, 11, 65]. The role of autophagy in cell death can be categorized into autophagy-dependent cell death (ADCD or ACD) and autophagy-mediated cell death (AMCD) [10]. The two forms of autophagy associated with cell death are not entirely independent and may coexist within the cell. In certain instances, these modes can intertwine during the process of cell death [66]. ADCD typically occurs when autophagy is excessively activated, and the lysosomal degradation capacity is inadequate to process the substantial number of autophagosomes, which may include endoplasmic reticulum phagocytosis, mitophagy, and self-mutilation. This form of cell death is characterized by the accumulation of autophagosomes, resulting in disruptions to the intracellular environment and damage to organelles. For instance, during reperfusion, the disruption of autophagic flux can result in the accumulation of autophagosomes, thereby creating a toxic intracellular environment that exacerbates cellular damage and may ultimately lead to cell death [7, 10, 11, 20]. This disruption may arise from an imbalance in lysosomal degradation capacity or from the inhibition of autophagosome–lysosome fusion [22]. Lysosomes play a crucial role as degradative organelles within cells, and their proper functioning is essential for maintaining intracellular homeostasis [7, 10, 11]. During I/R injury, the lysosomal degradation capacity may become compromised owing to various factors, including diminished lysosomal enzyme activity and inadequate energy supply resulting from ischemia, both of which impair the enzymatic degradation of substrates [26]. Furthermore, I/R injury may disrupt the intracellular acidic environment, leading to lysosomal acidification disorders that adversely affect lysosomal enzyme activity [33]. The fusion of autophagosomes with lysosomes represents a pivotal stage in the degradation of autophagic substrates [7]. However, during I/R injury, this fusion process may be impeded, leading to the inefficient breakdown of autophagic substrates [26]. AMCD is a distinct form of cell death that is entirely reliant on the autophagic process.

Various forms of cell death include autophagy, necrosis, pyroptosis, apoptosis, and ferroptosis [67]. Autophagy can interact with these cell death processes in complex ways, depending on the specific cellular environment and signaling pathways involved [10]. Biochemically, autophagy is marked by increased expression of autophagy-related proteins such as LC3-II and Beclin-1. In contrast, necrosis is a passive and uncontrolled process triggered by external factors such as physical or chemical damage, leading to cell swelling, membrane rupture, and release of intracellular contents, with biochemical markers including lactate dehydrogenase (LDH) release and increased levels of ROS production [68]. Morphologically, necrotic cells are characterized by organelle swelling and the loss of plasma membrane integrity, whereas apoptotic cells display contraction, nuclear condensation, and DNA fragmentation [69]. Apoptosis is a programmed cell death process initiated by internal or external signals, leading to caspase activation and subsequent cell division and dissolution [70]. Pyroptosis is integral to the immune response, facilitating the elimination of pathogen-infected cells and inducing inflammation to recruit immune cells [71]. In contrast, ferroptosis is distinct from pyroptosis as it is an iron-dependent form of cell death marked by uncontrolled lipid peroxidation within diverse and adaptable mechanisms [9, 63, 72]. Specific autophagic processes, including ferritinophagy, lipophagy, and clockophagy, contribute to the initiation or execution of iron-induced cell death by selectively degrading proteins or organelles that protect against damage [9]. Additionally, other forms of selective autophagy, such as reticulophagy and lysophagy, bolster cellular defenses against damage caused by iron phagophores [7]. For instance, within cardiac tissue, Mammalian sterile 20-like kinase 1 (Mst1), a component of the Hippo signaling pathway [73], exerts a protective influence against cardiac I/R injury. This is achieved through the activation of the Kelch-like ECH-associated protein 1 (Keap1)/nuclear factor erythroid 2-related factor (Nrf2) axis and the suppression of ROS production, indicating a significant role for Mst1 in the transitional management of cardiac I/R injury during heart transplantation [6]. Studies have demonstrated that smoking can exacerbate autophagy through various mechanisms, contributing to cellular aging and tissue damage: free radicals and reactive oxygen species generated by smoking can harm cellular lipids, proteins, and DNA, resulting in oxidative stress; carcinogens present in smoke can damage cellular DNA; and smoking also induces mitochondrial damage and inflammation, which can further impair autophagy [74]. Research has indicated that factors such as stress [75] and environmental pollution [76] may trigger excessive autophagy in the body, potentially leading to depression under chronic stress conditions and severe outcomes like miscarriage in pregnant females. Understanding the mechanism of autophagy in the context of I/R injury is of paramount importance. It is essential to investigate the potential of autophagy as a therapeutic target, ensuring that its activation during treatment aims to preserve tissues and organs rather than exacerbate cellular damage. This understanding is critical for accurately mitigating and regulating the detrimental effects of autophagy in clinical settings, such as organ transplantation.

The dual action mechanisms of autophagy

In the preceding section, we discussed the enigmatic dual function of autophagy in I/R injury, a process governed by a variety of molecular mechanisms that modulate signaling pathways and influence whether autophagy facilitates cellular survival or leads to cellular destruction following I/R injury [10]. As previously noted, the genes implicated in the regulation of the fundamental processes of this autophagy pathway, collectively referred to as ATGs, encode proteins that are crucial for the formation and maturation of autophagosomes [60, 61]. Previous research has identified that ATGs are integral to processes such as protein secretion, pathogen degradation, and the maintenance of genome stability [16]. The regulation of these ATGs is mediated through both genetic and epigenetic mechanisms, which modulate the intensity and duration of autophagic responses, thereby influencing their protective or harmful outcomes [61].

Furthermore, the extent of autophagy activation is intricately linked to its spatial and temporal dynamics [62]. The thioredoxin-interacting protein (TXNIP)/regulated in development and DNA damage responses 1 (Redd1) complex has been identified as a promoter of autophagosome formation during myocardial I/R, indicating an enhancement in autophagy activation [64]. While TXNIP is known to inhibit autophagosome clearance through the elevation of ROS levels, the formation of autophagosomes induced by TXNIP is not mediated by ROS. This conclusion is supported by evidence showing that the use of ROS scavengers does not impede the increased autophagosome formation observed in hearts overexpressing TXNIP [48, 64]. Ultimately, TXNIP directly interacts with and stabilizes the autophagy regulator Redd1, resulting in mammalian target of rapamycin (mTOR) inhibition and the activation of autophagy. This indicates that increased TXNIP/Redd1 expression represents a novel signaling pathway that exacerbates I/R injury by promoting excessive autophagy during reperfusion [64]. Furthermore, autophagy exhibits dual functions in tumorigenesis, serving as a tumor-suppressing mechanism in the early stages and a cancer-promoting factor in the later stages [77]. The dual mechanisms through which autophagy influences tumor development is illustrated in Fig. 3.

Fig. 3
figure 3

The dual role of autophagy in hepatic IRI. Autophagy plays a dual role in HIRI: moderate autophagy facilitates the clearance of damaged organelles and mitigates oxidative stress, whereas excessive autophagy can exacerbate cellular damage, leading to adverse outcomes. During the ischemic phase, protective autophagy is activated via AMPK stimulation owing to reduced ATP and oxygen levels, which inhibits mTOR, promotes ULK1 activation, and initiates the formation of protective autophagosomes. In the reperfusion phase, increased ROS levels activate BNIP3, resulting in the dissociation of the Beclin-1/Bcl-2 complex and the release of Beclin-1, thereby promoting autophagosome formation and contributing to the clearance of damaged organelles and proteins. However, overactivation of autophagy can lead to the excessive degradation of organelles and proteins, potentially forming harmful autophagosomes and exacerbating cellular damage. Therefore, maintaining autophagic homeostasis is crucial for alleviating hepatic ischemia–reperfusion injury. (Created using Adobe Illustrator)

Posttranslational modifications (PTMs) are crucial in the precise regulation of autophagy regulators’ functions [46, 78]. Building on prior research, the mechanism by which the primary amino acid sequence of proteins dictates the material properties of biomolecular condensates is well established [79]. Consequently, PTMs in proteins, including phosphorylation and ubiquitination, function as molecular switches to precisely regulate the dynamics of biomolecular condensates. Ubiquitination, a prevalent and reversible PTM, influences biomolecular assemblies via two primary mechanisms [78, 80]. Firstly, it modifies the molecular structure of protein constituents, thereby altering the physicochemical properties of the biomolecular assemblies. Secondly, it facilitates valence by engaging with binding partners within molecular networks that contain UBDs or ubiquitin-associated domains (UBAs) [80,81,82]. Furthermore, the accumulation of ubiquitin in individuals with neurodegenerative disorders, including amyotrophic lateral sclerosis (ALS), may be attributed to disruptions in autophagic activity and the homeostasis of stress granules [83]. Consequently, ubiquitination is crucial in regulating the dynamics of biomolecular aggregates associated with these diseases [78]. Drawing from these preclinical studies [78, 80,81,82], we posit that targeting ubiquitinated biomolecular aggregates holds significant promise in combating diseases such as ALS. PTMs, including phosphorylation and ubiquitination, function as molecular rheostats that dynamically modulate and regulate protein activity, stability, localization, and interactions among proteins such as ULK1, Beclin-1, and PI3K complexes. These processes are crucial in various biological functions, including cellular metabolism, growth, differentiation, and apoptosis [22, 84].

Beclin-1 participates in numerous biological processes, with its most extensively characterized function being its involvement in autophagy. It interacts with and modulates the activity of the PI3KC3/Vps34 lipid kinase, which generates PI3P, a critical regulator in the initiation of autophagy and intracellular membrane trafficking [84, 85]. These interactions can either augment or suppress autophagy, contingent upon the cellular context and the specific signaling pathways activated during I/R injury [22, 62]. For instance, the phosphorylation of ULK1 by AMPK facilitates the initiation of autophagy under energy-depleted conditions [22]. Research indicates that, in intestinal diseases, the expression level of Beclin-1 is frequently downregulated, potentially resulting in a reduction of autophagic activity [60]. Moreover, cyclic GMP-AMP synthetase (cGAS) functions as a DNA-sensing receptor and exhibits increased expression in both human and mouse models of colitis. Research indicates that a deficiency in cGAS can exacerbate colitis and decrease the levels of autophagic proteins, such as Beclin-1 and LC3-II. However, administration of the autophagy activator rapamycin has been shown to significantly alleviate the severity of colitis in cGAS knockout mice [86]. The data indicate that the ubiquitination of Beclin-1 may influence its interaction with anti-apoptotic proteins in the context of human inflammatory bowel disease (IBD) and mouse models of colitis, thereby modulating the equilibrium between autophagy and apoptosis to preserve intestinal epithelial homeostasis. Autophagy induced by ischemia is associated with the activation of AMPK and is suppressed by a dominant negative form of AMPK. However, during reperfusion, autophagy is characterized by an upregulation of Beclin-1 without concurrent AMPK activation [62]. Research indicates that ischemia activates autophagy via AMPK-dependent pathways during cardiac I/R injury, whereas ischemia/reperfusion itself induces autophagy through mechanisms dependent on Beclin-1 but independent of AMPK [21]. Specifically, during myocardial ischemia, myocardial cells experience significant stress due to energy deprivation and ATP depletion, rendering mitochondrial function a critical determinant [87]. Under ischemic and hypoxic conditions, anaerobic glycolysis predominates as the primary metabolic pathway, leading to the accumulation of lactate and hydrogen ions, thereby inducing intracellular acidosis [7]. The resultant decrease in pH and depletion of ATP activate the Na+/H+ ion exchanger and the Na+/HCO 3 transporter, while inhibiting the Na+/K+-ATPase, culminating in a substantial intracellular accumulation of sodium ions [31]. The accumulation of sodium ions and alterations in the Na+/Ca2+ exchanger on the muscle membrane contribute to elevated intracellular calcium levels and mitochondrial swelling [8]. Autophagy is activated as a response to I/R injury, oxidative stress, and energy depletion [8, 48]. Upon the restoration of blood flow and reoxygenation of the respiratory chain, ROS production is intensified, which triggers excessive autophagy and may ultimately result in cell death [48].

Autophagy has been demonstrated to exhibit a dual role in the pathological process of I/R injury [30]. Elucidating the mechanisms by which these molecular regulators operate is essential for comprehending the modulation of autophagy in the context of I/R injury [11, 28]. The biological milieu plays a pivotal role in the execution of autophagy, with its requirements varying across different stages of the autophagic process. Modulating autophagic pathways to alter the mode of cell death post-clinical intervention—transitioning from lethal to protective in the context of cardiovascular disease, or from protective to lethal in cancer therapy—could substantially influence therapeutic outcomes [88, 89]. For instance, in tumor cells, autophagy plays a crucial role in mitigating cytotoxicity by facilitating the removal of deleterious proteins and superfluous or damaged organelles, thereby inhibiting the progression of cellular malignancy. In tumor cells, the reduction in autophagic activity facilitates cellular evasion of apoptosis. Further research is required to substantiate the dual role of autophagy in tumors, which appears to be contingent upon the cellular microenvironment and the level of autophagic activity at specific temporal points [8, 48]. Moreover, the expression patterns of key autophagic molecules across various tumor types and their prognostic implications exhibit variability. To optimize therapeutic outcomes, it is essential to select suitable autophagy inhibitors or agonists on the basis of the specific autophagic and genetic characteristics of the tumor, thereby formulating a targeted and personalized treatment strategy. By focusing on these molecular regulators and their associated pathways, we can potentially augment the beneficial effects of autophagy while minimizing its detrimental impacts. This approach necessitates a comprehensive understanding of the intricate interactions between autophagy and other cellular processes, as well as the development of precise pharmacological tools to modulate these interactions. The primary challenge lies in effectively harnessing these molecular mechanisms to direct autophagy toward cellular rescue rather than cellular destruction.

