Skip to main content

Mitophagy in perioperative neurocognitive disorder: mechanisms and therapeutic strategies

Abstract

Perioperative neurocognitive disorder (PND) is a common neurological complication after surgery/anesthesia in elderly patients that affect postoperative outcome and long-term quality of life, which increases the cost of family and social resources. The pathological mechanism of PND is complex and not fully understood, and the methods of prevention and treatment of PND are very limited, so it is particularly important to analyze the mechanism of PND. Research indicates that mitochondrial dysfunction is pivotal in the initiation and progression of PND, although the precise mechanisms remain elusive and could involve disrupted mitophagy. We reviewed recent studies on the link between mitophagy and PND, highlighting the role of key proteins in abnormal mitophagy and discussing therapeutic strategies aimed at mitophagy regulation. This provides insights into the mechanisms underlying PND and potential therapeutic targets.

Introduction

Perioperative neurocognitive disorder (PND) is a prevalent and severe neurological complication in elderly patients following surgery/anesthesia. It encompasses both objectively measured cognitive decline and subjectively reported cognitive deficits, as well as changes in daily living activities before and after surgery [1]. Affecting 5–55% of the elderly patients [2], PND significantly increases the rates of postoperative morbidity, mortality, and the incidence of long-term cognitive dysfunction [3, 4]. The pathogenesis of PND involves various factors, there are no effective drugs or interventions to prevent PND yet. Mitochondria, often referred to as the “powerhouse of the cell”, play a crucial role in neuronal development and synaptic plasticity [5, 6]. Mitochondrial quality control is a key factor in the health and survival of brain neurons, and mitochondrial dysfunction is closely associated with a variety of neurodegenerative diseases [7,8,9,10]. Similarly, mitochondrial dysfunction is increasingly recognized as playing a critical role in the development and progression of PND [11,12,13], although the specific mechanisms remain poorly understood. Research in animal and cellular models of PND has led to the hypothesis that abnormal mitophagy, which lead to the accumulation of damaged mitochondria, may be a key factor [14,15,16]. This review examines the molecular mechanisms underlying mitophagy abnormalities associated with PND, as well as promising therapeutic strategies targeting mitophagy abnormalities in PND demonstrated in recent studies. The goal is to provide insights into the mechanisms behind the occurrence and progression of PND and to identify potential new therapeutic targets.

Mitophagy

Autophagy is the cellular process in which autophagosomes transport misfolded proteins, lipids, or damaged organelles within the cytoplasm to lysosomes for degradation and clearance, playing a significant role in hippocampal-dependent cognitive adaptation [17]. Mitophagy, a selective form of autophagy, preserves mitochondrial function and cellular homeostasis by specifically targeting damaged or redundant mitochondria for removal from the cytoplasm [18]. Mitophagy is crucial for maintaining intracellular mitochondrial quality and quantity equilibrium, thereby supporting normal mitochondrial function [19,20,21]. Under external stressors such as hypoxia, nutrient deprivation, and cellular senescence, damaged mitochondria produce substantial amounts of ROS, which triggers mitophagy to initiate self-clearance by removing damaged mitochondria, thereby reducing ROS accumulation and maintaining cellular stability [22]. Additionally, mitophagy can initiate caspase-family-induced apoptosis and support cell survival by eliminating damaged mitochondria and preventing the excessive release of cytochrome C associated with mitochondrial damage [23]. Neurons, as highly differentiated cells, demand substantial energy to sustain their intricate physiological functions, making mitophagy balance essential for neuronal health and survival [24]. Disruptions in neuronal mitophagy can lead to the accumulation of damaged mitochondria, increase ROS production, and compromise mitochondrial respiratory function, which in turn reduces energy supply. This energy deficit impairs neuronal function, diminishes physiological resilience, and ultimately trigger programmed cell death in neurons [25] (Fig. 1).

Fig. 1
figure 1

Physiological function of mitophagy in neurons. Under conditions such as hypoxia, nutrient deprivation, and cellular senescence, damaged mitochondria generate high levels of ROS, triggering mitophagy to clear these damaged mitochondria. Concurrently, mitophagy initiates caspase-family-induced apoptosis by removing damaged mitochondria, thus preventing excessive cytochrome C release due to mitochondrial injury. When mitophagy is impaired, the resulting ROS accumulation reduces ATP production and initiates programmed neuronal death. (This image was generated and provided under the BioRender license using BioRender. All rights and ownership of BioRender content belong to BioRender.)

The PINK1/Parkin pathway is the most extensively studied mitophagy pathway in neurological diseases [26]. PINK1 is a mitochondrial serine/threonine kinase, with Parkin functions as an E3 ubiquitin ligase. Upon mitochondrial damage, PINK1 enters the inner mitochondrial membrane (IMM), accumulates on the outer mitochondrial membrane (OMM), and phosphorylates the serine 65 residue on ubiquitin molecules [27]. Concurrently, Parkin is recruited to the damaged mitochondria and phosphorylated by PINK1, then activates its E3 ligase activity and promotes the ubiquitination of multiple OMM proteins, leading to the formation of ubiquitin chains [28, 29]. Adaptor proteins, such as P62/SQSTM1, OPTN, NBR1, NDP52, and TAX1BP1, recognize phosphorylated ubiquitin chains on mitochondrial proteins. Through binding to microtubule-associated protein 1A/1B light chain 3 (LC3), these adaptor proteins label damaged mitochondria for phagocytosis and degradation by autophagosomes [30,31,32].

Mitophagy regulates the molecular mechanism of PND

With advancing insights into the regulatory network of mitophagy, its intricate role in the pathogenesis of PND has gradually become apparent. Furthermore, the involvement of Nod-like receptor protein 3 (NLRP3) inflammasome, Synaptosome Associated Protein 25 (SNAP25), pyroptosis, and neurotoxic proteins in the mitophagy process has been demonstrated in cellular and animal models of PND (Table 1), while the conclusions regarding the role of mitophagy in the onset and progression of PND are not yet consistent (Fig. 2).

Table 1 Molecular mechanisms of mitophagy and interventions in PND
Fig. 2
figure 2

Molecular mechanism by which mitophagy regulates PND. A Surgery/anesthesia elevates ROS levels in the hippocampus, activates NLRP3 inflammasomes, stimulates the release of pro-inflammatory cytokines, intensifies neuroinflammatory responses, and contributes to the development of PND. Activation of the NLRP3 inflammasome induces mitochondrial damage, mtDNA buildup, and mtROS production, while inhibiting mitophagy by promoting caspase-1-dependent Parkin cleavage. B Surgery/anesthesia can inhibit mitophagy and promote the production of PND by promoting the production of proteins such as Aβ, tau and TNFAIP1. (This image was generated and provided under the BioRender license using BioRender. All rights and ownership of BioRender content belong to BioRender.)

In 2018, Ye et al. were the first to propose a connection between PND and mitophagy [14]. They performed abdominal exploratory surgery on 4-month-old female C57BL/6J mice under sevoflurane anesthesia, and observed that surgery/anesthesia upregulated the expression of autophagy-related proteins, including LC3-II, Beclin-1, Parkin, and PINK1, damages mitochondria, and induces behavioral and cognitive impairments in mice. Pretreatment with Honokiol (HNK) was found to further enhance autophagy biomarker expression, reduce mitochondrial ROS levels, alleviate mitochondrial structural damage, and improve postoperative cognitive function in surgery/anesthesia-induced mice. In contrast, the use of the autophagy inhibitor 3-Methyladenine (3-MA) reversed the effects of HNK on mitophagy and cognition in these mice. In addition, Chen et al. demonstrated that mitophagy dysfunction is a primary cause of sevoflurane-induced mitochondrial damage in H4 cells and PND in aged rats [15]. Sevoflurane treatment induces mitochondrial dysfunction and mitophagy deficiencies in H4 cells and aged rat hippocampal neurons, which include increased ROS levels, decreased membrane potential, impaired respiratory function, accumulation of the mitochondrial marker protein Tomm20, and reduced levels of the lysosomal marker protein LAMP1. Ac-YVAD-cmk can inhibit sevoflurane-induced intracellular lysosomal dysfunction of H4 cell, promote mitophagy, and alleviate intracellular ROS levels and mtROS accumulation [33]. Administration of rapamycin, an autophagy activator, reduced sevoflurane-induced ROS production, restored mitochondrial phagocytosis levels, alleviated mitochondrial damage and improved cognitive function in aged rats. Conversely, it is worth noting that Wang and colleagues performed laparotomy on 18-month-old male C57BL/6J mice under sevoflurane anesthesia and found promoted mitophagy and reduced mitochondrial area in hippocampal neurons, accompanied by ROS accumulation, neuronal apoptosis, and tau protein misfolding [16]. Preoperative administration of varenicline improved cognitive function in mice by reducing mitophagy levels, restoring mitochondrial function, lowering oxidative stress, and inhibiting tau phosphorylation. In all, these findings suggest that PND may be linked to the relative capacity of mitophagy and the accumulation of damaged mitochondria.