The regulation of autophagy in I/R Injury

The regulation of autophagy presents a promising novel strategy for addressing I/R injury; however, its underlying mechanisms are notably complex [28]. The primary molecular regulators of autophagy include mTORC1, AMPK, p53, and endoplasmic reticulum stress (ERS) [7]. Notably, mTOR and Beclin-1, as molecules associated with autophagy, are pivotal during various stages of myocardial ischemia–reperfusion injury (MIRI) [84]. During the ischemic phase, the mTOR operates via the AMPK/mTOR and PI3K/AKT/mTOR signaling pathways [62]. In contrast, the expression of Beclin-1 is upregulated during the reperfusion phase [7]. The regulation of autophagy can be achieved through various strategies, including pharmacological interventions, genetic modifications, and alterations of environmental factors [47, 72, 90].

Regarding pharmacological interventions, the administration of serine/threonine kinase inhibitors, calcium channel inhibitors, and highly selective sodium-glucose cotransporter 2 (SGLT2) inhibitors has been demonstrated to be associated with the self-regulation of autophagy, yielding positive outcomes in clinical treatment [21]. The PI3K/TOR signaling pathway is recognized as a negative regulator of autophagy in mammalian cells. Furthermore, depletion of cellular ATP significantly inhibits mTOR activity without impacting the activation of PI3K or altering intracellular amino acid concentrations [91]. As previously discussed, rapamycin serves as a potent and specific inhibitor of the mTOR pathway, demonstrating its ability to enhance autophagy and exhibiting therapeutic potential in preclinical models of I/R injury [92]. Empagliflozin has been shown to mitigate cardiac microvascular I/R damage through the activation of the AMPKα1/ULK1/FUNDC1/mitochondrial autophagy pathway [21]. In the context of colorectal cancer, excessive activation of the mTOR pathway impedes the initiation of autophagy, thereby facilitating tumor cell proliferation and survival [60].

While genetic manipulation offers precise control over the autophagy process, its application in clinical settings remains constrained. For instance, the inhibition of mTORC1 by circular RNA (circ-FoxO3) to enhance autophagy, or the knockout or overexpression of the ATG gene, exemplifies these limitations [59, 61]. Research indicates that the Sirt1/FoxO3α pathway provides a protective effect by modulating autophagy in hepatic ischemia–reperfusion injury (HIRI), a mechanism that can be disrupted by the specific Sirt1 inhibitor EX-527 [93]. At the molecular level, circular RNAs engage with autophagy-related microRNAs (miRNAs) [94, 95] and proteins [96] to modulate various pathological processes. The interplay between microRNAs (miRNAs) and autophagy is pivotal in the context of neurodegenerative diseases. Both autophagy and miRNAs exhibit dual roles in these diseases [97]. miRNAs are integral to the regulation of autophagy-related genes and signaling pathways, influencing the autophagic process; aberrant miRNA expression can result in autophagic dysfunction, thereby exacerbating the progression of neurodegenerative disorders [98]. Conversely, targeting specific miRNAs offers a therapeutic approach to modulate autophagy levels, potentially mitigating symptoms of neurodegenerative conditions. For instance, inhibiting miR-140, which enhances PINK1-mediated mitophagy, has been shown to alleviate symptoms of Alzheimer’s disease [99]. In the context of spinal cord ischemia–reperfusion injury (SCIRI), noncoding RNAs (ncRNAs) have the capacity to regulate apoptosis, inflammation, autophagy, and oxidative stress, thereby mitigating the effects of SCIRI [100]. The inhibition of the inhibitor Keap1, which regulates the Nrf2—an oxidative stress sensor and pivotal transcription factor for cellular protection against oxidative damage—results in the nuclear accumulation of Nrf2. This accumulation subsequently activates the transcription of genes responsible for encoding a range of cytoprotective, antioxidant, and anti-inflammatory proteins, thereby mitigating oxidative stress damage [6]. For instance, the Mst1 gene has been the subject of extensive research in the context of various reperfusion injuries associated with organ transplantation, including that of the heart [101]. As a pivotal mediator of oxidative stress, this serine/threonine kinase is intricately associated with mitochondrial function and autophagic processes [73]. Exposure to CoCl2 has been shown to upregulate Mst1 expression and activate the Keap1/Nrf2 signaling pathway, while exacerbating cellular oxidative damage through Mst1 gene ablation and inhibition of the Keap1/Nrf2 pathway [6]. Furthermore, apoptosis initiated by extrinsic pathways is mediated by transmembrane death receptors, which belong to the tumor necrosis factor receptor (TNFR) family and possess “death domains” [7]. Through the death domain, specific ligands and associated death receptors, such as the apoptosis-stimulating fragment ligand (FasL)/Fas receptor (FasR), TNF-α/TNFR1, TNF-related apoptosis-inducing ligand (TRAIL)/death receptor (DR), and TRAIL/DR5, mediate the transmission of apoptotic signals from the cell surface to intracellular pathways [102]. Additionally, the p53 gene serves as a crucial tumor suppressor, with the p53 protein in the cytoplasm capable of inhibiting the mTOR activity by suppressing AMPK activity [103]. In colorectal cancer, mutations and loss of function in the p53 gene are significantly associated with dysregulated autophagy and tumor progression [60].

Environmental factors are pivotal in investigating the regulatory mechanisms of autophagic activity, as they modulate various signaling pathways and molecular processes [80]. For instance, low-dose hydrogen sulfide (H2S) has been shown to mitigate the neuronal damage associated with cerebral ischemia–reperfusion injury (CIRI) [104]. The ubiquitination pathway, along with the PTEN-activated PINK1-Parkin pathway, represents typical regulatory mechanisms operative during mitosis [33, 84]. Furthermore, autophagy fulfills various physiological functions, including the maintenance of cellular homeostasis, promotion of cell survival, regulation of the cell cycle, modulation of oxidative stress, facilitation of muscle regeneration, preservation of the quiescent state of stem cells, and promotion of muscle cell differentiation [105]. In eukaryotic cells, the ubiquitin–proteasome system (UPS) and autophagy constitute two principal cellular degradation pathways that are essential for the clearance of misfolded or unfolded proteins. These pathways are critical for maintaining cellular and tissue homeostasis, preventing alterations associated with aging, and mitigating a range of human diseases [82]. For instance, the inhibition of the UPS results in the compensatory activation of autophagy via multiple mechanisms. Conversely, the suppression of autophagy can either activate or impair the proteasomal pathway, contingent upon the specific cellular context and environmental conditions [51, 83]. Furthermore, components of either system may serve as proteolytic targets for the other [16]. Cells must precisely regulate the induction of autophagy in response to diverse stress conditions. Reversible ubiquitination of the core autophagy-inducing factor, specifically the ULK1 and PI3K complex subunits, has been identified as a universal mechanism for both initiating and terminating autophagy across various cellular contexts [13, 65]. The initiation of autophagy is orchestrated by the ULK1 serine/threonine kinase, which associates with FIP200, ATG13, and ATG101 to form functional complexes [20, 84]. In numerous cellular stress responses, the activation of ULK1 initiates the phosphorylation of downstream factors, subsequently inducing a cascade of autophagy [22]. A key effector of ULK1 is the PI3K-II complex, comprising the lipid kinase Vps34 and the regulatory proteins Beclin-1, Vps15, and ATG14 [22, 84]. ULK1 facilitates the activation of PI3K-III complexes and recruits them to sites of autophagosome formation, where the produced PI3P plays a critical role in the nucleation process of autophagosomes [106]. ATG9, the sole transmembrane protein integral to the core autophagy machinery, is postulated to supply membrane resources essential for autophagosome formation [22]. Furthermore, ATG9 interacts with ATG2 and WIPI proteins (ATG18, a phosphatidylinositol 3-phosphate effector in yeast), playing a crucial role in the initial phases of autophagosome formation originating from the endoplasmic reticulum [65]. Additionally, the ubiquitination process contributes to the timely induction of autophagy through a mechanism of positive feedback [27]. The upregulation of Beclin-1 expression, a pivotal protein involved in the regulation of autophagosome formation and processing, is responsible for the activation of autophagy during reperfusion [84, 85]. In vitro studies have demonstrated that Beclin-1-mediated autophagy is modulated by the BCL-2 protein in cardiomyocytes under conditions of nutrient deprivation, such as amino acid and serum deficiencies [107]. In the human breast cancer cell line MCF-7, Beclin-1 protein expression is markedly reduced and, in certain instances, undetectable [108]. Stable transfection of the Beclin-1 gene has been shown to significantly enhance cellular autophagic activity, thereby reducing carcinogenic potential. Furthermore, ROS may induce Beclin-1-mediated autophagy during reperfusion [84]. In this context, elevated ROS levels serve not only as indicators of an energy crisis but also as critical promoters of autophagy. The overexpression of Beclin-1 resulting from reperfusion is associated with increased oxidative stress [85]. In addition to modulating the expression of Beclin-1, ROS also alter the oxidation state and activity of autophagy-related proteins, thereby facilitating the LC3, which subsequently triggers autophagy [85]. Studies have demonstrated that Beclin-1 inhibits tumorigenesis by inducing autophagic cell death in tumor cells. However, the downregulation of Beclin-1 gene expression markedly diminishes the autophagic response, shielding tumor cells from autophagic cell death and consequently promoting their continued proliferation [109]. Receptor-mediated mitophagy, encompassing the BNIP3 and FUNDC1 pathways, facilitates the recruitment of damaged mitochondria to autophagosomes through the binding to specific proteins [110]. In addition to the role of autophagy in I/R, mitophagy is critically involved as a mechanism of cell death. An analysis of the expanding significance of mitophagy in various other diseases is presented in Table 1.

Table 1 Effect of mitophagy in diseases

To investigate the regulatory mechanisms of mitophagy, it has been identified that, beyond the classical receptors BNIP3 and FUNDC1, a range of autophagy receptors, including BNIP3L/NIX, BCL2L13, AMBRA1, and FKBP8, are localized within the outer mitochondrial membrane (OMM). These receptors possess the capability to directly recruit LC3/GABARAP proteins, thereby facilitating mitochondrial degradation independently of ubiquitin signaling [50, 166, 167]. Conversely, the inner mitochondrial membrane (IMM) typically remains impermeable; however, during ischemic injury, characterized by simultaneous nutrient and oxygen deprivation, the mitochondrial permeability transition pore (mPTP) opens nonselectively. This opening results in the uncoupling of oxidative phosphorylation, ATP hydrolysis, and the accumulation of intramitochondrial inorganic phosphate [31]. CL plays a critical role in this regulatory process, as most CL-mediated mitochondrial autophagy receptors contain LC3 interaction regions (LIRs), which enable them to bind tightly to LC3/GABARAP proteins [168]. This interaction facilitates the induction of mitophagy under cellular stress conditions through the association of CL with LC3 [50]. Moreover, lipids such as ceramides may serve as signaling molecules or receptors for compromised mitochondria [169]. Hypoxia and nutrient deprivation are potent inducers of autophagy; under anoxic conditions, cells activate the autophagic pathway to degrade and recycle intracellular components to maintain energy homeostasis and ensure survival [170, 171]. Autophagy is significantly upregulated in response to nutrient deprivation, particularly in the context of energy depletion due to deficiencies in amino acids and glucose [12, 72]. This process constitutes a cellular strategy to acquire essential nutrients and support survival [170]. Additional regulatory mechanisms include the dysregulation of lysosomal clearance, the involvement of NLRP3 inflammasomes in mediating pyroptosis, the role of iron metabolism-related proteins in influencing ferroptosis, and the regulation of mitochondrial damage by BCL-2 family proteins [172,173,174]. In recent years, ERS has also garnered significant attention as a novel regulatory pathway of apoptosis [175]. ERS is implicated in a wide range of physiological and pathological processes, including protein folding, intracellular Ca2+ storage, oxidative stress, hypoxia, ischemia, and lipid metabolism disorders, and is intricately associated with myocardial IRI [29]. Although ERS is essential for cellular survival, its prolonged activation can lead to apoptosis [7]. The myocardial damage resulting from the accumulation of unfolded proteins during ERS can further exacerbate ERS, thereby altering the metabolic state of cardiomyocytes and causing more severe injury [176]. In the context of MIRI, ERS levels increase, and the attenuation of ERS has been demonstrated to alleviate the effects of MIRI [29, 87, 100]. However, it is crucial to recognize that not all ERS responses are harmful. For example, the ERS transcription factor ATF6 has been shown to provide cardiomyocyte protection against ischemia–reperfusion injury [7]. Collectively, these mechanisms contribute to the impaired clearance of damaged proteins and organelles within cells, leading to their intracellular accumulation. This accumulation subsequently triggers apoptosis, necrosis, and other forms of cell death, thereby exacerbating myocardial damage [20]. Stress granules, which are membraneless organelles located within the cytoplasm, are formed in response to a variety of environmental stressors, such as elevated temperatures, oxidative stress, and viral infections [177]. These granules consist of messenger ribonucleoprotein complexes (mRNPs), including stalled mRNA, RNA-binding proteins (RBPs), translation initiation factors, and various other proteins [78]. Recent studies have highlighted the critical role of ubiquitination in regulating the dynamics of stress granules, particularly concerning their assembly, disassembly, and degradation processes [81, 177, 178]. The regulation of stress granule dynamics is significantly influenced by PTMs, including phosphorylation and methylation [178]. Different stressors induce distinct patterns of ubiquitination within the stress granule proteome; for instance, heat shock results in substantial ubiquitination of stress granule components, whereas arsenite, a common inducer of stress granules, does not [81].

We propose the novel use of autophagy regulation as a therapeutic strategy to mitigate organ damage and systemic effects resulting from I/R injury. To accomplish this, it is imperative to explore the molecular mechanisms underlying the dual role of autophagy and to develop pharmacological agents that are both safe and efficacious, ensuring their timely and precise administration. The challenge lies in identifying the critical factors, timing, and extent of autophagy activation, as well as synchronizing these interventions with the dynamics of I/R injury. Consequently, it is imperative to rigorously evaluate the impact of these regulatory strategies in preclinical models to ascertain their efficacy and potential side effects. The exploration of autophagy’s therapeutic potential is currently progressing, with each step offering new insights into the intricate balance between its protective and detrimental properties. Here, we offer a summary of the factors that can influence or regulate the process of autophagy, as delineated in Table 2.

Table 2 Autophagy-related influencing factors

The effects on organ damage and the whole body

Phagocytosis has extensive and significant implications for I/R-induced clinical organ injury and its systemic effects [60]. As previously discussed, autophagy represents a distinct form of cell death that operates independently of other apoptotic pathways or excessive autophagic processes [10].

Cardiovascular disease represents a significant global public health challenge, emerging as a leading cause of morbidity and mortality worldwide [183]. The investigation of autophagy within the context of cardiovascular diseases (CVDs) encompasses intricate vascular pathological processes that result in the impairment of vascular architecture and cardiac functionality, thereby imposing a substantial burden on global health systems and economic resources [184]. These conditions encompass atherosclerosis, hypertension, MIRI, myocardial infarction, myocardial hypertrophy, heart failure, and dilated cardiomyopathy [185, 186]. Adverse consequences of autophagy in MIRI are illustrated in Fig. 4.

Fig. 4
figure 4

Deleterious effects of autophagy in MIRI. During ischemic conditions, cardiomyocytes shift to anaerobic metabolism, which leads to a reduction in ATP levels and an elevation in intracellular Na+ and Ca2+ concentrations. Upon reperfusion, there is an exacerbation of intracellular Ca2+ accumulation and ROS generation, culminating in MIRI. The principal apoptotic signaling pathways involved are the intrinsic pathway, mediated by mitochondrial mechanisms, and the extrinsic pathway, mediated by death receptors. During ischemia and the initial phase of reperfusion, cellular acidosis, disruption of homeostasis, and various other factors facilitate the enzymatic release of iron equivalents or iron ions. This process activates the Fenton reaction, resulting in elevated levels of ROS, ultimately inducing ferroptosis in cardiomyocytes. (Created using Adobe Illustrator)

Mortality rates associated with cardiovascular disease have decreased as a result of proactive preventive strategies and advancements in pharmacological treatments and medical technology [166]. Nonetheless, the overall burden of cardiovascular disease remains substantial, primarily owing to inadequate implementation of prevention guidelines, challenges in adhering to preventive measures, and the prevalent occurrence of risk factors for coronary heart disease, including lipid metabolism disorders, hypertension, and diabetes mellitus [107]. The issue of IRI in the context of myocardial infarction presents a significant challenge in cardiovascular medicine. The acute loss of myocardial tissue due to an ischemic event results in profound metabolic and ionic disturbances within the affected myocardium, culminating in cell death [187]. Even when blood flow is subsequently restored, the heart’s limited regenerative capacity poses a substantial obstacle to cell replacement [188]. The role of autophagy in cardiac function is complex and somewhat paradoxical. In conditions such as obesity and high-fat diet-induced cardiac dysfunction, autophagy plays a protective role by modulating cardiometabolism and safeguarding the heart against ischemia–reperfusion damage [30, 189, 190]. Conversely, cardiomyocytes are heavily reliant on oxygen for oxidative phosphorylation to sustain their function. Under hypoxic conditions, the reduction in mitochondrial ATP production leads to calcium overload, acidosis, and oxidative stress, all of which contribute to myocardial injury [191]. In the context of cardioprotective effects, pretreatment has been shown to enhance autophagic flux by activating AMPK and PI3K while inhibiting mTOR [192]. Additionally, it has been proposed that drug posttreatment (PPC), administered within minutes during the early stages of reperfusion, offers greater efficacy and flexibility compared with conditioning or ischemic conditions [193]. Several studies have indicated that fluctuations in oxygen levels, rather than sustained low oxygen levels, constitute the most detrimental factor in myocardial hypoxia–reperfusion injury. Therefore, it is imperative to regulate the reperfusion process in hypoxia–reperfusion injury to prevent significant myocardial damage caused by abrupt, extensive blood reperfusion [194].

Cerebral ischemic injury is a significant contributor to global morbidity and mortality, precipitating various central nervous system disorders, including AIS and chronic ischemic AD [111]. Research indicates that, during cerebral ischemia/reperfusion events, there is a reduction in ATP levels, which activates the intracellular energy sensor AMPK. This activation subsequently inhibits mTORC1, resulting in the dephosphorylation of ATG13 and ULK1. These molecular events facilitate the assembly of ULK1 complexes and expedite the initiation of autophagy [195, 196]. During the initial phase of reperfusion, autophagy plays a cytoprotective role by degrading damaged organelles and misfolded proteins, thereby releasing amino acids and nucleotides for recycling [197]. Conversely, prolonged activation of autophagy in the later stages of reperfusion may result in the excessive degradation of healthy organelles and proteins, culminating in autophagic cell death and secondary injury to histiocytes. This indicates that autophagy has a dual role in brain I/R injury [198].

HIRI represents a significant complication associated with hepatectomy and liver transplantation, profoundly affecting patient outcomes [199]. Research indicates that upregulated autophagy plays a crucial role in the restoration of liver function following I/R injury. From an energy metabolism perspective, autophagy contributes to the maintenance of metabolic homeostasis by facilitating the removal of damaged organelles and proteins, thereby recycling their constituent nutrients and supplying energy to cells [200]. During oxidative stress, autophagy plays a crucial role in mitigating cellular damage by eliminating deleterious substances, including ROS. This process helps to attenuate the detrimental effects of oxidative stress on cells. Furthermore, within the context of the inflammatory response, autophagy serves to inhibit the release of inflammatory cytokines, thereby reducing the inflammatory cascade and alleviating liver injury [27, 201]. Nonetheless, it is important to note that excessive autophagy can result in the degradation of normal organelles and proteins, which may impair cellular function and potentially exacerbate liver injury [202]. The regulatory mechanisms governing autophagy are intricate, and the interactions among various pathways render the role of autophagy in HIRI a subject of ongoing debate [201]. The AMPK/mTOR signaling pathway is unequivocally central to the regulation of autophagy and remains a prominent subject of contemporary research. Studies have demonstrated that the induction of autophagy correlates with reduced expression and activity of mTOR during HIRI. Furthermore, autophagy displays dual regulatory roles in the progression of HIRI, exerting a protective effect on cells during the early stages, while potentially contributing to adverse outcomes in prolonged ischemic conditions [202]. As previously discussed, the removal of damaged mitochondria is dependent on the selective autophagic process mediated by the PINK1/Parkin pathway, known as mitophagy [203]. The upregulation of PINK1 protein via this pathway has been demonstrated to trigger mitophagy, subsequently inhibiting the NLRP3 inflammatory pathway and mitigating HIRI [204].

Autophagy is an essential mechanism for the kidneys to sustain normal physiological functions, including the preservation of podocyte morphology and functionality [13, 61]. The targeted deletion of ATG5 or ATG7 in renal epithelial cells has been demonstrated to induce CKD in murine models, characterized by podocyte and tubular dysfunction, glomerular and tubulointerstitial damage, and progressive organ failure [205]. Moreover, the specific deletion of ATG5 in mouse podocytes facilitates the onset of age-dependent glomerulopathy, evidenced by the accumulation of oxidative and ubiquitinated proteins, heightened endoplasmic reticulum stress, podocyte loss, and proteinuria [61]. These studies have substantiated the critical role of autophagy in mitigating age-related glomerular disease and the deterioration of renal function [61]. Beyond glomerular disease, the damage and apoptosis of tubular epithelial cells represent a significant characteristic of AKI, which has the potential to progress to CKD if the injury is recurrent or inadequately repaired [206]. Cisplatin-induced tubular apoptosis has been demonstrated to decrease when autophagy inhibitors, such as 3-methyladenosine or bafilomycin, are used, or when Beclin-1 expression is downregulated. This is particularly relevant for chemotherapeutic agents such as cisplatin, which induce AKI through the promotion of autophagy [207]. Nonetheless, certain studies have indicated that the enhancement of autophagy by cisplatin may, in contrast, aggravate renal injury and apoptosis [208]. Various injuries, including renal I/R, sepsis, and exposure to nephrotoxins, can result in nutrient depletion and oxidative stress, which subsequently trigger the activation of autophagy [13, 14, 61]. Renal fibrosis is a characteristic feature of CKD, with transforming growth factor-β1 (TGF-β1) playing a pivotal role in its progression. TGF-β1 not only facilitates the activation of fibroblasts but also significantly contributes to the development of renal fibrosis [209, 210]. In the context of chronic kidney diseases, including diabetic nephropathy, primary nephrotic syndrome, immunoglobulin A nephropathy, and doxorubicin-induced nephropathy, autophagy may be activated as an intrinsic protective mechanism within renal tubular epithelial cells and podocytes. The extent of autophagic disruption is associated with the severity of chronic kidney disease exacerbation [211,212,213].

Furthermore, autophagy is essential for preserving endothelial cell function and vascular integrity during pulmonary I/R injury [185]. Autophagy, a crucial cellular degradation and recycling mechanism, plays a vital role in preserving damaged mitochondria and preventing the release of cytotoxic substances, thereby sustaining mitochondrial function and cellular viability. The regulation of autophagy within endothelial cells influences the equilibrium between pro-survival and pro-apoptotic signaling pathways, potentially altering cellular fate under ischemic conditions [214]. Furthermore, autophagy inhibits apoptosis, maintains intracellular homeostasis, and safeguards blood vessels from damage by modulating angiogenesis [25]. For instance, P66shc facilitates the removal of damaged mitochondria through the promotion of mitophagy, thereby contributing to the maintenance of mitochondrial function and vascular integrity in endothelial cells [185]. Dysfunction of pulmonary endothelial cells (ECs) is a key characteristic of pulmonary I/R injury, leading to excessive fluid accumulation in the lungs (i.e., edema), reduced efficiency of gas exchange, and diminished lung elasticity [215]. The occurrence of I/R injury following lung transplantation is unavoidable and can result in the initial dysfunction of the transplanted organ. This condition contributes to heightened morbidity and mortality among postoperative patients and may also precipitate immune rejection, a critical determinant of postoperative mortality in recipients [216]. Dysfunction in autophagy can result in vascular injury and disease. Consequently, strategies aimed at modulating autophagy, including the application of autophagy inducers, inhibitors, or gene therapy, are anticipated to enhance vascular integrity and offer therapeutic benefits for vascular diseases [84, 92]. Moreover, interventions targeting autophagy-related signaling pathways, as well as the use of natural products and lifestyle modifications, present additional avenues for modulating autophagy to improve vascular health [217, 218].

The impact of autophagy on the immune system’s inflammatory response is significant: autophagy has the potential to attenuate inflammation by degrading pro-inflammatory cytokines and modulating antigen presentation to immune cells [27]. Nevertheless, an imbalance in this regulatory mechanism can lead to exacerbated inflammation, potentially initiating a cascade of events culminating in multi-organ dysfunction syndrome [12]. Nod-like receptors (NLRs), including NOD1 and NOD2 signaling pathways, play a crucial role in immune defense by inducing autophagy and suppressing inflammatory responses. Additionally, autophagy modulates inflammatory pathways in macrophages, such as the NF-κB pathway, the RIG-I/STING pathway, and the inflammasome pathway [60, 201]. Disruptions in autophagy can result in heightened inflammatory responses, including conditions such as IBD, systemic lupus erythematosus (SLE), and arthritis. For instance, a deficiency in ATG7 is associated with elevated levels of IL-1β and pyroptosis, while a deficiency in ATG5 increases vulnerability to Mycobacterium tuberculosis [46, 201]. Consequently, investigating the interplay between autophagy and macrophage function is crucial for elucidating the mechanisms underlying inflammatory responses and for devising novel therapeutic strategies for inflammatory diseases [27]. Comprehending the organ-specific and systemic functions of autophagy in I/R injury is essential for the advancement of targeted therapeutic strategies. It is imperative to sustain a delicate equilibrium to avert dysregulation in the role of autophagy within these processes. The dual roles of autophagy in the IRI process of different organs are listed in Table 3.