NLRP3 inflammasome

Neuroinflammation plays a central role in the pathogenesis of PND [34]. Surgery/anesthesia can trigger a systemic inflammatory response that activates inflammation in the central nervous system through various pathways, such as impairing neuronal function, hindering neuronal regeneration, and inducing apoptosis, which collectively lead to cognitive decline [35]. NLRP3 inflammasome is well expressed in neuronal tissues and plays a crucial role in the progression of chronic neurodegenerative diseases [36]. The NLRP3 inflammasome is a complex composed of sensors (NOD-like receptor protein 3, NLRP3), adaptors (apoptosis-associated speck-like protein with a caspase recruitment domain, ASC), and effectors (caspase-1) that are vital for inflammatory regulation [37]. Upon detecting exogenous pathogens or endogenous cell damage signals, NLRP3 recruits ASC proteins, triggering inflammasome assembly and activating caspase-1. Activated caspase-1 then promotes the maturation and secretion of inflammatory cytokines IL-1β and TNF-α, intensifying inflammation and initiating pyroptosis [38]. Mitochondrial dysfunction prompts the overproduction of mitochondrial ROS (mtROS) and the release of mitochondrial DNA (mtDNA), which activates the NLRP3 inflammasome and induces an inflammatory response [39]. The activation of the NLRP3 inflammasome, in turn, damages mitochondria, reduces mitochondrial membrane potential, accumulates mtDNA, elevates mtROS production, and blocks mitophagy by activating caspase-1-dependent cleavage of Parkin. Caspase-1, a key cysteine protease activated by the NLRP3 inflammasome, cleaves Parkin into N-terminal (Parkin-N) and C-terminal (Parkin-C) fragments [40]. This cleavage disrupts Parkin’s structure, leading to the loss of its E3 ubiquitin ligase activity. Consequently, mitochondrial outer membrane proteins cannot be ubiquitinated effectively, blocking the initiation of mitophagy. This impairment further exacerbates mitochondrial dysfunction, elevating intracellular ROS levels and intensifying the inflammatory response, which inhibits mitophagy, intensifies the mitochondrial inflammatory response, and further exacerbates mitochondrial dysfunction [41, 42]. In aged mice, surgery/anesthesia was shown to increase hippocampal mtROS levels, activate NLRP3 inflammasomes, stimulate the release of pro-inflammatory cytokines, amplify neuroinflammatory responses, and impair spatial cognition [43]. Enhancing mitophagy can mitigate mitochondrial structural damage, lower mtROS and MDA production [14], inhibit NLRP3 inflammasome activation, reduce neuroinflammatory responses, and improve cognitive function [44]. Zheng et al. demonstrated that sevoflurane raised the expression of NLRP3, cleaved caspase-1, IL-1β, and IL-18 in the hippocampus of aged mice. The caspase-1 inhibitor Ac-YVAD-cmk was able to alleviate mitochondrial dysfunction and restore mitophagic balance by inhibiting NLRP3 inflammasome activation, thereby improving sevoflurane-induced cognitive impairment [33]. After lipopolysaccharide (LPS) treatment, the expression levels of NLRP3, caspase-1 and IL-1β in BV2 microglia cell were significantly higher than those in normal BV2 microglia cell, and the mitochondrial membrane potential was lower than that in normal BV2 microglia cell. Rapamycin could significantly reduce the activation of NLRP3 inflammasome [45]. Additionally, a retrospective clinical study demonstrated that elevated levels of serum NLRP3 protein, IL-18, and IL-1β after cardiac surgery were associated with an increased risk of cognitive impairment 7 days after surgery. After adjusting for confounding variables, high serum NLRP3 protein levels immediately after surgery were identified as an independent risk factor for the development of PND [46]. This evidence supports the involvement of NLRP3 in PND in human studies. Altogether, NLRP3 inflammasome activation can inhibit mitophagy, promote mitochondrial damage, and then impair cognitive function. The enhancement of mitophagy can alleviate mitochondrial damage and inhibit the activation of NLRP3 inflammasome, thereby improving cognitive impairment.

SNAP25

As a presynaptic protein, SNAP25 is a critical component of the SNARE complex, playing a significant role in regulating neurotransmitter release, synaptic plasticity, neuronal repair, and long-term memory formation [47,48,49]. SNAP25 is the only protein known to localize to mitochondrial OMMs and has been shown to influence autophagy in neuronal cells [50, 51]. Reduced expression of SNAP25 is also associated with the inactivation of the PINK1/Parkin pathway [52]. Studies have shown that the reduction of SNAP25 leads to defects in presynaptic short-term plasticity, abnormal dendritic spine morphology, and a significant decrease in the long time potentiation amplitude of postsynaptic terminals, which in turn affects learning and memory functions [53]. At the same time, the downregulation of SNAP25 expression was detected in the brain tissues of AD patients and PND model mice induced by anesthesia [54, 55]. Wang et al. found that surgery/anesthesia inhibited the expression of PINK1 and LC3 while promoting the cleavage of caspase-3 and GSDME. Overexpression of SNAP25 using AAV9 particles counteracted these effects, reducing PND severity. In SH-SY5Y cells treated with isoflurane and lipopolysaccharide (LPS), levels of PINK1, Parkin, and LC3-II decreased, while P62 accumulated. SNAP25 knockdown blocked the accumulation of PINK1 in the OMM and the transport of Parkin to the mitochondria, further worsening mitophagy defects induced by isoflurane and LPS. Conversely, SNAP25 overexpression helped inhibit cell death in SH-SY5Y neuronal cells by restoring PINK1-dependent mitophagy [56]. The expression of TNFAIP1 was significantly upregulated in the hippocampus of mice after surgery/anesthesia. TNFAIP1 is a ubiquitin ligase whose N-terminal region contains a BTB domain capable of binding to SNAP25. Through the K48-linked polyubiquitination pathway, TNFAIP1 can be targeted for degradation of SNAP25. Targeted knockdown of TNFAIP1 expression could ameliorate surgery/anesthesia induced memory deficits and PINK1/Parkin-dependent mitophagy defects by stabilizing SNAP25 [57]. Therefore, stabilizing or upregulating the expression of SNAP25, which can restore PINK1/Parkin-dependent mitophagy, contributes to the improvement of cognitive dysfunction induced by surgical anesthesia.

Pyroptosis

Pyroptosis, an inflammatory form of programmed cell death mediated by gasdermin (GSDM) family proteins, plays a critical role in the pathogenesis of PND [58, 59]. GSDMS is a gene family with conserved structural motifs, which plays an important role in cell differentiation and proliferation, cell death, mitochondrial homeostasis, anti-microbial, inflammation and tumorigenesis [60]. All members of the GSDM family (except DFNB59) contain a cytotoxic n-terminal domain (NT) and a c-terminal inhibitory domain (CT), which are connected by a flexible connecting region [61]. After being activated by pathogenic or damaging signals, the N-terminal domains of GSDM proteins A–E induce pyroptosis by binding to membrane lipids after cleavage by activated caspases, forming pores in the cell membrane. This leads to osmotic imbalance, cell swelling, and eventual rupture of the cell membrane [62]. The NLRP3/caspase-1/GSDMD pathway is a classic pyroptosis pathway, and its activation in microglia and astrocytes has been linked to postoperative cognitive deficits [63]. In neonatal rats, continuous exposure to 2% sevoflurane for 6 h significantly increased the expression of pro-apoptotic proteins such as Bax and pyroptosis-related proteins, including cleaved caspase-1, cleaved GSDMD, NLRP3, and ASC, leading to both apoptosis and pyroptosis of hippocampal neurons [64]. In aged mice, surgery/anesthesia-induced hippocampal mitochondrial dysfunction activates NLRP3 inflammasome–caspase-1-dependent pyroptosis, which impairs learning and memory in behavioral tests [65]. Chen et al. recently demonstrated that dexmedetomidine promotes mitophagy by upregulating PINK1, reducing caspase-1/11–GSDMD-dependent hippocampal neuron death, and improving postoperative cognitive function in elderly rats [66]. Interestingly, GSDME is more highly expressed than GSDMD in the brain and in certain neuronal cell lines, such as SH-SY5Y neuroblastoma and HT-22 hippocampal neurons, suggesting a significant role for GSDME in nervous system pyroptosis [67]. Wang et al. highlighted in in vivo and in vitro studies that PINK1 downregulation promotes caspase-3/GSDME-dependent pyroptosis by reducing mitophagy, leading to PND-like behaviors in rats [68]. In animal experiments, surgery/anesthesia induced downregulation of PINK1 and LC3-II in the hippocampus of 12-month-old male rats, along with abnormal accumulation of P62. Further PINK1 knockdown inhibited mitophagy, promoted caspase-3/GSDME-dependent pyroptosis, and worsened cognitive dysfunction. Conversely, PINK1 overexpression alleviated cognitive impairment, restored mitophagy, and inhibited GSDME-dependent pyroptosis. In in vitro studies, LPS-treated SH-SY5Y cells displayed PINK1-mediated mitophagy deficiency and GSDME-dependent pyroptosis, with these effects worsening when PINK1 function was lost. Accordingly, pyroptosis through the NLRP3/caspase-1/GSDMD and caspase-3/GSDME pathways, regulates the expression of PINK1 and mitophagy, playing a key role in the pathogenesis of PND.

Neurotoxic proteins

Recent studies have revealed the increased levels of tau and Aβ proteins in the cerebrospinal fluid of patients undergoing anesthesia [69,70,71,72,73], which is associated with the incidence and severity of postoperative delirium [74, 75]. Tau protein, a member of the microtubule-binding protein family, is primarily enriched around neuronal axons and plays a crucial role in regulating and maintaining microtubule stability, which is essential for neuronal axon transport [76]. Abnormal phosphorylation and aggregation of tau protein disrupt neuronal structure and function, are linked to mitochondrial dysfunction, and contribute significantly to the pathogenesis of surgically induced cognitive dysfunction and neurodegenerative diseases such as AD [77,78,79]. Aberrant tau phosphorylation and aggregation can impact mitochondrial dynamics, bioenergetics, and mitophagy, either by inhibiting Parkin translocation to mitochondria [80] or by altering mitochondrial membrane potential [81]. Aβ protein, a neurotoxic peptide of 39–43 amino acids, is produced through the cleavage of amyloid precursor protein (APP) by β-secretase and γ-secretase [82]. Aβ protein can accumulate within mitochondria by interacting with mitochondrial proteins, which promotes excessive ROS production and leads to mitochondrial damage. Additionally, Aβ protein can impair the mitophagy–lysosomal pathway by causing lysosomal dysfunction, leading to abnormal aggregation of mitophagosomes and substrates [83]. Enhancing mitophagy has been shown to reduce Aβ protein accumulation and tau hyperphosphorylation, thereby reversing memory impairment in mice [84]. In conclusion, the abnormal accumulation of tau and Aβ proteins after anesthesia is closely related to PND. By regulating the abnormal accumulation of these proteins and enhancing mitophagy, it may provide new strategies for the prevention and treatment of PND and AD.

Mitophagy-related intervention strategies and potential clinical applications in PND research models

PND affects the prognosis and quality of life of patients after surgery, and increases the consumption of family and medical resources. It is urgent to adopt effective prevention and treatment strategies to reduce the occurrence and development of PND. In view of the role of mitophagy in PND, we summarized the existing pharmacological and non-pharmacological strategies targeting mitophagy in order to provide new ideas for the prevention and treatment of PND.

Pharmacological strategies

Rapamycin is an immunosuppressant that binds specifically to the mammalian target of rapamycin (mTOR) by forming a complex with a 12-kDa FK506 binding protein (FKBP12) [85]. Mammalian target of rapamycin (mTOR) is a highly conserved serine-threonine kinase, can regulate protein synthesis, energy metabolism, lipid metabolism, mitochondrial and lysosomal biogenesis [86]. Rapamycin has been shown to reverse sevoflurane-induced autophagic flux damage through the mTOR signaling, thereby improving cognitive deficits in aged rats [87]. In sevoflurane-treated hippocampal tissue, rapamycin counteracts the increased levels of mitochondrial markers Tomm20 and P62 and the decreased expression of the lysosomal marker LAMP1, promoting mitophagy and reducing the number of damaged mitochondria. At the same time, rapamycin improving mitochondrial quality in neuronal cells, which significantly alleviates sevoflurane-induced cognitive impairment in rats [15]. Additionally, rapamycin promotes mitophagy in LPS-treated HT22 cells, reducing mitochondrial hyperdivision and improving mitochondrial function [88].

Honokiol (HNK), a natural bisphenol compound derived from magnolia officinalis, exhibits multiple pharmacological activities, including anti-tumor, antioxidant, anti-inflammatory, and neuroprotective properties [89]. As a small polyphenolic molecule, HNK easily crosses the blood–brain barrier, inhibiting intracellular Ca2 + influx, caspase-3 activity, and the abnormal aggregation of Aβ protein, thereby providing neuroprotection [90]. HNK has been shown to support microglial phagocytic function by improving mitochondrial function [91]. Furthermore, HNK alleviates neuroinflammation and reverses surgery- and anesthesia-induced learning and memory deficits in mice by promoting mitophagy, lowering mtROS levels, and inhibiting NLRP3 inflammasome activation [14].

Varenicline, a non-nicotine smoking cessation drug approved by the U.S. Food and Drug Administration, acts as a selective partial agonist of the α4β2-nicotinic acetylcholine receptor (nAChR) [92]. The α4β2-nAChR, a ligand-gated ion channel widely distributed in the nervous system, regulates neurotransmitters such as acetylcholine, γ-aminobutyric acid, and norepinephrine, playing a central role in learning, memory, cognition, attention, inflammation, and pain [93]. This nAChR subunit in the central nervous system has been proposed as a potential target for treating age-related cognitive decline and various neurodegenerative and psychiatric disorders [94, 95]. Studies have shown that varenicline reduces neuroinflammation, tau misfolding, DNA damage, and apoptosis, thereby mitigating PND-like behaviors in aged mice [96]. Varenicline also alleviates surgery  and anesthesia-induced hippocampal oxidative stress, mitochondrial dysfunction, and aberrant mitophagy, reducing tau phosphorylation and improving cognitive function in mice through the PKR/STAT3 pathway [16].

Liraglutide, a glucagon-like peptide-1 (GLP-1) receptor agonist, is primarily used to treat type 2 diabetes and obesity [97]. Recent research has expanded our understanding of liraglutide’s pharmacological effects, showing potential efficacy in managing neurological diseases. Liraglutide improves neural encoding in the ventromedial prefrontal cortex and ventral striatum, correcting adaptation mispredictions in patients with impaired insulin sensitivity and normalizing sensory-related learning deficits in obese patients [98]. In AD mouse models, liraglutide improves learning and memory by reducing Aβ protein deposition and tau hyperphosphorylation, promoting synaptic plasticity, and reducing neuronal degeneration [99, 100]. In alcohol-dependent mice, liraglutide significantly increased dendritic spine density and synaptic protein levels, alleviating anxiety and memory deficits linked to alcohol withdrawal [101]. Additionally, liraglutide mitigates neuroinflammation, reduces synaptic loss and impaired plasticity, and alleviates anxiety-like behaviors and cognitive deficits in aged mice post-cardiac surgery by increasing mitophagy and inhibiting NLRP3 inflammasome and microglial activation [44]. Recently, results from a Phase 2b clinical trial presented at the Alzheimer's Association International Conference (AAIC) demonstrated that liraglutide can slow the atrophy of key brain regions in AD patients and reduce the rate of cognitive decline [102]. However, it has not yet been reported in clinical trials of PND, and further research is still needed.

Elamipretide (SS-31), a novel mitochondria-targeted antioxidant, primarily localizes to the inner mitochondrial membrane, where it directly scavenges ROS and stabilizes the electron transport chain. SS-31 has demonstrated neuroprotective effects across various neurological disorders [103,104,105,106]. It has been shown to reverse isoflurane-induced mitochondrial dysfunction in the hippocampus of aged mice, promote brain-derived neurotrophic factor (BDNF) signaling and synaptic plasticity, reduce neuronal damage, and improve cognitive function [107]. In mouse models with LPS-induced neuroinflammation, SS-31 significantly increased energy production and mitochondrial membrane potential in the hippocampus, indicating its potential to restore function in damaged neurons [105]. SS-31 inhibits activation of the NLRP3 inflammasome–caspase-1 pathway in the hippocampus following surgery or anesthesia, thereby reducing lesion-related cell death and neuroinflammation, rescuing neuronal damage and synaptic dysfunction, and alleviating cognitive impairment [65]. Furthermore, SS-31 promotes PHB2-mediated mitophagy activation, inhibiting mtDNA release, M1 microglial polarization, and inflammation via the cGAS–STING pathway, ultimately restoring neurocognitive function in elderly mice after surgery [108].