Table 3 The dual role of autophagy in ischemia–reperfusion injury in different organs

Future therapeutic approaches must meticulously modulate autophagic activity to optimize its protective benefits while mitigating the potential risk of harm [12]. An important consideration is that autophagy inhibitors impact various stages of the autophagic process, resulting in distinct therapeutic outcomes. During the initiation phase of autophagy, inhibitors such as 3-methyladenosine, wortmannin, and LY294002 can impede the onset of autophagy. This inhibition leads to a downregulation in the expression of autophagy-related proteins, specifically LC3-II and Beclin-1, consequently diminishing autophagic flux. During the fusion phase of autophagy, inhibitors such as pafimycin A1 and chloroquine impede the fusion of lysosomes with autophagosomes, thereby obstructing the degradation of autophagic contents. This results in a reduction of autophagic activity, yet concurrently leads to an upregulation in the expression of autophagy-related proteins and an enhancement in autophagic flux [199]. This therapeutic approach within precision medicine necessitates an in-depth and comprehensive understanding of the cellular context, as well as the interactions between autophagy and other cell death mechanisms. Such understanding is essential to accurately discern the intricate balance between autophagy and cellular rescue or destruction following I/R injury. This knowledge may offer novel insights for the development of innovative treatments for I/R injury.

Conclusions

The dual role of autophagy in ischemia–reperfusion (I/R) injury presents both a challenge and an opportunity in therapeutic strategies. As a double-edged sword, autophagy can either protect or harm cells, depending on the context and extent of activation. This duality demands a nuanced understanding of its mechanisms and regulatory pathways. During the ischemic phase, autophagy acts as a protective mechanism by clearing dysfunctional organelles and misfolded proteins, thus maintaining cellular integrity. This is particularly important in organs such as the brain and heart, where rapid response to damage is critical. Autophagy recycles cellular components to restore energy balance, thereby facilitating recovery upon reperfusion. Conversely, excessive autophagy during reperfusion can exacerbate tissue damage. Overactivation might lead to autophagic cell death or an imbalance in cellular homeostasis, as seen in cases where it contributes to mitochondrial fragmentation and ATP depletion. Understanding the thresholds of protective versus detrimental autophagy is crucial, especially in therapeutic contexts such as cancer treatment or organ transplants, where modulation of autophagy could shift outcomes significantly. The therapeutic potential lies in precisely regulating autophagy to harness its protective benefits while minimizing harmful effects. This requires a sophisticated approach, potentially involving the use of autophagy modulators at specific stages of the process. For instance, targeting the AMPK/mTOR pathway could effectively modulate autophagy in liver and kidney injuries, improving outcomes in organ transplantation and chronic disease management. The future of I/R injury treatment could be revolutionized by therapies that finely tune autophagic responses. Investigating molecular targets such as the PINK1/Parkin pathway in mitochondrial autophagy could provide insights into reducing oxidative stress and inflammation. Additionally, understanding the interplay between autophagy and other cell death pathways could lead to more effective and personalized therapeutic strategies.

In conclusion, autophagy’s dual role in I/R injury underscores the need for targeted therapeutic approaches that leverage its protective aspects while controlling its potential to cause harm. As research advances, the potential for autophagy-based therapies in improving outcomes for diseases involving I/R injury appears promising, provided that we achieve a deeper comprehension of its regulatory mechanisms and their clinical implications.

Availability of data and materials

Data sharing is not applicable to this article as no datasets were generated or analyzed during the current study. All information is derived from publicly available articles and datasets.

Abbreviations

Aβ:

β-Amyloid

ACD:

Autophagic cell death

AD:

Alzheimer’s disease

ADCD:

Autophagy-dependent cell death

AIS:

Acute ischemic stroke

AKI:

Acute kidney injury

ALI:

Acute lung injury

ALS:

Amyotrophic lateral sclerosis

AMBRA1:

Activating molecules of the autophagy regulator 1

AMCD:

Autophagy-mediated cell death

AMD:

Age-related macular degeneration

AMP:

Adenosine monophosphate

AMPK:

AMP-activated protein kinase

ATGs:

Autophagy-related genes

ATP:

Adenosine triphosphate

BAK:

BCL-2 antagonist/killer 1

BAX:

BCL-2-associated X

BBB:

Blood–brain barrier

BCL-2:

B-cell lymphoma 2

BIM:

BCL-2 interacting mediator of cell death

CIRI:

Cerebral ischemia–reperfusion injury

circ-FoxO3:

Circular RNA FoxO3

cGAS:

Cyclic GMP-AMP synthetase

CKD:

Chronic kidney disease

CL:

Cardiolipin

CMA:

Chaperone-mediated autophagy

COPD:

Chronic obstructive pulmonary disease

CVDs:

Cardiovascular diseases

DAMPs:

Damage-associated molecular patterns

DR:

Death receptor

Drp1:

Dynein-associated protein 1

ECs:

Endothelial cells

ERS:

Endoplasmic reticulum stress

FasL:

Fas ligand

FasR:

Fas receptor

GABARAP:

Gamma-aminobutyric acid receptor-associated protein

H2S:

Hydrogen sulfide

HF:

Heart failure

HIRI:

Hepatic ischemia–reperfusion injury

HSC:

Heat shock cognate

IBD:

Inflammatory bowel disease

IMM:

Inner mitochondrial membrane

I/R:

Ischemia–reperfusion

Keap1:

Kelch-like ECH-associated protein 1

LAMP:

Lysosome-associated membrane protein

LC3:

Microtubule-associated protein 1 light chain 3

LIRs:

LC3 interaction regions

LRRK2:

Leucine-rich repeat kinase 2

MAPK:

Mitogen-activated protein kinase

MIRI:

Myocardial ischemia–reperfusion injury

miRNA:

MicroRNA

MODS:

Multiple organ dysfunction syndrome

mPTP:

Mitochondrial permeability transition pore

mRNPs:

Messenger ribonucleoprotein complexes

Mst1:

Mammalian sterile 20-like kinase 1

mtDNA:

Mitochondrial DNA

mTOR:

Mammalian target of rapamycin

mTORC1:

Mammalian target of rapamycin complex 1

MUL1:

Mitochondrial E3 ubiquitin ligase

NAFLD:

Nonalcoholic fatty liver disease

NBR1:

Neighbor of BRCA1 gene 1

ncRNAs:

Non-coding RNAs

NDP52:

Nuclear dot protein 52

NLRP3:

Nucleotide-binding domain and leucine-rich repeat containing protein 3

NLRs:

Nod-like receptors

Nrf2:

Nuclear factor erythroid 2-related factor

OMM:

Outer mitochondrial membrane

Opa1:

Optic atrophy 1

PI3K:

Phosphatidylinositol 3-kinase

PI3P:

Phosphatidylinositol 3-phosphate

PIP3:

Phosphatidylinositol (3,4,5)-trisphosphate

PKB:

Protein kinase B

pTau:

Tau protein

PTMs:

Posttranslational modifications

RBPs:

RNA-binding proteins

RGCs:

Retinal ganglion cells

ROS:

Reactive oxygen species

SCIRI:

Spinal cord ischemia–reperfusion injury

SGLT2:

Sodium-glucose co-transporter 2

SIRS:

Systemic inflammatory response syndrome

SLE:

Systemic lupus erythematosus

SNARE:

Soluble N-ethylmaleimide-sensitive factor attachment protein receptors

SOD:

Superoxide dismutase

SQSTM1:

Sequestosome 1

TGF-β1:

Transforming growth factor-β1

TNF-α:

Tumor necrosis factor-alpha

TNFR:

Tumor necrosis factor receptor

TRAIL:

TNF-related apoptosis-inducing ligand

TXNIP:

Thioredoxin-interacting protein

UBAs:

Ubiquitin-associated domains

UBDs:

Ubiquitin binding domains

ULK:

Unc-51 like autophagy activating kinase

UPS:

Ubiquitin–proteasome system

Vps:

Vacuolar protein sorting

References

  1. Wu MY, Yiang GT, Liao WT, Tsai AP, Cheng YL, Cheng PW, et al. Current mechanistic concepts in ischemia and reperfusion injury. Cell Physiol Biochem. 2018;46(4):1650–67.

    Article  CAS  PubMed  Google Scholar 

  2. Eltzschig HK, Eckle T. Ischemia and reperfusion—from mechanism to translation. Nat Med. 2011;17(11):1391–401.

    Article  CAS  PubMed  Google Scholar 

  3. Newton K, Strasser A, Kayagaki N, Dixit VM. Cell death. Cell. 2024;187(2):235–56.

    Article  CAS  PubMed  Google Scholar 

  4. Zhou H, He L, Xu G, Chen L. Mitophagy in cardiovascular disease. Clin Chim Acta. 2020;507:210–8.

    Article  CAS  PubMed  Google Scholar 

  5. Su L, Zhang J, Gomez H, Kellum JA, Peng Z. Mitochondria ROS and mitophagy in acute kidney injury. Autophagy. 2023;19(2):401–14.

    Article  CAS  PubMed  Google Scholar 

  6. Fei Q, Liu J, Qiao L, Zhang M, Xia H, Lu D, et al. Mst1 attenuates myocardial ischemia/reperfusion injury following heterotopic heart transplantation in mice through regulating Keap1/Nrf2 axis. Biochem Biophys Res Commun. 2023;644:140–8.

    Article  CAS  PubMed  Google Scholar 

  7. He J, Liu D, Zhao L, Zhou D, Rong J, Zhang L, et al. Myocardial ischemia/reperfusion injury: mechanisms of injury and implications for management (Review). Exp Ther Med. 2022;23(6):430.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Youle RJ, Narendra DP. Mechanisms of mitophagy. Nat Rev Mol Cell Biol. 2011;12(1):9–14.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Chen X, Tsvetkov AS, Shen H-M, Isidoro C, Ktistakis NT, Linkermann A, et al. International consensus guidelines for the definition, detection, and interpretation of autophagy-dependent ferroptosis. Autophagy. 2024;20(6):1213–46.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Liu S, Yao S, Yang H, Liu S, Wang Y. Autophagy: regulator of cell death. Cell Death Dis. 2023. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41419-023-06154-8.

    Article  PubMed  PubMed Central  Google Scholar 

  11. Mizushima N, Komatsu M. Autophagy: renovation of cells and tissues. Cell. 2011;147(4):728–41.

    Article  CAS  PubMed  Google Scholar 

  12. Dugbartey GJ. Cellular and molecular mechanisms of cell damage and cell death in ischemia–reperfusion injury in organ transplantation. Mol Biol Rep. 2024;51(1):473.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Choi ME. Autophagy in kidney disease. Annu Rev Physiol. 2020;82(1):297–322.

    Article  CAS  PubMed  Google Scholar 

  14. Tang C, Livingston MJ, Liu Z, Dong Z. Autophagy in kidney homeostasis and disease. Nat Rev Nephrol. 2020;16(9):489–508.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Miceli C, Leri M, Stefani M, Bucciantini M. Autophagy-related proteins: Potential diagnostic and prognostic biomarkers of aging-related diseases. Ageing Res Rev. 2023;89: 101967.

    Article  CAS  PubMed  Google Scholar 

  16. Levine B, Kroemer G. Biological functions of autophagy genes: a disease perspective. Cell. 2019;176(1–2):11–42.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Jiang S, Sun J, Mohammadtursun N, Hu Z, Li Q, Zhao Z, et al. Dual role of autophagy/mitophagy in chronic obstructive pulmonary disease. Pulm Pharmacol Ther. 2019;56:116–25.

    Article  CAS  PubMed  Google Scholar 

  18. Tekirdag K, Cuervo AM. Chaperone-mediated autophagy and endosomal microautophagy: joint by a chaperone. J Biol Chem. 2018;293(15):5414–24.

    Article  CAS  PubMed  Google Scholar 

  19. Oku M, Sakai Y. Three distinct types of microautophagy based on membrane dynamics and molecular machineries. BioEssays. 2018;40(6): e1800008.

    Article  PubMed  Google Scholar 

  20. Liu Y, Li L, Wang Z, Zhang J, Zhou Z. Myocardial ischemia–reperfusion injury; molecular mechanisms and prevention. Microvas Res. 2023. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.mvr.2023.104565.

    Article  Google Scholar 

  21. Cai C, Guo Z, Chang X, Li Z, Wu F, He J, et al. Empagliflozin attenuates cardiac microvascular ischemia/reperfusion through activating the AMPKα1/ULK1/FUNDC1/mitophagy pathway. Redox Biol. 2022. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.redox.2022.102288.

    Article  PubMed  PubMed Central  Google Scholar 

  22. Li X, He S, Ma B. Autophagy and autophagy-related proteins in cancer. Mol Cancer. 2020;19(1):12.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Rizzuto R, Bernardi P, Pozzan T. Mitochondria as all-round players of the calcium game. J Physiol. 2000;529(1):37–47.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Lamark T, Svenning S, Johansen T. Regulation of selective autophagy: the p62/SQSTM1 paradigm. Essays Biochem. 2017;61(6):609–24.

    Article  PubMed  Google Scholar 

  25. Ren J, Zhang Y. Targeting autophagy in aging and aging-related cardiovascular diseases. Trends Pharmacol Sci. 2018;39(12):1064–76.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Shen Q, Shi Y, Liu J, Su H, Huang J, Zhang Y, et al. Acetylation of STX17 (syntaxin 17) controls autophagosome maturation. Autophagy. 2021;17(5):1157–69.

    Article  CAS  PubMed  Google Scholar 

  27. Wu MY, Lu JH. Autophagy and macrophage functions: inflammatory response and phagocytosis. Cells. 2019;9(1):70.

    Article  PubMed  PubMed Central  Google Scholar 

  28. Yang M, Linn BS, Zhang Y, Ren J. Mitophagy and mitochondrial integrity in cardiac ischemia–reperfusion injury. Biochim Biophys Acta Mol Basis Dis. 2019;1865(9):2293–302.