Dexmedetomidine (Dex), is a highly selective α2 adrenergic receptor agonist with sedative, anxiolytic, hypnotic, and analgesic properties that is commonly used in the induction and maintenance of general anesthesia [109]. In recent years, studies have found that Dex has a positive effect on cognitive impairment, especially PND, which is expected to become an ideal drug for the treatment of PND. Clinical studies have found that the prophylactic use of low-dose Dex significantly reduces postoperative delirium in older patients after cardiac and noncardiac surgery [110]. In animal experiments, Chen et al. found that Dex improved motor symptoms in Parkinson mice by enhancing PINK1/Parkin-induced mitophagy, improving mitochondrial function, and protecting dopaminergic neurons [111]. Suo and Wang found that Dex alleviated sevoflurane-induced neurotoxic effects by activating mitophagy in the hippocampus of rats and improved learning and memory ability in rats [112], it provides a potential strategy for the treatment of PND.

Propofol is a general anesthetic widely used in the induction and maintenance of general anesthesia and sedation of regional anesthesia. Propofol produces sedation, hypnosis, and amnesia effects by activating the inhibitory transmitter γ-aminobutyric acid (GABA) in the central nervous system [113]. Dai et al. found that propofol could reduce the damage of mitochondria by inhibiting the PINK1/Parkin mitophagy pathway, and alleviate the impairment of learning and memory in sleep-deprived rats [114]. However, Liang et al. found that propofol time-dependently decreased the expression levels of PINK1 mRNA and protein, inducing apoptosis in hippocampal neurons [115]. As early as 2015, Han et al. suggested that repeated use of low-dose propofol had no apparent effect on cognitive function. However, repeated administration of propofol at recommended or higher doses caused a significant increase in the expression of apoptotic factors and pro-inflammatory cytokine in the hippocampus of neonatal rats, impairing cognitive function [116]. Therefore, more research is needed to explore the dose and timing of propofol administration to mitigate side effects and improve its role in the prevention and treatment of PND.

In conclusion, various drugs have shown significant potential therapeutic effects in animal models by improving PND through modulation of mitophagy. For example, sirolimus improves cognitive function by restoring mitophagy and reducing the accumulation of damaged mitochondria, and HNK reduces neuroinflammation by enhancing mitophagy and reducing oxidative stress. By reducing Aβ protein deposition and tau protein hyperphosphorylation, liraglutide enhances mitophagy and improves cognitive function. However, these strategies still face challenges in clinical application. Future studies need to further optimize drug dosage and administration time to improve treatment efficacy and reduce side effects. For example, the effects of the dose and timing of administration of propofol on cognitive function vary and require further investigation. In addition, long-term follow-up studies are needed to evaluate the durable effects of the drug and to consider the accessibility and safety of the drug, especially for older patients. Meanwhile, exploring the synergy between drugs, such as the combined use of rapamycin and Dex, may improve PND more effectively by enhancing mitophagy and inhibiting neuroinflammation.

Non-pharmacological treatment strategies

Electroacupuncture (EA) is an innovative therapy that combines physical nerve stimulation with Traditional Chinese Medicine (TCM) acupuncture and is widely applied to treat various cognitive dysfunctions [117]. Preoperative EA therapy has been shown to reduce the incidence of PND and decrease levels of inflammatory markers in elderly patients [118]. Studies suggest that the mechanisms by which EA alleviates PND may involve neuroinflammation, oxidative stress, autophagy, and the microbiota–gut–brain axis [119]. In PND rat models, EA has been found to improve spatial memory by reducing mitochondrial damage from oxidative stress and increasing mitophagy to prevent the buildup of damaged mitochondria [120]. Additionally, inhibiting ROS production and IL-1β expression has been shown to further improve spatial memory in these models [121].

Cognitive training (CT) is to improve cognitive function through the guidance of different cognitive domains such as memory, attention, execution and language [122]. Common CT methods include computerized CT (CCT), multimedia education, paper-and-pencil cognitive training, memory training, and communication with patients [123]. CT can increase the density of dopamine D1 receptors in the cortex and improve the capacity of working memory [124]. At the same time, CT can also improve the cognitive reserve of patients [125] and reduce the incidence of PND [126]. In AD mice, repeated CT reduced glycogen synthase kinase-3β (gsk3β) activity, reduced phosphorylated tau deposition, and improved spatial memory [127]. However, more research is needed to determine whether CT can affect mitophagy to improve cognitive function.

Physical activity (PA) is widely regarded as beneficial for physical and mental health, and can significantly improve cognitive function [128]. Studies have shown that regular exercise before surgery and early postoperative exercise can significantly reduce the incidence of PND [129, 130]. PA can increase BDNF levels, increase hippocampal volume, and improve memory function [131]. PA can also affect the activation of microglia and astrocytes, reduce neuroinflammation, and enhance cognitive function in AD patients [132]. Continuous aerobic exercise for 12 weeks can promote the level of mitophagy in the hippocampus of APP/PS1 transgenic AD mice, clear damaged or abnormal mitochondria, maintain mitochondrial protein homeostasis, and improve the learning and spatial memory levels of mice [133]. Zhao et al. also found that PA can enhance PINK1/Parkin-mediated mitophagy activity by upregulating the SIRT1–FOXO1/3 axis, reduce Aβ protein deposition, and improve the learning and memory ability of AD mice [134]. These findings suggest that PA-activated mitophagy may be a promising strategy for the prevention of PND.

In summary, non-pharmacological strategies such as EA, CT, and PA show potential positive effects in the prevention and treatment of PND. For example, EA modulates neuroinflammation and oxidative stress by physical nerve stimulation, increases mitophagy, and improves cognitive function; CT reduces the incidence of PND by enhancing cognitive reserve; PA improves cognitive function by increasing BDNF levels and enhancing mitophagy. However, further optimization and long-term efficacy evaluation of these strategies need to be further investigated. The effects of different types and durations of CT and PA on PND need to be explored in the future and their long-term effects evaluated. At the same time, accessibility and safety of non-pharmacological strategies need to be considered. In addition, explore the synergy of non-pharmacological strategies with pharmacological treatments to more effectively improve PND.

Conclusion

PND is a common complication in elderly patients following surgery or anesthesia, with its underlying pathophysiological mechanisms primarily involving neuroinflammation, oxidative stress, insufficient energy supply, and neuronal apoptosis. Recent studies suggest that mitochondrial dysfunction plays a crucial role in the development of PND. Mitophagy, a key mechanism to maintain mitochondrial homeostasis, alleviates oxidative stress by clearing damaged mitochondria and protecting neuronal function. This review explores the relationship between PND and abnormal mitophagy, detailing the molecular mechanisms involved and potential intervention strategies. Studies have indicated that the occurrence of PND is closely related to the balance of mitophagy and the accumulation of damaged mitochondria. In the PND model, NLRP3 inflammasome activation inhibits mitophagy, exacerbating mitochondrial damage, while enhancing mitophagy alleviates inflammation and improves cognitive function. Furthermore, proteins such as SNAP25, pyroptosis mediators, and neurotoxic proteins play pivotal roles in PND pathogenesis and are linked to mitophagy regulation. Although some pharmacological and non-pharmacological interventions have demonstrated potential in improving PND by regulating mitophagy in animal models, their clinical efficacy and safety remain to be fully validated. Future research should focus on identifying the specific regulatory pathways of mitophagy in PND and exploring the temporal interplay of various pathological processes during its pathogenesis. Additionally, more clinical trials are necessary to evaluate the safety and efficacy of current intervention strategies and to develop more effective prevention and treatment approaches for PND.

Data availability

No datasets were generated or analysed during the current study.

References

  1. Evered L, Silbert B, Knopman DS, Scott DA, DeKosky ST, Rasmussen LS, et al. Recommendations for the nomenclature of cognitive change associated with anaesthesia and surgery-2018. Br J Anaesth. 2018;121(5):1005–12.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  2. Daiello LA, Racine AM, Yun Gou R, Marcantonio ER, Xie Z, Kunze LJ, et al. Postoperative delirium and postoperative cognitive dysfunction: overlap and divergence. Anesthesiology. 2019;131(3):477–91.