    Article  CAS  PubMed  Google Scholar 

  29. Sukumaran P, Nascimento Da Conceicao V, Sun Y, Ahamad N, Saraiva LR, Selvaraj S, et al. Calcium signaling regulates autophagy and apoptosis. Cells. 2021;10(8):2125.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Aghaei M, Motallebnezhad M, Ghorghanlu S, Jabbari A, Enayati A, Rajaei M, et al. Targeting autophagy in cardiac ischemia/reperfusion injury: a novel therapeutic strategy. J Cell Physiol. 2019;234(10):16768–78.

    Article  CAS  PubMed  Google Scholar 

  31. Anzell AR, Maizy R, Przyklenk K, Sanderson TH. Mitochondrial quality control and disease: insights into ischemia–reperfusion injury. Mol Neurobiol. 2018;55(3):2547–64.

    Article  CAS  PubMed  Google Scholar 

  32. Tang C, Cai J, Yin XM, Weinberg JM, Venkatachalam MA, Dong Z. Mitochondrial quality control in kidney injury and repair. Nat Rev Nephrol. 2021;17(5):299–318.

    Article  CAS  PubMed  Google Scholar 

  33. Shen L, Gan Q, Yang Y, Reis C, Zhang Z, Xu S, et al. Mitophagy in cerebral ischemia and ischemia/reperfusion injury. Front Aging Neurosci. 2021. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fnagi.2021.687246.

    Article  PubMed  PubMed Central  Google Scholar 

  34. Deepak K, Roy PK, Das CK, Mukherjee B, Mandal M. Mitophagy at the crossroads of cancer development: exploring the role of mitophagy in tumor progression and therapy resistance. Biochim Biophys Acta. 2024;1871(6): 119752.

    Article  CAS  Google Scholar 

  35. Adlimoghaddam A, Fayazbakhsh F, Mohammadi M, Babaei Z, Behrooz AB, Tabasi F, et al. Sex and region-specific disruption of autophagy and mitophagy in Alzheimer’s disease: linking cellular dysfunction to cognitive decline. bioRxiv. 2024;141(1):58593.

    Google Scholar 

  36. McCabe M, Boya P, Chen RH, Chu CT, Colombo MI, Delgui L, et al. Women in Autophagy: an initiative to promote gender parity in science. Nat Cell Biol. 2024;26(12):2009–12.

    Article  CAS  PubMed  Google Scholar 

  37. Wang S, Long H, Hou L, Feng B, Ma Z, Wu Y, et al. The mitophagy pathway and its implications in human diseases. Signal Transduct Target Ther. 2023;8(1):304.

    Article  PubMed  PubMed Central  Google Scholar 

  38. Debnath J, Gammoh N, Ryan KM. Autophagy and autophagy-related pathways in cancer. Nat Rev Mol Cell Biol. 2023;24(8):560–75.

    Article  CAS  PubMed  Google Scholar 

  39. Lees KR, Bluhmki E, von Kummer R, Brott TG, Toni D, Grotta JC, et al. Time to treatment with intravenous alteplase and outcome in stroke: an updated pooled analysis of ECASS, ATLANTIS, NINDS, and EPITHET trials. Lancet (London, England). 2010;375(9727):1695–703.

    Article  CAS  PubMed  Google Scholar 

  40. Berkhemer OA, Fransen PS, Beumer D, van den Berg LA, Lingsma HF, Yoo AJ, et al. A randomized trial of intraarterial treatment for acute ischemic stroke. N Engl J Med. 2015;372(1):11–20.

    Article  PubMed  Google Scholar 

  41. Jovin TG, Chamorro A, Cobo E, de Miquel MA, Molina CA, Rovira A, et al. Thrombectomy within 8 hours after symptom onset in ischemic stroke. N Engl J Med. 2015;372(24):2296–306.

    Article  CAS  PubMed  Google Scholar 

  42. Kumar R, Bukowski MJ, Wider JM, Reynolds CA, Calo L, Lepore B, et al. Mitochondrial dynamics following global cerebral ischemia. Mol Cell Neurosci. 2016;76:68–75.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Kim H, Scimia MC, Wilkinson D, Trelles RD, Wood MR, Bowtell D, et al. Fine-tuning of Drp1/Fis1 availability by AKAP121/Siah2 regulates mitochondrial adaptation to hypoxia. Mol Cell. 2011;44(4):532–44.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Sanderson TH, Raghunayakula S, Kumar R. Neuronal hypoxia disrupts mitochondrial fusion. Neuroscience. 2015;301:71–8.

    Article  CAS  PubMed  Google Scholar 

  45. Tang J, Hu Z, Tan J, Yang S, Zeng L. Parkin protects against oxygen-glucose deprivation/reperfusion insult by promoting Drp1 degradation. Oxid Med Cell Longev. 2016;2016:8474303.

    Article  PubMed  PubMed Central  Google Scholar 

  46. Figlia G, Willnow P, Teleman AA. Metabolites regulate cell signaling and growth via covalent modification of proteins. Dev Cell. 2020;54(2):156–70.

    Article  CAS  PubMed  Google Scholar 

  47. Cai Y, Yang E, Yao X, Zhang X, Wang Q, Wang Y, et al. FUNDC1-dependent mitophagy induced by tPA protects neurons against cerebral ischemia–reperfusion injury. Redox Biol. 2021;38:101792.

    Article  CAS  PubMed  Google Scholar 

  48. Li L, Tan J, Miao Y, Lei P, Zhang Q. ROS and autophagy: interactions and molecular regulatory mechanisms. Cell Mol Neurobiol. 2015;35(5):615–21.

    Article  PubMed  PubMed Central  Google Scholar 

  49. Vurusaner B, Gargiulo S, Testa G, Gamba P, Leonarduzzi G, Poli G, et al. The role of autophagy in survival response induced by 27-hydroxycholesterol in human promonocytic cells. Redox Biol. 2018;17:400–10.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Iriondo MN, Etxaniz A, Varela YR, Ballesteros U, Hervas JH, Montes LR, et al. LC3 subfamily in cardiolipin-mediated mitophagy: a comparison of the LC3A, LC3B and LC3C homologs. Autophagy. 2022;18(12):2985–3003.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Zha Z, Wang J, Wang X, Lu M, Guo Y. Involvement of PINK1/Parkin-mediated mitophagy in AGE-induced cardiomyocyte aging. Int J Cardiol. 2017;227:201–8.

    Article  PubMed  Google Scholar 

  52. Johansen T, Lamark T. Selective autophagy: ATG8 family proteins, LIR motifs and cargo receptors. J Mol Biol. 2020;432(1):80–103.

    Article  CAS  PubMed  Google Scholar 

  53. Kirkin V, Lamark T, Sou YS, Bjørkøy G, Nunn JL, Bruun JA, et al. A role for NBR1 in autophagosomal degradation of ubiquitinated substrates. Mol Cell. 2009;33(4):505–16.

    Article  CAS  PubMed  Google Scholar 

  54. Turco E, Savova A, Gere F, Ferrari L, Romanov J, Schuschnig M, et al. Reconstitution defines the roles of p62, NBR1 and TAX1BP1 in ubiquitin condensate formation and autophagy initiation. Nat Commun. 2021;12(1):5212.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Prudent J, Zunino R, Sugiura A, Mattie S, Shore GC, McBride HM. MAPL SUMOylation of drp1 stabilizes an ER/mitochondrial platform required for cell death. Mol Cell. 2015;59(6):941–55.

    Article  CAS  PubMed  Google Scholar 

  56. Villa E, Proïcs E, Rubio-Patiño C, Obba S, Zunino B, Bossowski JP, et al. Parkin-independent mitophagy controls chemotherapeutic response in cancer cells. Cell Rep. 2017;20(12):2846–59.

    Article  CAS  PubMed  Google Scholar 

  57. Tokuyama T, Yanagi S. Role of mitochondrial dynamics in heart diseases. Genes. 2023. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/genes14101876.

    Article  PubMed  PubMed Central  Google Scholar 

  58. Fang EF, Hou Y, Palikaras K, Adriaanse BA, Kerr JS, Yang B, et al. Mitophagy inhibits amyloid-β and tau pathology and reverses cognitive deficits in models of Alzheimer’s disease. Nat Neurosci. 2019;22(3):401–12.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Yang Z, Huang C, Wen X, Liu W, Huang X, Li Y, et al. Circular RNA circ-FoxO3 attenuates blood-brain barrier damage by inducing autophagy during ischemia/reperfusion. Mol Ther. 2022;30(3):1275–87.

    Article  CAS  PubMed  Google Scholar 

  60. Shi Y, Jiang B, Zhao J. Induction mechanisms of autophagy and endoplasmic reticulum stress in intestinal ischemia–reperfusion injury, inflammatory bowel disease, and colorectal cancer. Biomed Pharmacother. 2024. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.biopha.2023.115984.

    Article  PubMed  Google Scholar 

  61. Hartleben B, Godel M, Meyer-Schwesinger C, Liu S, Ulrich T, Kobler S, et al. Autophagy influences glomerular disease susceptibility and maintains podocyte homeostasis in aging mice. J Clin Invest. 2010;120(4):1084–96.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Matsui Y, Takagi H, Qu X, Abdellatif M, Sakoda H, Asano T, et al. Distinct roles of autophagy in the heart during ischemia and reperfusion. Circ Res. 2007;100(6):914–22.

    Article  CAS  PubMed  Google Scholar 

  63. Xu C, Liu Z, Xiao J. Ferroptosis: a double-edged sword in gastrointestinal disease. Int J Mol Sci. 2021;22(22):12403.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Tao L, Yan W, Gao E, Zhang L, Wang H, Liu Y, et al. TXNIP/Redd1 signalling and excessive autophagy: a novel mechanism of myocardial ischaemia/reperfusion injury in mice. Cardiovasc Res. 2020;116(3):645–57.

    Article  PubMed  Google Scholar 

  65. Levine B, Kroemer G. Autophagy in the pathogenesis of disease. Cell. 2008;132(1):27–42.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Yeo BK, Hong CJ, Chung KM, Woo H, Kim K, Jung S, et al. Valosin-containing protein is a key mediator between autophagic cell death and apoptosis in adult hippocampal neural stem cells following insulin withdrawal. Mol Brain. 2016. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s13041-016-0212-8.

    Article  PubMed  PubMed Central  Google Scholar 

  67. D’Arcy MS. Cell death: a review of the major forms of apoptosis, necrosis and autophagy. Cell Biol Int. 2019;43(6):582–92.

    Article  PubMed  Google Scholar 

  68. Ai Y, Meng Y, Yan B, Zhou Q, Wang X. The biochemical pathways of apoptotic, necroptotic, pyroptotic, and ferroptotic cell death. Mol Cell. 2024;84(1):170–9.

    Article  CAS  PubMed  Google Scholar 

  69. Chen T, Ren Q, Ma F. New insights into constitutive neutrophil death. Cell Death Discov. 2025;11(1):6.

    Article  PubMed  PubMed Central  Google Scholar 

  70. Ketelut-Carneiro N, Fitzgerald KA. Apoptosis, pyroptosis, and necroptosis—Oh my! The many ways a cell can die. J Mol Biol. 2022;434(4): 167378.

    Article  CAS  PubMed  Google Scholar 

  71. Lin L, Zhang MX, Zhang L, Zhang D, Li C, Li YL. Autophagy, pyroptosis, and ferroptosis: new regulatory mechanisms for atherosclerosis. Front Cell Dev Biol. 2021;9: 809955.

    Article  PubMed  Google Scholar 

  72. Yan H-f, Zou T, Tuo Q-Z, Xu S, Li H, Belaidi AA, et al. Ferroptosis: mechanisms and links with diseases. Signal Transd Targeted Therapy. 2021;6(1):430.

    Google Scholar 

  73. Maejima Y, Zablocki D, Nah J, Sadoshima J. The role of the Hippo pathway in autophagy in the heart. Cardiovasc Res. 2023;118(17):3320–30.

    Article  PubMed  Google Scholar 

  74. Akhtari M, Jalalvand M, Sadr M, Sharifi H. Autophagy in the cellular consequences of tobacco smoking: insights into senescence. J Biochem Mol Toxicol. 2024;38(12): e70065.

    Article  CAS  PubMed  Google Scholar 

  75. Xu K, Wang M, Wang H, Zhao S, Tu D, Gong X, et al. HMGB1/STAT3/p65 axis drives microglial activation and autophagy exert a crucial role in chronic stress-induced major depressive disorder. J Adv Res. 2024;59:79–96.

    Article  CAS  PubMed  Google Scholar 

  76. Wan S, Wang X, Chen W, Xu Z, Zhao J, Huang W, et al. Polystyrene nanoplastics activate autophagy and suppress trophoblast cell migration/invasion and migrasome formation to induce miscarriage. ACS Nano. 2024;18(4):3733–51.

    Article  CAS  PubMed  Google Scholar 

  77. Yun CW, Jeon J, Go G, Lee JH, Lee SH. The dual role of autophagy in cancer development and a therapeutic strategy for cancer by targeting autophagy. Int J Mol Sci. 2020. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/ijms22010179.

    Article  PubMed  PubMed Central  Google Scholar 

  78. Liang P, Zhang J, Wang B. Emerging roles of ubiquitination in biomolecular condensates. Cells. 2023. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/cells12182329.