    Article  PubMed  Google Scholar 

  3. Inouye SK, Marcantonio ER, Kosar CM, Tommet D, Schmitt EM, Travison TG, et al. The short-term and long-term relationship between delirium and cognitive trajectory in older surgical patients. Alzheimers Dement. 2016;12(7):766–75.

    Article  PubMed  Google Scholar 

  4. Boone MD, Sites B, von Recklinghausen FM, Mueller A, Taenzer AH, Shaefi S. Economic burden of postoperative neurocognitive disorders among US medicare patients. JAMA Netw Open. 2020;3(7): e208931.

    Article  PubMed  PubMed Central  Google Scholar 

  5. Li S, Xiong GJ, Huang N, Sheng ZH. The cross-talk of energy sensing and mitochondrial anchoring sustains synaptic efficacy by maintaining presynaptic metabolism. Nat Metab. 2020;2(10):1077–95.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  6. Iwata R, Casimir P, Erkol E, Boubakar L, Planque M, Gallego López IM, et al. Mitochondria metabolism sets the species-specific tempo of neuronal development. Science. 2023;379(6632):eabn4705.

    Article  PubMed  CAS  Google Scholar 

  7. Chang Y, Wang C, Zhu J, Zheng S, Sun S, Wu Y, et al. SIRT3 ameliorates diabetes-associated cognitive dysfunction via regulating mitochondria-associated ER membranes. J Transl Med. 2023;21(1):494.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  8. Zhao P, Zhang J, Kuai J, Li L, Li X, Feng N, et al. TAT-PEP alleviated cognitive impairment by alleviating neuronal mitochondria damage and apoptosis after cerebral ischemic reperfusion injury. Mol Neurobiol. 2023;60(10):5655–71.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  9. Samanta S, Akhter F, Roy A, Chen D, Turner B, Wang Y, et al. New cyclophilin D inhibitor rescues mitochondrial and cognitive function in Alzheimer’s disease. Brain. 2024;147(5):1710–25.

    Article  PubMed  Google Scholar 

  10. Wu DP, Wei YS, Du YX, Liu LL, Yan QQ, Zhao YD, et al. Ameliorative role of mitochondrial therapy in cognitive function of vascular dementia mice. J Alzheimers Dis. 2024;97(3):1381–92.

    Article  PubMed  CAS  Google Scholar 

  11. Netto MB, de Oliveira Junior AN, Goldim M, Mathias K, Fileti ME, da Rosa N, et al. Oxidative stress and mitochondrial dysfunction contributes to postoperative cognitive dysfunction in elderly rats. Brain Behav Immun. 2018;73:661–9.

    Article  PubMed  CAS  Google Scholar 

  12. Yang Y, Liu Y, Zhu J, Song S, Huang Y, Zhang W, et al. Neuroinflammation-mediated mitochondrial dysregulation involved in postoperative cognitive dysfunction. Free Radic Biol Med. 2022;178:134–46.

    Article  PubMed  CAS  Google Scholar 

  13. Xu X, Zhou B, Liu J, Ma Q, Zhang T, Wu X. Ru360 alleviates postoperative cognitive dysfunction in aged mice by inhibiting MCU-mediated mitochondrial dysfunction. Neuropsychiatr Dis Treat. 2023;19:1531–42.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  14. Ye JS, Chen L, Lu YY, Lei SQ, Peng M, Xia ZY. Honokiol-mediated mitophagy ameliorates postoperative cognitive impairment induced by surgery/sevoflurane via inhibiting the activation of NLRP3 inflammasome in the hippocampus. Oxid Med Cell Longev. 2019;2019:8639618.

    Article  PubMed  PubMed Central  Google Scholar 

  15. Chen Y, Zhang P, Lin X, Zhang H, Miao J, Zhou Y, et al. Mitophagy impairment is involved in sevoflurane-induced cognitive dysfunction in aged rats. Aging (Albany NY). 2020;12(17):17235–56.

    Article  PubMed  CAS  Google Scholar 

  16. Wang J, Zhu S, Lu W, Li A, Zhou Y, Chen Y, et al. Varenicline improved laparotomy-induced cognitive impairment by restoring mitophagy in aged mice. Eur J Pharmacol. 2022;916: 174524.

    Article  PubMed  CAS  Google Scholar 

  17. Glatigny M, Moriceau S, Rivagorda M, Ramos-Brossier M, Nascimbeni AC, Lante F, et al. Autophagy is required for memory formation and reverses age-related memory decline. Curr Biol. 2019;29(3):435-48.e8.

    Article  PubMed  CAS  Google Scholar 

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

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  19. Ashrafi G, Schwarz TL. The pathways of mitophagy for quality control and clearance of mitochondria. Cell Death Differ. 2013;20(1):31–42.

    Article  PubMed  CAS  Google Scholar 

  20. Cen X, Zhang M, Zhou M, Ye L, Xia H. Mitophagy regulates neurodegenerative diseases. Cells. 2021;10(8):1876.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  21. Li X, Huang L, Lan J, Feng X, Li P, Wu L, et al. Molecular mechanisms of mitophagy and its roles in neurodegenerative diseases. Pharmacol Res. 2021;163: 105240.

    Article  PubMed  CAS  Google Scholar 

  22. Schofield JH, Schafer ZT. Mitochondrial reactive oxygen species and mitophagy: a complex and nuanced relationship. Antioxid Redox Signal. 2021;34(7):517–30.

    Article  PubMed  CAS  Google Scholar 

  23. Wanderoy S, Hees JT, Klesse R, Edlich F, Harbauer AB. Kill one or kill the many: interplay between mitophagy and apoptosis. Biol Chem. 2020;402(1):73–88.

    Article  PubMed  Google Scholar 

  24. Han S, Zhang M, Jeong YY, Margolis DJ, Cai Q. The role of mitophagy in the regulation of mitochondrial energetic status in neurons. Autophagy. 2021;17(12):4182–201.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  25. Yang K, Yan Y, Yu A, Zhang R, Zhang Y, Qiu Z, et al. Mitophagy in neurodegenerative disease pathogenesis. Neural Regen Res. 2024;19(5):998–1005.

    Article  PubMed  CAS  Google Scholar 

  26. Li J, Yang D, Li Z, Zhao M, Wang D, Sun Z, et al. PINK1/Parkin-mediated mitophagy in neurodegenerative diseases. Ageing Res Rev. 2023;84: 101817.

    Article  PubMed  CAS  Google Scholar 

  27. Matsuda N, Sato S, Shiba K, Okatsu K, Saisho K, Gautier CA, et al. PINK1 stabilized by mitochondrial depolarization recruits Parkin to damaged mitochondria and activates latent Parkin for mitophagy. J Cell Biol. 2010;189(2):211–21.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  28. Kondapalli C, Kazlauskaite A, Zhang N, Woodroof HI, Campbell DG, Gourlay R, et al. PINK1 is activated by mitochondrial membrane potential depolarization and stimulates Parkin E3 ligase activity by phosphorylating Serine 65. Open Biol. 2012;2(5): 120080.

    Article  PubMed  PubMed Central  Google Scholar 

  29. Fritsch LE, Moore ME, Sarraf SA, Pickrell AM. Ubiquitin and receptor-dependent mitophagy pathways and their implication in neurodegeneration. J Mol Biol. 2020;432(8):2510–24.

    Article  PubMed  CAS  Google Scholar 

  30. Geisler S, Holmström KM, Skujat D, Fiesel FC, Rothfuss OC, Kahle PJ, et al. PINK1/Parkin-mediated mitophagy is dependent on VDAC1 and P62/SQSTM1. Nat Cell Biol. 2010;12(2):119–31.

    Article  PubMed  CAS  Google Scholar 

  31. Lazarou M, Sliter DA, Kane LA, Sarraf SA, Wang C, Burman JL, et al. The ubiquitin kinase PINK1 recruits autophagy receptors to induce mitophagy. Nature. 2015;524(7565):309–14.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  32. Padman BS, Nguyen TN, Uoselis L, Skulsuppaisarn M, Nguyen LK, Lazarou M. LC3/GABARAPs drive ubiquitin-independent recruitment of Optineurin and NDP52 to amplify mitophagy. Nat Commun. 2019;10(1):408.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  33. Zheng D, Wang H, Zhou Y, Chen Y, Chen G. Ac-YVAD-cmk ameliorated sevoflurane-induced cognitive dysfunction and revised mitophagy impairment. PLoS ONE. 2023;18(1): e0280914.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  34. Leslie M. The post-op brain. Science. 2017;356(6341):898–900.

    Article  PubMed  CAS  Google Scholar 

  35. Terrando N, Monaco C, Ma D, Foxwell BM, Feldmann M, Maze M. Tumor necrosis factor-alpha triggers a cytokine cascade yielding postoperative cognitive decline. Proc Natl Acad Sci U S A. 2010;107(47):20518–22.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  36. Sarkar S, Rokad D, Malovic E, Luo J, Harischandra DS, Jin H, et al. Manganese activates NLRP3 inflammasome signaling and propagates exosomal release of ASC in microglial cells. Sci Signal. 2019;12(563):eaat9900.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  37. Fu J, Wu H. Structural mechanisms of NLRP3 inflammasome assembly and activation. Annu Rev Immunol. 2023;41:301–16.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  38. Xu J, Núñez G. The NLRP3 inflammasome: activation and regulation. Trends Biochem Sci. 2023;48(4):331–44.

    Article  PubMed  CAS  Google Scholar 

  39. Mishra SR, Mahapatra KK, Behera BP, Patra S, Bhol CS, Panigrahi DP, et al. Mitochondrial dysfunction as a driver of NLRP3 inflammasome activation and its modulation through mitophagy for potential therapeutics. Int J Biochem Cell Biol. 2021;136: 106013.

    Article  PubMed  CAS  Google Scholar 

  40. Yu J, Nagasu H, Murakami T, Hoang H, Broderick L, Hoffman HM, et al. Inflammasome activation leads to Caspase-1-dependent mitochondrial damage and block of mitophagy. Proc Natl Acad Sci U S A. 2014;111(43):15514–9.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  41. Youm YH, Grant RW, McCabe LR, Albarado DC, Nguyen KY, Ravussin A, et al. Canonical Nlrp3 inflammasome links systemic low-grade inflammation to functional decline in aging. Cell Metab. 2013;18(4):519–32.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  42. Xu Y, Wang J, Xu W, Ding F, Ding W. Prohibitin 2-mediated mitophagy attenuates renal tubular epithelial cells injury by regulating mitochondrial dysfunction and NLRP3 inflammasome activation. Am J Physiol Renal Physiol. 2019;316(2):F396-407.

    Article  PubMed  Google Scholar 

  43. Jiang W, Liu F, Li H, Wang K, Cao X, Xu X, et al. TREM2 ameliorates anesthesia and surgery-induced cognitive impairment by regulating mitophagy and NLRP3 inflammasome in aged C57/BL6 mice. Neurotoxicology. 2022;90:216–27.

    Article  PubMed  CAS  Google Scholar 

  44. Jia M, Lv X, Zhu T, Shen JC, Liu WX, Yang JJ. Liraglutide ameliorates delirium-like behaviors of aged mice undergoing cardiac surgery by mitigating microglia activation via promoting mitophagy. Psychopharmacology. 2023. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s00213-023-06492-7.

    Article  PubMed  PubMed Central  Google Scholar 

  45. Lu J, Zong Y, Tao X, Dai H, Song J, Zhou H. Anesthesia/surgery-induced learning and memory dysfunction by inhibiting mitophagy-mediated NLRP3 inflammasome inactivation in aged mice. Exp Brain Res. 2024;242(2):417–27.

    Article  PubMed  CAS  Google Scholar 

  46. Ma G, Sun P, Chen Y, Jiang X, Zhang C, Qu B, et al. NLRP3 inflammasome activation contributes to the cognitive decline after cardiac surgery. Front Surg. 2022;9: 992769.

    Article  PubMed  PubMed Central  Google Scholar 

  47. Wang W, Wang F, Liu J, Zhao W, Zhao Q, He M, et al. SNAP25 ameliorates sensory deficit in rats with spinal cord transection. Mol Neurobiol. 2014;50(2):290–304.

    Article  PubMed  CAS  Google Scholar 

  48. Zhang H, Therriault J, Kang MS, Ng KP, Pascoal TA, Rosa-Neto P, et al. Cerebrospinal fluid synaptosomal-associated protein 25 is a key player in synaptic degeneration in mild cognitive impairment and Alzheimer’s disease. Alzheimers Res Ther. 2018;10(1):80.

    Article  PubMed  PubMed Central  Google Scholar 

  49. Yang L, Shi LJ, Shen SY, Yang JY, Lv SS, Wang ZC, et al. Toward antifragility: social defeat stress enhances learning and memory in young mice via hippocampal synaptosome associated protein 25. Psychol Sci. 2023;34(5):616–32.

    Article  PubMed  Google Scholar 

  50. Chen M, Qiu T, Wu J, Yang Y, Wright GD, Wu M, et al. Extracellular anti-angiogenic proteins augment an endosomal protein trafficking pathway to reach mitochondria and execute apoptosis in HUVECs. Cell Death Differ. 2018;25(11):1905–20.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  51. Mu Y, Yan X, Li D, Zhao D, Wang L, Wang X, et al. NUPR1 maintains autolysosomal efflux by activating SNAP25 transcription in cancer cells. Autophagy. 2018;14(4):654–70.

    Article  PubMed  CAS  Google Scholar 

  52. Li D, Zheng J, Wang M, Feng L, Ren Z, Liu Y, et al. Changes of TSPO-mediated mitophagy signaling pathway in learned helplessness mice. Psychiatry Res. 2016;245:141–7.

    Article  PubMed  Google Scholar 

  53. Irfan M, Gopaul KR, Miry O, Hökfelt T, Stanton PK, Bark C. SNAP-25 isoforms differentially regulate synaptic transmission and long-term synaptic plasticity at central synapses. Sci Rep. 2019;9(1):6403.

    Article  PubMed  PubMed Central  Google Scholar 

  54. Bereczki E, Francis PT, Howlett D, Pereira JB, Höglund K, Bogstedt A, et al. Synaptic proteins predict cognitive decline in Alzheimer’s disease and Lewy body dementia. Alzheimers Dement. 2016;12(11):1149–58.

    Article  PubMed  Google Scholar 

  55. Zhang X, Wu W, Luo Y, Wang Z. Transcranial photobiomodulation therapy ameliorates perioperative neurocognitive disorder through modulation of mitochondrial function in aged mice. Neuroscience. 2022;490:236–49.

    Article  PubMed  CAS  Google Scholar 

  56. Wang W, Gao W, Zhang L, Xia Z, Zhao B. SNAP25 ameliorates postoperative cognitive dysfunction by facilitating PINK1-dependent mitophagy and impeding caspase-3/GSDME-dependent pyroptosis. Exp Neurol. 2023;367: 114463.

    Article  PubMed  CAS  Google Scholar 

  57. Wang W, Gao W, Gong P, Song W, Bu X, Hou J, et al. Neuronal-specific TNFAIP1 ablation attenuates postoperative cognitive dysfunction via targeting SNAP25 for K48-linked ubiquitination. Cell Commun Signal. 2023;21(1):356.

    Article  PubMed  PubMed Central  Google Scholar 

  58. Que YY, Zhu T, Zhang FX, Peng J. Neuroprotective effect of DUSP14 overexpression against isoflurane-induced inflammatory response, pyroptosis and cognitive impairment in aged rats through inhibiting the NLRP3 inflammasome. Eur Rev Med Pharmacol Sci. 2020;24(12):7101–13.

    PubMed  Google Scholar 

  59. Hsu SK, Li CY, Lin IL, Syue WJ, Chen YF, Cheng KC, et al. Inflammation-related pyroptosis, a novel programmed cell death pathway, and its crosstalk with immune therapy in cancer treatment. Theranostics. 2021;11(18):8813–35.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  60. Yang J, Jiang J. Gasdermins: a dual role in pyroptosis and tumor immunity. Front Immunol. 2024;15:1322468.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  61. Broz P, Pelegrín P, Shao F. The gasdermins, a protein family executing cell death and inflammation. Nat Rev Immunol. 2020;20(3):143–57.

    Article  PubMed  CAS  Google Scholar 

  62. Moujalled D, Strasser A, Liddell JR. Molecular mechanisms of cell death in neurological diseases. Cell Death Differ. 2021;28(7):2029–44.