    Article  PubMed  PubMed Central  Google Scholar 

  79. Li J, Zhang M, Ma W, Yang B, Lu H, Zhou F, et al. Post-translational modifications in liquid-liquid phase separation: a comprehensive review. Mol Biomed. 2022. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s43556-022-00075-2.

    Article  PubMed  PubMed Central  Google Scholar 

  80. Park JS, Ma H, Roh YS. Ubiquitin pathways regulate the pathogenesis of chronic liver disease. Biochem Pharmacol. 2021;193: 114764.

    Article  CAS  PubMed  Google Scholar 

  81. Maxwell BA, Gwon Y, Mishra A, Peng J, Nakamura H, Zhang K, et al. Ubiquitination is essential for recovery of cellular activities after heat shock. Science (New York, NY). 2021;372(6549):3593.

    Article  Google Scholar 

  82. Chen R-H, Chen Y-H, Huang T-Y. Ubiquitin-mediated regulation of autophagy. J Biomed Sci. 2019. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12929-019-0569-y.

    Article  PubMed  PubMed Central  Google Scholar 

  83. Ji CH, Kwon YT. Crosstalk and interplay between the ubiquitin-proteasome system and autophagy. Mol Cells. 2017;40(7):441–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  84. Hu YJ, Zhong JT, Gong L, Zhang SC, Zhou SH. Autophagy-related beclin 1 and head and neck cancers. Onco Targets Ther. 2020;13:6213–27.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  85. Zhu H, He L. Beclin 1 biology and its role in heart disease. Curr Cardiol Rev. 2015;11(3):229–37.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. Khan S, Mentrup HL, Novak EA, Siow VS, Wang Q, Crawford EC, et al. Cyclic GMP-AMP synthase contributes to epithelial homeostasis in intestinal inflammation via Beclin-1-mediated autophagy. FASEB J. 2022;36(5): e22282.

    Article  CAS  PubMed  Google Scholar 

  87. Xia Z, Li H, Irwin MG. Myocardial ischaemia reperfusion injury: the challenge of translating ischaemic and anaesthetic protection from animal models to humans. Br J Anaesth. 2016;117(2):44–62.

    Article  Google Scholar 

  88. Gao B, Yu W, Lv P, Liang X, Sun S, Zhang Y. Parkin overexpression alleviates cardiac aging through facilitating K63-polyubiquitination of TBK1 to facilitate mitophagy. Biochim Biophys Acta Mol Basis Dis. 2021;1867(1): 165997.

    Article  CAS  PubMed  Google Scholar 

  89. Humpton TJ, Alagesan B, DeNicola GM, Lu D, Yordanov GN, Leonhardt CS, et al. Oncogenic KRAS induces NIX-mediated mitophagy to promote pancreatic cancer. Cancer Discov. 2019;9(9):1268–87.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  90. Klionsky DJ, Petroni G, Amaravadi RK, Baehrecke EH, Ballabio A, Boya P, et al. Autophagy in major human diseases. EMBO J. 2021;40(19): e108863.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  91. Wang X. The expanding role of mitochondria in apoptosis. Genes Dev. 2001;15(22):2922–33.

    CAS  PubMed  Google Scholar 

  92. Chao X, Williams SN, Ding WX. Role of mechanistic target of rapamycin in autophagy and alcohol-associated liver disease. Am J Physiol Cell Physiol. 2022;323(4):C1100–11.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  93. Dusabimana T, Kim SR, Kim HJ, Park SW, Kim H. Nobiletin ameliorates hepatic ischemia and reperfusion injury through the activation of SIRT-1/FOXO3a-mediated autophagy and mitochondrial biogenesis. Exp Mol Med. 2019;51(4):1–16.

    Article  CAS  PubMed  Google Scholar 

  94. Gan X, Zhu H, Jiang X, Obiegbusi SC, Yong M, Long X, et al. CircMUC16 promotes autophagy of epithelial ovarian cancer via interaction with ATG13 and miR-199a. Mol Cancer. 2020;19(1):45.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  95. Han B, Zhang Y, Zhang Y, Bai Y, Chen X, Huang R, et al. Novel insight into circular RNA HECTD1 in astrocyte activation via autophagy by targeting MIR142-TIPARP: implications for cerebral ischemic stroke. Autophagy. 2018;14(7):1164–84.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  96. Du WW, Yang W, Li X, Awan FM, Yang Z, Fang L, et al. A circular RNA circ-DNMT1 enhances breast cancer progression by activating autophagy. Oncogene. 2018;37(44):5829–42.

    Article  CAS  PubMed  Google Scholar 

  97. Mohseni M, Behzad G, Farhadi A, Behroozi J, Mohseni H, Valipour B. MicroRNAs regulating autophagy: opportunities in treating neurodegenerative diseases. Front Neurosci. 2024;18:1397106.

    Article  PubMed  PubMed Central  Google Scholar 

  98. Zhang L, Fang Y, Zhao X, Zheng Y, Ma Y, Li S, et al. miR-204 silencing reduces mitochondrial autophagy and ROS production in a murine AD model via the TRPML1-activated STAT3 pathway. Mol Therapy Nucleic Acids. 2021;24:822–31.

    Article  CAS  Google Scholar 

  99. Liang C, Mu Y, Tian H, Wang D, Zhang S, Wang H, et al. MicroRNA-140 silencing represses the incidence of Alzheimer’s disease. Neurosci Lett. 2021;758: 135674.

    Article  CAS  PubMed  Google Scholar 

  100. Ling X, Lu J, Yang J, Qin H, Zhao X, Zhou P, et al. Non-coding RNAs: emerging therapeutic targets in spinal cord ischemia–reperfusion injury. Front Neurol. 2021. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fneur.2021.680210.

    Article  PubMed  PubMed Central  Google Scholar 

  101. Yu W, Xu M, Zhang T, Zhang Q, Zou C. Mst1 promotes cardiac ischemia–reperfusion injury by inhibiting the ERK-CREB pathway and repressing FUNDC1-mediated mitophagy. J Physiol Sci. 2019;69(1):113–27.

    Article  CAS  PubMed  Google Scholar 

  102. Randhawa PK, Gupta MK. Melatonin as a protective agent in cardiac ischemia–reperfusion injury: vision/illusion? Eur J Pharmacol. 2020;885: 173506.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  103. Zhu XZ, Qiu Z, Lei SQ, Leng Y, Li WY, Xia ZY. The role of P53 in myocardial ischemia–reperfusion injury. Cardiovasc Drugs Therapy. 2023;39(1):195.

    Article  Google Scholar 

  104. Deng G, Muqadas M, Adlat S, Zheng H, Li G, Zhu P, et al. Protective effect of hydrogen sulfide on cerebral ischemia–reperfusion injury. Cell Mol Neurobiol. 2023;43(1):15–25.

    Article  CAS  PubMed  Google Scholar 

  105. Chu Y, Yuan X, Tao Y, Yang B, Luo J. Autophagy in muscle regeneration: mechanisms, targets, and therapeutic perspective. Int J Mol Sci. 2024;25(22):11901.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  106. Zhang K, Zhu S, Li J, Jiang T, Feng L, Pei J, et al. Targeting autophagy using small-molecule compounds to improve potential therapy of Parkinson’s disease. Acta Pharm Sin B. 2021;11(10):3015–34.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  107. Van Camp G. Cardiovascular disease prevention. Acta Clin Belg. 2014;69(6):407–11.

    Article  PubMed  Google Scholar 

  108. Liang XH, Jackson S, Seaman M, Brown K, Kempkes B, Hibshoosh H, et al. Induction of autophagy and inhibition of tumorigenesis by beclin 1. Nature. 1999;402(6762):672–6.

    Article  CAS  PubMed  Google Scholar 

  109. Wang Z, Xu L, Duan Z, Zeng L, Yan N, Peng Z. Beclin 1-mediated macroautophagy involves regulation of caspase-9 expression in cervical cancer HeLa cells. Gynecol Oncol. 2007;107(1):107–13.

    Article  CAS  PubMed  Google Scholar 

  110. Tang C, Han H, Liu Z, Liu Y, Yin L, Cai J, et al. Activation of BNIP3-mediated mitophagy protects against renal ischemia–reperfusion injury. Cell Death Dis. 2019;10(9):677.

    Article  PubMed  PubMed Central  Google Scholar 

  111. Luo H, Guo H, Zhou Y, Fang R, Zhang W, Mei Z. Neutrophil extracellular traps in cerebral ischemia/reperfusion injury: friend and foe. Curr Neuropharmacol. 2023;21(10):2079–96.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  112. Xie C, Zhuang XX, Niu Z, Ai R, Lautrup S, Zheng S, et al. Amelioration of Alzheimer’s disease pathology by mitophagy inducers identified via machine learning and a cross-species workflow. Nat Biomed Eng. 2022;6(1):76–93.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  113. Pan XJ, Misrani A, Tabassum S, Yang L. Mitophagy pathways and Alzheimer’s disease: from pathogenesis to treatment. Mitochondrion. 2021;59:37–47.

    Article  CAS  PubMed  Google Scholar 

  114. Liu X, Ye M, Ma L. The emerging role of autophagy and mitophagy in tauopathies: from pathogenesis to translational implications in Alzheimer’s disease. Front Aging Neurosci. 2022;14:1022821.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  115. Moskal N, Riccio V, Bashkurov M, Taddese R, Datti A, Lewis PN, et al. ROCK inhibitors upregulate the neuroprotective Parkin-mediated mitophagy pathway. Nat Commun. 2020;11(1):88.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  116. Wang X. Destructive cellular paths underlying familial and sporadic Parkinson disease converge on mitophagy. Autophagy. 2017;13(11):1998–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  117. Shaltouki A, Hsieh CH, Kim MJ, Wang X. Alpha-synuclein delays mitophagy and targeting Miro rescues neuron loss in Parkinson’s models. Acta Neuropathol. 2018;136(4):607–20.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  118. Chen J, Ren Y, Gui C, Zhao M, Wu X, Mao K, et al. Phosphorylation of Parkin at serine 131 by p38 MAPK promotes mitochondrial dysfunction and neuronal death in mutant A53T α-synuclein model of Parkinson’s disease. Cell Death Dis. 2018;9(6):700.

    Article  PubMed  PubMed Central  Google Scholar 

  119. Zimprich A, Benet-Pagès A, Struhal W, Graf E, Eck SH, Offman MN, et al. A mutation in VPS35, encoding a subunit of the retromer complex, causes late-onset Parkinson disease. Am J Hum Genet. 2011;89(1):168–75.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  120. Guo X, Sun X, Hu D, Wang YJ, Fujioka H, Vyas R, et al. VCP recruitment to mitochondria causes mitophagy impairment and neurodegeneration in models of Huntington’s disease. Nat Commun. 2016;7:12646.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  121. Jurcau A, Jurcau CM. Mitochondria in Huntington’s disease: implications in pathogenesis and mitochondrial-targeted therapeutic strategies. Neural Regen Res. 2023;18(7):1472–7.

    Article  CAS  PubMed  Google Scholar 

  122. Sharma A, Behl T, Sharma L, Aelya L, Bungau S. Mitochondrial dysfunction in Huntington’s disease: pathogenesis and therapeutic opportunities. Curr Drug Targets. 2021;22(14):1637–67.

    Article  CAS  PubMed  Google Scholar 

  123. Rosen DR. Mutations in Cu/Zn superoxide dismutase gene are associated with familial amyotrophic lateral sclerosis. Nature. 1993;364(6435):362.

    Article  CAS  PubMed  Google Scholar 

  124. Tak YJ, Park JH, Rhim H, Kang S. ALS-related mutant SOD1 aggregates interfere with mitophagy by sequestering the autophagy receptor optineurin. Int J Mol Sci. 2020. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/ijms21207525.

    Article  PubMed  PubMed Central  Google Scholar 

  125. Song M, Gong G, Burelle Y, Gustafsson ÅB, Kitsis RN, Matkovich SJ, et al. Interdependence of parkin-mediated mitophagy and mitochondrial fission in adult mouse hearts. Circ Res. 2015;117(4):346–51.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  126. Luan Y, Luan Y, Feng Q, Chen X, Ren KD, Yang Y. Emerging role of mitophagy in the heart: therapeutic potentials to modulate mitophagy in cardiac diseases. Oxid Med Cell Longev. 2021;2021:3259963.

    Article  PubMed  PubMed Central  Google Scholar 

  127. Ajoolabady A, Chiong M, Lavandero S, Klionsky DJ, Ren J. Mitophagy in cardiovascular diseases: molecular mechanisms, pathogenesis, and treatment. Trends Mol Med. 2022;28(10):836–49.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  128. Xu C, Cao Y, Liu R, Liu L, Zhang W, Fang X, et al. Mitophagy-regulated mitochondrial health strongly protects the heart against cardiac dysfunction after acute myocardial infarction. J Cell Mol Med. 2022;26(4):1315–26.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  129. Gao G, Chen W, Yan M, Liu J, Luo H, Wang C, et al. Rapamycin regulates the balance between cardiomyocyte apoptosis and autophagy in chronic heart failure by inhibiting mTOR signaling. Int J Mol Med. 2020;45(1):195–209.

    PubMed  Google Scholar 

  130. Ma L, Zhu J, Gao Q, Rebecchi MJ, Wang Q, Liu L. Restoring pharmacologic preconditioning in the aging heart: role of mitophagy/autophagy. J Gerontol A Biol Sci Med Sci. 2017;72(4):489–98.