    Article  PubMed  PubMed Central  Google Scholar 

  63. Zhang Z, Ma Q, Velagapudi R, Barclay WE, Rodriguiz RM, Wetsel WC, et al. Annexin-A1 tripeptide attenuates surgery-induced neuroinflammation and memory deficits through regulation the NLRP3 inflammasome. Front Immunol. 2022;13: 856254.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  64. Wu Z, Tan J, Lin L, Zhang W, Yuan W. microRNA-140-3p protects hippocampal neuron against pyroptosis to attenuate sevoflurane inhalation-induced post-operative cognitive dysfunction in rats via activation of HTR2A/ERK/Nrf2 axis by targeting DNMT1. Cell Death Discov. 2022;8(1):290.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  65. Zuo Y, Yin L, Cheng X, Li J, Wu H, Liu X, et al. Elamipretide attenuates pyroptosis and perioperative neurocognitive disorders in aged mice. Front Cell Neurosci. 2020;14:251.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  66. Chen Y, Wei G, Zhang L, Feng X, Lei E. Dexmedetomidine enhances mitophagy via PINK1 to alleviate hippocampal neuronal pyroptosis and improve postoperative cognitive dysfunction in elderly rat. Exp Neurol. 2024;379: 114842.

    Article  PubMed  CAS  Google Scholar 

  67. Wang Y, Gao W, Shi X, Ding J, Liu W, He H, et al. Chemotherapy drugs induce pyroptosis through caspase-3 cleavage of a gasdermin. Nature. 2017;547(7661):99–103.

    Article  PubMed  CAS  Google Scholar 

  68. Wang W, Zhao B, Gao W, Song W, Hou J, Zhang L, et al. Inhibition of PINK1-mediated mitophagy contributes to postoperative cognitive dysfunction through activation of caspase-3/GSDME-dependent pyroptosis. ACS Chem Neurosci. 2023;14(7):1249–60.

    Article  PubMed  CAS  Google Scholar 

  69. Cunningham EL, McGuinness B, McAuley DF, Toombs J, Mawhinney T, O’Brien S, et al. CSF Beta-amyloid 1–42 concentration predicts delirium following elective arthroplasty surgery in an observational cohort study. Ann Surg. 2019;269(6):1200–5.

    Article  PubMed  Google Scholar 

  70. Ballweg T, White M, Parker M, Casey C, Bo A, Farahbakhsh Z, et al. Association between plasma tau and postoperative delirium incidence and severity: a prospective observational study. Br J Anaesth. 2021;126(2):458–66.

    Article  PubMed  CAS  Google Scholar 

  71. Fong TG, Vasunilashorn SM, Gou Y, Libermann TA, Dillon S, Schmitt E, et al. Association of CSF Alzheimer’s disease biomarkers with postoperative delirium in older adults. Alzheimers Dement (N Y). 2021;7(1): e12125.

    Article  PubMed  Google Scholar 

  72. Parker M, White M, Casey C, Kunkel D, Bo A, Blennow K, et al. Cohort analysis of the association of delirium severity with cerebrospinal fluid amyloid-tau-neurodegeneration pathologies. J Gerontol A Biol Sci Med Sci. 2022;77(3):494–501.

    Article  PubMed  CAS  Google Scholar 

  73. Leung JM, Rojas JC, Tang C, Chan B, Lario-Lago A, Boxer AL, et al. The presence of preoperative neurodegeneration biofluid markers in patients with postoperative delirium. Anesthesiology. 2023. https://doiorg.publicaciones.saludcastillayleon.es/10.1097/ALN.0000000000004666.

    Article  PubMed  Google Scholar 

  74. Tang JX, Baranov D, Hammond M, Shaw LM, Eckenhoff MF, Eckenhoff RG. Human Alzheimer and inflammation biomarkers after anesthesia and surgery. Anesthesiology. 2011;115(4):727–32.

    Article  PubMed  CAS  Google Scholar 

  75. Zhang B, Tian M, Zheng H, Zhen Y, Yue Y, Li T, et al. Effects of anesthetic isoflurane and desflurane on human cerebrospinal fluid Aβ and τ level. Anesthesiology. 2013;119(1):52–60.

    Article  PubMed  CAS  Google Scholar 

  76. Parra Bravo C, Naguib SA, Gan L. Cellular and pathological functions of tau. Nat Rev Mol Cell Biol. 2024. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41580-024-00753-9.

    Article  PubMed  Google Scholar 

  77. Grundke-Iqbal I, Iqbal K, Tung YC, Quinlan M, Wisniewski HM, Binder LI. Abnormal phosphorylation of the microtubule-associated protein tau (tau) in Alzheimer cytoskeletal pathology. Proc Natl Acad Sci U S A. 1986;83(13):4913–7.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  78. Gao Y, Tan L, Yu JT, Tan L. Tau in Alzheimer’s disease: mechanisms and therapeutic strategies. Curr Alzheimer Res. 2018;15(3):283–300.

    Article  PubMed  CAS  Google Scholar 

  79. Huang C, Irwin MG, Wong G, Chang R. Evidence of the impact of systemic inflammation on neuroinflammation from a non-bacterial endotoxin animal model. J Neuroinflamm. 2018;15(1):147.

    Article  Google Scholar 

  80. Cummins N, Tweedie A, Zuryn S, Bertran-Gonzalez J, Götz J. Disease-associated tau impairs mitophagy by inhibiting Parkin translocation to mitochondria. EMBO J. 2019;38(3): e99360.

    Article  PubMed  Google Scholar 

  81. Bardai FH, Ordonez DG, Bailey RM, Hamm M, Lewis J, Feany MB. Lrrk promotes tau neurotoxicity through dysregulation of actin and mitochondrial dynamics. PLoS Biol. 2018;16(12): e2006265.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  82. Li M, Chen L, Lee DH, Yu LC, Zhang Y. The role of intracellular amyloid beta in Alzheimer’s disease. Prog Neurobiol. 2007;83(3):131–9.

    Article  PubMed  CAS  Google Scholar 

  83. Suire CN, Leissring MA. Cathepsin D: a candidate link between amyloid β-protein and tauopathy in Alzheimer disease. J Exp Neurol. 2021;2(1):10–5.

    PubMed  PubMed Central  Google Scholar 

  84. 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  PubMed  PubMed Central  CAS  Google Scholar 

  85. Li J, Kim SG, Blenis J. Rapamycin: one drug, many effects. Cell Metab. 2014;19(3):373–9.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  86. Liu GY, Sabatini DM. mTOR at the nexus of nutrition, growth, ageing and disease. Nat Rev Mol Cell Biol. 2020;21(4):183–203.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  87. Zhang X, Zhou Y, Xu M, Chen G. Autophagy is involved in the sevoflurane anesthesia-induced cognitive dysfunction of aged rats. PLoS ONE. 2016;11(4): e0153505.

    Article  PubMed  PubMed Central  Google Scholar 

  88. Li Y, Li Y, Chen L, Li Y, Liu K, Hong J, et al. Reciprocal interaction between mitochondrial fission and mitophagy in postoperative delayed neurocognitive recovery in aged rats. CNS Neurosci Ther. 2023;29(11):3322–38.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  89. Li X, Yuan Z, Wang Y, Wang W, Shi J. Recent advances of honokiol: pharmacological activities, manmade derivatives and structure–activity relationship. Eur J Med Chem. 2024;272: 116471.

    Article  PubMed  CAS  Google Scholar 

  90. Talarek S, Listos J, Barreca D, Tellone E, Sureda A, Nabavi SF, et al. Neuroprotective effects of honokiol: from chemistry to medicine. BioFactors. 2017;43(6):760–9.

    Article  PubMed  CAS  Google Scholar 

  91. Li W, Wang S, Zhang H, Li B, Xu L, Li Y, et al. Honokiol restores microglial phagocytosis by reversing metabolic reprogramming. J Alzheimers Dis. 2021;82(4):1475–85.

    Article  PubMed  CAS  Google Scholar 

  92. Faessel HM, Obach RS, Rollema H, Ravva P, Williams KE, Burstein AH. A review of the clinical pharmacokinetics and pharmacodynamics of varenicline for smoking cessation. Clin Pharmacokinet. 2010;49(12):799–816.

    Article  PubMed  CAS  Google Scholar 

  93. Yu R, Tae HS, Xu Q, Craik DJ, Adams DJ, Jiang T, et al. Molecular dynamics simulations of dihydro-β-erythroidine bound to the human α4β2 nicotinic acetylcholine receptor. Br J Pharmacol. 2019;176(15):2750–63.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  94. Zanardi A, Leo G, Biagini G, Zoli M. Nicotine and neurodegeneration in ageing. Toxicol Lett. 2002;127(1–3):207–15.

    Article  PubMed  CAS  Google Scholar 

  95. Tata AM, Velluto L, D’Angelo C, Reale M. Cholinergic system dysfunction and neurodegenerative diseases: cause or effect. CNS Neurol Disord Drug Targets. 2014;13(7):1294–303.