    CAS  PubMed  Google Scholar 

  131. Riffelmacher T, Richter FC, Simon AK. Autophagy dictates metabolism and differentiation of inflammatory immune cells. Autophagy. 2018;14(2):199–206.

    Article  CAS  PubMed  Google Scholar 

  132. Gkikas I, Palikaras K, Tavernarakis N. The role of mitophagy in innate immunity. Front Immunol. 2018;9:1283.

    Article  PubMed  PubMed Central  Google Scholar 

  133. Song Y, Zhou Y, Zhou X. The role of mitophagy in innate immune responses triggered by mitochondrial stress. Cell Commun Signal. 2020;18(1):186.

    Article  PubMed  PubMed Central  Google Scholar 

  134. Fu C, Cao N, Liu W, Zhang Z, Yang Z, Zhu W, et al. Crosstalk between mitophagy and innate immunity in viral infection. Front Microbiol. 2022;13:1064045.

    Article  PubMed  PubMed Central  Google Scholar 

  135. Gupta SS, Sharp R, Hofferek C, Kuai L, Dorn GW 2nd, Wang J, et al. NIX-mediated mitophagy promotes effector memory formation in antigen-specific CD8(+) T Cells. Cell Rep. 2019;29(7):1862-77.e7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  136. O’Sullivan TE, Johnson LR, Kang HH, Sun JC. BNIP3- and BNIP3L-mediated mitophagy promotes the generation of natural killer cell memory. Immunity. 2015;43(2):331–42.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  137. Wang S, Tao J, Chen H, Kandadi MR, Sun M, Xu H, et al. Ablation of Akt2 and AMPKα2 rescues high fat diet-induced obesity and hepatic steatosis through Parkin-mediated mitophagy. Acta Pharm Sin B. 2021;11(11):3508–26.

    Article  CAS  Google Scholar 

  138. Picca A, Faitg J, Auwerx J, Ferrucci L, D’Amico D. Mitophagy in human health, ageing and disease. Nat Metab. 2023;5(12):2047–61.

    Article  PubMed  Google Scholar 

  139. Wu H, Wang Y, Li W, Chen H, Du L, Liu D, et al. Deficiency of mitophagy receptor FUNDC1 impairs mitochondrial quality and aggravates dietary-induced obesity and metabolic syndrome. Autophagy. 2019;15(11):1882–98.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  140. Shan Z, Fa WH, Tian CR, Yuan CS, Jie N. Mitophagy and mitochondrial dynamics in type 2 diabetes mellitus treatment. Aging. 2022;14(6):2902–19.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  141. Sidarala V, Pearson GL, Parekh VS, Thompson B, Christen L, Gingerich MA, et al. Mitophagy protects β cells from inflammatory damage in diabetes. JCI Insight. 2020;5(24):141138.

    Article  PubMed  Google Scholar 

  142. Guan Y, Wang Y, Li B, Shen K, Li Q, Ni Y, et al. Mitophagy in carcinogenesis, drug resistance and anticancer therapeutics. Cancer Cell Int. 2021;21(1):350.

    Article  PubMed  PubMed Central  Google Scholar 

  143. Wang Y, Liu HH, Cao YT, Zhang LL, Huang F, Yi C. The role of mitochondrial dynamics and mitophagy in carcinogenesis, metastasis and therapy. Front Cell Dev Biol. 2020;8:413.

    Article  PubMed  PubMed Central  Google Scholar 

  144. Lin Q, Chen J, Gu L, Dan X, Zhang C, Yang Y. New insights into mitophagy and stem cells. Stem Cell Res Ther. 2021;12(1):452.

    Article  PubMed  PubMed Central  Google Scholar 

  145. Naik PP, Panigrahi S, Parida R, Praharaj PP, Bhol CS, Patil S, et al. Metabostemness in cancer: linking metaboloepigenetics and mitophagy in remodeling cancer stem cells. Stem Cell Rev Rep. 2022;18(1):198–213.

    Article  PubMed  Google Scholar 

  146. Chatzinikita E, Maridaki M, Palikaras K, Koutsilieris M, Philippou A. The role of mitophagy in skeletal muscle damage and regeneration. Cells. 2023. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/cells12050716.

    Article  PubMed  PubMed Central  Google Scholar 

  147. Lei Y, Gan M, Qiu Y, Chen Q, Wang X, Liao T, et al. The role of mitochondrial dynamics and mitophagy in skeletal muscle atrophy: from molecular mechanisms to therapeutic insights. Cell Mol Biol Lett. 2024;29(1):59.

    Article  PubMed  PubMed Central  Google Scholar 

  148. Wang Y, Cai J, Tang C, Dong Z. Mitophagy in acute kidney injury and kidney repair. Cells. 2020. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/cells9020338.

    Article  PubMed  PubMed Central  Google Scholar 

  149. Bhatia D, Choi ME. The emerging role of mitophagy in kidney diseases. J Life Sci (Westlake Village, Calif). 2019;1(3):13–22.

    Google Scholar 

  150. Yang K, Li T, Geng Y, Zou X, Peng F, Gao W. The role of mitophagy in the development of chronic kidney disease. PeerJ. 2024;12: e17260.

    Article  PubMed  PubMed Central  Google Scholar 

  151. Williams JA, Ding WX. A mechanistic review of mitophagy and its role in protection against alcoholic liver disease. Biomolecules. 2015;5(4):2619–42.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  152. Lemasters JJ, Zhong Z. Mitophagy in hepatocytes: types, initiators and role in adaptive ethanol metabolism. Liver research. 2018;2(3):125–32.

    Article  PubMed  Google Scholar 

  153. Zhang NP, Liu XJ, Xie L, Shen XZ, Wu J. Impaired mitophagy triggers NLRP3 inflammasome activation during the progression from nonalcoholic fatty liver to nonalcoholic steatohepatitis. Lab Invest. 2019;99(6):749–63.

    Article  CAS  PubMed  Google Scholar 

  154. Luo Z, Yan S, Chao Y, Shen M. Unveiling the mitophagy puzzle in non-alcoholic fatty liver disease (NAFLD): six hub genes for early diagnosis and immune modulatory roles. Heliyon. 2024;10(7): e28935.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  155. Letsiou E, Sammani S, Wang H, Belvitch P, Dudek SM. Parkin regulates lipopolysaccharide-induced proinflammatory responses in acute lung injury. Trans Res. 2017;181:71–82.

    Article  CAS  Google Scholar 

  156. Zhong Y, Xia S, Wang G, Liu Q, Ma F, Yu Y, et al. The interplay between mitophagy and mitochondrial ROS in acute lung injury. Mitochondrion. 2024;78: 101920.

    Article  CAS  PubMed  Google Scholar 

  157. Zhang Z, Chen Z, Liu R, Liang Q, Peng Z, Yin S, et al. Bcl-2 proteins regulate mitophagy in lipopolysaccharide-induced acute lung injury via PINK1/parkin signaling pathway. Oxid Med Cell Longev. 2020;2020:6579696.

    PubMed  PubMed Central  Google Scholar 

  158. Albano GD, Montalbano AM, Gagliardo R, Profita M. Autophagy/mitophagy in airway diseases: impact of oxidative stress on epithelial cells. Biomolecules. 2023. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/biom13081217.

    Article  PubMed  PubMed Central  Google Scholar 

  159. Zhou WC, Qu J, Xie SY, Sun Y, Yao HW. Mitochondrial dysfunction in chronic respiratory diseases: implications for the pathogenesis and potential therapeutics. Oxid Med Cell Longev. 2021;2021:5188306.

    Article  PubMed  PubMed Central  Google Scholar 

  160. Sreedhar A, Aguilera-Aguirre L, Singh KK. Mitochondria in skin health, aging, and disease. Cell Death Dis. 2020;11(6):444.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  161. Jiménez-Loygorri JI, Benítez-Fernández R, Viedma-Poyatos Á, Zapata-Muñoz J, Villarejo-Zori B, Gómez-Sintes R, et al. Mitophagy in the retina: viewing mitochondrial homeostasis through a new lens. Prog Retin Eye Res. 2023;96: 101205.

    Article  PubMed  Google Scholar 

  162. Sridevi Gurubaran I, Viiri J, Koskela A, Hyttinen JMT, Paterno JJ, Kis G, et al. Mitophagy in the retinal pigment epithelium of dry age-related macular degeneration investigated in the NFE2L2/PGC-1α(-/-) mouse model. Int J Mol Sci. 2020. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/ijms21061976.

    Article  PubMed  PubMed Central  Google Scholar 

  163. Stavropoulos D, Grewal MK, Petriti B, Chau KY, Hammond CJ, Garway-Heath DF, et al. The role of mitophagy in glaucomatous neurodegeneration. Cells. 2023. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/cells12151969.

    Article  PubMed  PubMed Central  Google Scholar 

  164. Dai Y, Hu X, Sun X. Overexpression of parkin protects retinal ganglion cells in experimental glaucoma. Cell Death Dis. 2018;9(2):88.

    Article  PubMed  PubMed Central  Google Scholar 

  165. Peng F, Jiang D, Xu W, Sun Y, Zha Z, Tan X, et al. AMPK/MFF activation: role in mitochondrial fission and mitophagy in Dry Eye. Invest Ophthalmol Vis Sci. 2022;63(12):18.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  166. Li G, Li J, Shao R, Zhao J, Chen M. FUNDC1: a promising mitophagy regulator at the mitochondria-associated membrane for cardiovascular diseases. Front Cell Dev Biol. 2021;9:788634.

    Article  PubMed  PubMed Central  Google Scholar 

  167. Xie Y, Kang R, Klionsky DJ, Tang D. GPX4 in cell death, autophagy, and disease. Autophagy. 2023;19(10):2621–38.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  168. Penna C, Perrelli MG, Pagliaro P. Mitochondrial pathways, permeability transition pore, and redox signaling in cardioprotection: therapeutic implications. Antioxid Redox Signal. 2013;18(5):556–99.

    Article  CAS  PubMed  Google Scholar 

  169. Pilátová MB, Solárová Z, Mezencev R, Solár P. Ceramides and their roles in programmed cell death. Adv Med Sci. 2023;68(2):417–25.

    Article  PubMed  Google Scholar 

  170. Daskalaki I, Gkikas I, Tavernarakis N. Hypoxia and selective autophagy in cancer development and therapy. Front Cell Dev Biol. 2018;6:104.

    Article  PubMed  PubMed Central  Google Scholar 

  171. Zhang W, Ren H, Xu C, Zhu C, Wu H, Liu D, et al. Hypoxic mitophagy regulates mitochondrial quality and platelet activation and determines severity of I/R heart injury. Elife. 2016. https://doiorg.publicaciones.saludcastillayleon.es/10.7554/eLife.21407.

    Article  PubMed  PubMed Central  Google Scholar 

  172. Qiao L, Hu J, Qiu X, Wang C, Peng J, Zhang C, et al. LAMP2A, LAMP2B and LAMP2C: similar structures, divergent roles. Autophagy. 2023;19(11):2837–52.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  173. Czabotar PE, Garcia-Saez AJ. Mechanisms of BCL-2 family proteins in mitochondrial apoptosis. Nat Rev Mol Cell Biol. 2023;24(10):732–48.

    Article  CAS  PubMed  Google Scholar 

  174. Zheng X, Wan J, Tan G. The mechanisms of NLRP3 inflammasome/pyroptosis activation and their role in diabetic retinopathy. Front Immunol. 2023;14:1151185.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  175. Liao H, Liu S, Ma Q, Huang H, Goel A, Torabian P, et al. Endoplasmic reticulum stress induced autophagy in cancer and its potential interactions with apoptosis and ferroptosis. Biochim Biophys Acta. 2025;1872(1): 119869.

    Article  CAS  Google Scholar 

  176. Jiang T, Ma C, Chen H. Unraveling the ultrastructure and dynamics of autophagic vesicles: Insights from advanced imaging techniques. FASEB Bioadv. 2024;6(7):189–99.

    Article  PubMed  PubMed Central  Google Scholar 

  177. Buchan JR, Parker R. Eukaryotic stress granules: the ins and outs of translation. Mol Cell. 2009;36(6):932–41.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  178. Ohn T, Anderson P. The role of posttranslational modifications in the assembly of stress granules. Wiley Interdisc Rev RNA. 2010;1(3):486–93.

    Article  CAS  Google Scholar 

  179. Wu X, Zheng Y, Liu M, Li Y, Ma S, Tang W, et al. BNIP3L/NIX degradation leads to mitophagy deficiency in ischemic brains. Autophagy. 2020;17(8):1934–46.

    Article  PubMed  PubMed Central  Google Scholar 

  180. Yamamoto H, Zhang S, Mizushima N. Autophagy genes in biology and disease. Nat Rev Genet. 2023;24(6):382–400.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  181. Hui Q, Karlstetter M, Xu Z, Yang J, Zhou L, Eilken HM, et al. Inhibition of the Keap1–Nrf2 protein–protein interaction protects retinal cells and ameliorates retinal ischemia–reperfusion injury. Free Radical Biol Med. 2020;146:181–8.

    Article  CAS  Google Scholar 

  182. Cai J, Chen X, Liu X, Li Z, Shi A, Tang X, et al. AMPK: the key to ischemia–reperfusion injury. J Cell Physiol. 2022;237(11):4079–96.

    Article  CAS  PubMed  Google Scholar 

  183. Zhao D, Liu J, Wang M, Zhang X, Zhou M. Epidemiology of cardiovascular disease in China: current features and implications. Nat Rev Cardiol. 2019;16(4):203–12.

    Article  PubMed  Google Scholar 

  184. Roshandel G, Khoshnia M, Poustchi H, Hemming K, Kamangar F, Gharavi A, et al. Effectiveness of polypill for primary and secondary prevention of cardiovascular diseases (PolyIran): a pragmatic, cluster-randomised trial. The Lancet. 2019;394(10199):672–83.

    Article  CAS  Google Scholar 

  185. Boengler K, Bornbaum J, Schlüter K-D, Schulz R. P66shc and its role in ischemic cardiovascular diseases. Basic Res Cardiol. 2019. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s00395-019-0738-x.

    Article  PubMed  Google Scholar 

  186. Li H, Sureda A, Devkota HP, Pittalà V, Barreca D, Silva AS, et al. Curcumin, the golden spice in treating cardiovascular diseases. Biotechnol Advd. 2020. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.biotechadv.2019.01.010.

    Article  Google Scholar 

  187. Welt FGP, Batchelor W, Spears JR, Penna C, Pagliaro P, Ibanez B, et al. Reperfusion injury in patients with acute myocardial infarction: JACC scientific statement. J Am Coll Cardiol. 2024;83(22):2196–213.

    Article  PubMed  Google Scholar 

  188. Buja LM. Pathobiology of myocardial and cardiomyocyte injury in ischemic heart disease: perspective from seventy years of cell injury research. Exp Mol Pathol. 2024;140: 104944.

    Article  CAS  PubMed  Google Scholar 

  189. Campos JC, Queliconi BB, Bozi LHM, Bechara LRG, Dourado PMM, Andres AM, et al. Exercise reestablishes autophagic flux and mitochondrial quality control in heart failure. Autophagy. 2017;13(8):1304–17.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  190. Shao D, Kolwicz SC Jr, Wang P, Roe ND, Villet O, Nishi K, et al. Increasing fatty acid oxidation prevents high-fat diet-induced cardiomyopathy through regulating parkin-mediated mitophagy. Circulation. 2020;142(10):983–97.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  191. Guo Z, Tian Y, Gao J, Zhou B, Zhou X, Chang X, et al. Enhancement of mitochondrial homeostasis: a novel approach to attenuate hypoxic myocardial injury. Int J Med Sci. 2024;21(15):2897–911.

    Article  PubMed  PubMed Central  Google Scholar 

  192. Popov SV, Mukhomedzyanov AV, Voronkov NS, Derkachev IA, Boshchenko AA, Fu F, et al. Regulation of autophagy of the heart in ischemia and reperfusion. Apoptosis. 2023;28(1–2):55–80.

    Article  CAS  PubMed  Google Scholar 

  193. Wu Y, Liu H, Wang X. Cardioprotection of pharmacological postconditioning on myocardial ischemia/reperfusion injury. Life Sci. 2021;264: 118628.

    Article  CAS  PubMed  Google Scholar 

  194. Mancardi D, Ottolenghi S, Attanasio U, Tocchetti CG, Paroni R, Pagliaro P, et al. Janus, or the inevitable battle between too much and too little oxygen. Antioxid Redox Signal. 2022;37(13–15):972–89.

    Article  CAS  PubMed  Google Scholar 

  195. Yang H, Li L, Zhou K, Wang Y, Guan T, Chai C, et al. Shengmai injection attenuates the cerebral ischemia/reperfusion induced autophagy via modulation of the AMPK, mTOR and JNK pathways. Pharm Biol. 2016;54(10):2288–97.

    Article  CAS  PubMed  Google Scholar 

  196. Huang YG, Tao W, Yang SB, Wang JF, Mei ZG, Feng ZT. Autophagy: novel insights into therapeutic target of electroacupuncture against cerebral ischemia/ reperfusion injury. Neural Regen Res. 2019;14(6):954–61.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  197. Fan YY, Hu WW, Nan F, Chen Z. Postconditioning-induced neuroprotection, mechanisms and applications in cerebral ischemia. Neurochem Int. 2017;107:43–56.

    Article  CAS  PubMed  Google Scholar 

  198. Zhang Q, Jia M, Wang Y, Wang Q, Wu J. Cell death mechanisms in cerebral ischemia–reperfusion injury. Neurochem Res. 2022;47(12):3525–42.

    Article  CAS  PubMed  Google Scholar 

  199. Mao B, Yuan W, Wu F, Yan Y, Wang B. Autophagy in hepatic ischemia–reperfusion injury. Cell Death Disc. 2023. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41420-023-01387-0.

    Article  Google Scholar 

  200. Ma X, McKeen T, Zhang J, Ding W-X. Role and mechanisms of mitophagy in liver diseases. Cells. 2020. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/cells9040837.

    Article  PubMed  PubMed Central  Google Scholar 

  201. Dowdell AS, Colgan SP. Metabolic host-microbiota interactions in autophagy and the pathogenesis of inflammatory bowel disease (IBD). Pharmaceuticals (Basel). 2021;14(8):708.

    Article  CAS  PubMed  Google Scholar 

  202. Ge Y, Zhang Q, Jiao Z, Li H, Bai G, Wang H. Adipose-derived stem cells reduce liver oxidative stress and autophagy induced by ischemia–reperfusion and hepatectomy injury in swine. Life Sci. 2018;214:62–9.

    Article  CAS  PubMed  Google Scholar 

  203. Onishi M, Yamano K, Sato M, Matsuda N, Okamoto K. Molecular mechanisms and physiological functions of mitophagy. EMBO J. 2021;40(3): e104705.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  204. Xu Y, Tang Y, Lu J, Zhang W, Zhu Y, Zhang S, et al. PINK1-mediated mitophagy protects against hepatic ischemia/reperfusion injury by restraining NLRP3 inflammasome activation. Free Radical Biol Med. 2020;160:871–86.

    Article  CAS  Google Scholar 

  205. Kawakami T, Gomez IG, Ren S, Hudkins K, Roach A, Alpers CE, et al. Deficient autophagy results in mitochondrial dysfunction and FSGS. J Am Soc Nephrol. 2015;26(5):1040–52.

    Article  CAS  PubMed  Google Scholar 

  206. Livingston MJ, Shu S, Fan Y, Li Z, Jiao Q, Yin X-M, et al. Tubular cells produce FGF2 via autophagy after acute kidney injury leading to fibroblast activation and renal fibrosis. Autophagy. 2022;19(1):256–77.

    Article  PubMed  PubMed Central  Google Scholar 

  207. Periyasamy-Thandavan S, Jiang M, Wei Q, Smith R, Yin XM, Dong Z. Autophagy is cytoprotective during cisplatin injury of renal proximal tubular cells. Kidney Int. 2008;74(5):631–40.

    Article  CAS  PubMed  Google Scholar 

  208. Inoue K, Kuwana H, Shimamura Y, Ogata K, Taniguchi Y, Kagawa T, et al. Cisplatin-induced macroautophagy occurs prior to apoptosis in proximal tubules in vivo. Clin Exp Nephrol. 2010;14(2):112–22.

    Article  CAS  PubMed  Google Scholar 

  209. Lee SY, Kim SI, Choi ME. Therapeutic targets for treating fibrotic kidney diseases. Trans Res. 2015;165(4):512–30.

    Article  CAS  Google Scholar 

  210. Tanaka T. A mechanistic link between renal ischemia and fibrosis. Med Mol Morphol. 2017;50(1):1–8.

    Article  CAS  PubMed  Google Scholar 

  211. Lin TA, Wu VC, Wang CY. Autophagy in chronic kidney diseases. Cells. 2019;8(1):61.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  212. De Rechter S, Decuypere JP, Ivanova E, van den Heuvel LP, De Smedt H, Levtchenko E, et al. Autophagy in renal diseases. Pediatr Nephrol (Berlin, Germany). 2016;31(5):737–52.

    Article  Google Scholar 

  213. Liu N, Shi Y, Zhuang S. Autophagy in chronic kidney diseases. Kidney Dis (Basel, Switzerland). 2016;2(1):37–45.

    Google Scholar 

  214. Ta HQ, Kuppusamy M, Sonkusare SK, Roeser ME, Laubach VE. The endothelium: gatekeeper to lung ischemia–reperfusion injury. Respir Res. 2024;25(1):172.

    Article  PubMed  PubMed Central  Google Scholar 

  215. Rizzo AN, Schmidt EP. The role of the alveolar epithelial glycocalyx in acute respiratory distress syndrome. Am J Physiol Cell Physiol. 2023;324(4):C799-c806.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  216. Chen-Yoshikawa TF. Ischemia–reperfusion injury in lung transplantation. Cells. 2021. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/cells10061333.

    Article  PubMed  PubMed Central  Google Scholar 

  217. Lederhuber H, Massey LH, Abeysiri S, Roman MA, Rajaretnam N, McDermott FD, et al. Preoperative intravenous iron and the risk of blood transfusion in colorectal cancer surgery: meta-analysis of randomized clinical trials. Br J Surg. 2024;111(1):320.

    Article  Google Scholar 

  218. Kim S, Choi S, Kang D. Quantitative and qualitative analysis of autophagy flux using imaging. BMB Rep. 2020;53(5):241–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  219. Li L, Cheng H, Zhou Y, Zhao D, Zhang X, Wang Y, et al. METTL3 regulates autophagy of hypoxia-induced cardiomyocytes by targeting ATG7. Cell Death Discov. 2025;11(1):37.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  220. Chen L, Mao LS, Xue JY, Jian YH, Deng ZW, Mazhar M, et al. Myocardial ischemia–reperfusion injury: the balance mechanism between mitophagy and NLRP3 inflammasome. Life Sci. 2024;355: 122998.

    Article  CAS  PubMed  Google Scholar 

  221. Shi B, Ma M, Zheng Y, Pan Y, Lin X. mTOR and Beclin1: Two key autophagy-related molecules and their roles in myocardial ischemia/reperfusion injury. J Cell Physiol. 2019;234(8):12562–8.

    Article  CAS  PubMed  Google Scholar 

  222. Liu M, Yao Y, Tan F, Wang J, Hu R, Du J, et al. Sodium-glucose co-transporter 2 (SGLT-2) inhibitors ameliorate renal ischemia–reperfusion injury (IRI) by modulating autophagic processes. Trans Res. 2025;277:27–38.

    Article  CAS  Google Scholar 

  223. Dutta S, Ganguly A, Ghosh RS. An overview of the unfolded protein response (upr) and autophagy pathways in human viral oncogenesis. Int Rev Cell Mol Biol. 2024;386:81–131.

    Article  PubMed  Google Scholar 

  224. Jarocki M, Turek K, Saczko J, Tarek M, Kulbacka J. Lipids associated with autophagy: mechanisms and therapeutic targets. Cell Death Discov. 2024;10(1):460.

    Article  PubMed  PubMed Central  Google Scholar 

  225. Mao B, Yuan W, Wu F, Yan Y, Wang B. Autophagy in hepatic ischemia–reperfusion injury. Cell Death Discov. 2023;9(1):115.

    Article  PubMed  PubMed Central  Google Scholar 

  226. Mehta P, Shende P. Dual role of autophagy for advancements from conventional to new delivery systems in cancer. Biochim Biophys Acta. 2023;1867(10): 130430.

    Article  CAS  Google Scholar 

  227. Ghosh I, Sankhe R, Mudgal J, Arora D, Nampoothiri M. Spermidine, an autophagy inducer, as a therapeutic strategy in neurological disorders. Neuropeptides. 2020;83: 102083.

    Article  CAS  PubMed  Google Scholar 

  228. Mo Y, Sun YY, Liu KY. Autophagy and inflammation in ischemic stroke. Neural Regen Res. 2020;15(8):1388–96.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  229. Wang X, Wu Y. Protective effects of autophagy inhibitor 3-methyladenine on ischemia–reperfusion-induced retinal injury. Int Ophthalmol. 2020;40(5):1095–101.

    Article  PubMed  Google Scholar 

  230. Mathew B, Chennakesavalu M, Sharma M, Torres LA, Stelman CR, Tran S, et al. Autophagy and post-ischemic conditioning in retinal ischemia. Autophagy. 2021;17(6):1479–99.

    Article  CAS  PubMed  Google Scholar 

Download references

Funding

This study was funded by the Fund Project of National Natural Science Foundation of China (82302421) and the Fund Project of National Natural Science Foundation of China (82272214).

Author information

Authors and Affiliations

Authors

Contributions

Lingxuan Tang, Wangzheqi Zhang, and Yan Liao contributed to the manuscript writing and figure preparation, Lingxuan Tang, Weijie Wang, and Wenwen Shi designed the work, and Changli Wang and Xiaoming Deng supervised the work. All authors read and approved the final manuscript.

Corresponding authors

Correspondence to Xiaoming Deng, Changli Wang or Wenwen Shi.

Ethics declarations

Ethics approval and consent to participate

Not applicable. This manuscript does not contain any studies with human participants or animals performed by any of the authors.

Consent for publication

Not applicable. This manuscript does not include details, images, or videos relating to an individual person.

Competing interests

The authors declare that they have no competing interests.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Tang, L., Zhang, W., Liao, Y. et al. Autophagy: a double-edged sword in ischemia–reperfusion injury. Cell Mol Biol Lett 30, 42 (2025). https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s11658-025-00713-x

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s11658-025-00713-x

Keywords