    Article  PubMed  CAS  Google Scholar 

  96. Huang C, Chu JM, Liu Y, Chang RC, Wong GT. Varenicline reduces DNA damage, tau mislocalization and post surgical cognitive impairment in aged mice. Neuropharmacology. 2018;143:217–27.

    Article  PubMed  CAS  Google Scholar 

  97. Slomski A. Liraglutide may reduce diabetic kidney disease. JAMA. 2017;318(16):1532.

    PubMed  Google Scholar 

  98. Hanssen R, Rigoux L, Kuzmanovic B, Iglesias S, Kretschmer AC, Schlamann M, et al. Liraglutide restores impaired associative learning in individuals with obesity. Nat Metab. 2023;5(8):1352–63.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  99. Han WN, Hölscher C, Yuan L, Yang W, Wang XH, Wu MN, et al. Liraglutide protects against amyloid-β protein-induced impairment of spatial learning and memory in rats. Neurobiol Aging. 2013;34(2):576–88.

    Article  PubMed  CAS  Google Scholar 

  100. Chen S, Sun J, Zhao G, Guo A, Chen Y, Fu R, et al. Liraglutide improves water maze learning and memory performance while reduces hyperphosphorylation of tau and neurofilaments in APP/PS1/Tau triple transgenic mice. Neurochem Res. 2017;42(8):2326–35.

    Article  PubMed  CAS  Google Scholar 

  101. Liu W, Wang Z, Wang W, Wang Z, Xing Y, Hölscher C. Liraglutide reduces alcohol consumption, anxiety, memory impairment, and synapse loss in alcohol dependent mice. Neurochem Res. 2024;49(4):1061–75.

    Article  PubMed  CAS  Google Scholar 

  102. Femminella GD, Frangou E, Love SB, Busza G, Holmes C, Ritchie C, et al. Evaluating the effects of the novel GLP-1 analogue liraglutide in Alzheimer’s disease: study protocol for a randomised controlled trial (ELAD study). Trials. 2019;20(1):191.

    Article  PubMed  PubMed Central  Google Scholar 

  103. Reddy PH, Manczak M, Kandimalla R. Mitochondria-targeted small molecule SS31: a potential candidate for the treatment of Alzheimer’s disease. Hum Mol Genet. 2017;26(8):1483–96.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  104. Wu J, Hao S, Sun XR, Zhang H, Li H, Zhao H, et al. Elamipretide (SS-31) ameliorates isoflurane-induced long-term impairments of mitochondrial morphogenesis and cognition in developing rats. Front Cell Neurosci. 2017;11:119.

    Article  PubMed  PubMed Central  Google Scholar 

  105. Zhao W, Xu Z, Cao J, Fu Q, Wu Y, Zhang X, et al. Elamipretide (SS-31) improves mitochondrial dysfunction, synaptic and memory impairment induced by lipopolysaccharide in mice. J Neuroinflamm. 2019;16(1):230.

    Article  CAS  Google Scholar 

  106. Du X, Zeng Q, Luo Y, He L, Zhao Y, Li N, et al. Application research of novel peptide mitochondrial-targeted antioxidant SS-31 in mitigating mitochondrial dysfunction. Mitochondrion. 2024;75: 101846.

    Article  PubMed  CAS  Google Scholar 

  107. Wu J, Zhang M, Li H, Sun X, Hao S, Ji M, et al. BDNF pathway is involved in the protective effects of SS-31 on isoflurane-induced cognitive deficits in aging mice. Behav Brain Res. 2016;305:115–21.

    Article  PubMed  CAS  Google Scholar 

  108. Ji Y, Ma Y, Ma Y, Wang Y, Zhao X, Jin D, et al. SS-31 inhibits mtDNA-cGAS-STING signaling to improve POCD by activating mitophagy in aged mice. Inflamm Res. 2024;73(4):641–54.

    Article  PubMed  CAS  Google Scholar 

  109. Mahmoud M, Mason KP. Dexmedetomidine: review, update, and future considerations of paediatric perioperative and periprocedural applications and limitations. Br J Anaesth. 2015;115(2):171–82.

    Article  PubMed  CAS  Google Scholar 

  110. van Norden J, Spies CD, Borchers F, Mertens M, Kurth J, Heidgen J, et al. The effect of peri-operative dexmedetomidine on the incidence of postoperative delirium in cardiac and non-cardiac surgical patients: a randomised, double-blind placebo-controlled trial. Anaesthesia. 2021;76(10):1342–51.

    Article  PubMed  Google Scholar 

  111. Chen C, Chen Y, Liu T, Song D, Ma D, Cheng O. Dexmedetomidine can enhance PINK1/parkin-mediated mitophagy in MPTP-induced PD mice model by activating AMPK. Oxid Med Cell Longev. 2022;2022:7511393.

    PubMed  PubMed Central  Google Scholar 

  112. Suo L, Wang M. Dexmedetomidine alleviates sevoflurane-induced neurotoxicity via mitophagy signaling. Mol Biol Rep. 2020;47(10):7893–901.

    Article  PubMed  CAS  Google Scholar 

  113. Song XX, Yu BW. Anesthetic effects of propofol in the healthy human brain: functional imaging evidence. J Anesth. 2015;29(2):279–88.

    Article  PubMed  Google Scholar 

  114. Dai W, Xiao Y, Tu Y, Xiao F, Lu Y, Qin Y, et al. Propofol protects hippocampal neurons in sleep-deprived rats by inhibiting mitophagy and autophagy. Ann Transl Med. 2021;9(18):1427.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  115. Liang C, Du F, Cang J, Xue Z. Pink1 attenuates propofol-induced apoptosis and oxidative stress in developing neurons. J Anesth. 2018;32(1):62–9.

    Article  PubMed  Google Scholar 

  116. Han D, Jin J, Fang H, Xu G. Long-term action of propofol on cognitive function and hippocampal neuroapoptosis in neonatal rats. Int J Clin Exp Med. 2015;8(7):10696–704.

    PubMed  PubMed Central  CAS  Google Scholar 

  117. Lin JG, Chen CJ, Yang HB, Chen YH, Hung SY. Electroacupuncture promotes recovery of motor function and reduces dopaminergic neuron degeneration in rodent models of Parkinson’s disease. Int J Mol Sci. 2017;18(9):1846.

    Article  PubMed  PubMed Central  Google Scholar 

  118. Ho YS, Zhao FY, Yeung WF, Wong GT, Zhang HQ, Chang RC. Application of acupuncture to attenuate immune responses and oxidative stress in postoperative cognitive dysfunction: what do we know so far. Oxid Med Cell Longev. 2020;2020:9641904.

    Article  PubMed  PubMed Central  Google Scholar 

  119. Zhao W, Zou W. Effects of electroacupuncture on postoperative cognitive dysfunction and its underlying mechanisms: a literature review of rodent studies. Front Aging Neurosci. 2024;16:1384075.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  120. Wang H, Chen S, Zhang Y, Xu H, Sun H. Electroacupuncture ameliorates neuronal injury by Pink1/Parkin-mediated mitophagy clearance in cerebral ischemia-reperfusion. Nitric Oxide. 2019;91:23–34.

    Article  PubMed  Google Scholar 

  121. Guo J, Niu K, Ma BF, Sun LN, Fang QW, An JX. Electroacupuncture ameliorates surgery-induced spatial memory deficits by promoting mitophagy in rats. Ann Transl Med. 2023;11(2):74.

    Article  PubMed  PubMed Central  Google Scholar 

  122. Mondini S, Madella I, Zangrossi A, Bigolin A, Tomasi C, Michieletto M, et al. Cognitive reserve in dementia: implications for cognitive training. Front Aging Neurosci. 2016;8:84.

    Article  PubMed  PubMed Central  Google Scholar 

  123. O’Gara BP, Mueller A, Gasangwa D, Patxot M, Shaefi S, Khabbaz K, et al. Prevention of early postoperative decline: a randomized, controlled feasibility trial of perioperative cognitive training. Anesth Analg. 2020;130(3):586–95.

    Article  PubMed  PubMed Central  Google Scholar 

  124. McNab F, Varrone A, Farde L, Jucaite A, Bystritsky P, Forssberg H, et al. Changes in cortical dopamine D1 receptor binding associated with cognitive training. Science. 2009;323(5915):800–2.

    Article  PubMed  CAS  Google Scholar 

  125. Petrelli A, Kaesberg S, Barbe MT, Timmermann L, Rosen JB, Fink GR, et al. Cognitive training in Parkinson’s disease reduces cognitive decline in the long term. Eur J Neurol. 2015;22(4):640–7.

    Article  PubMed  CAS  Google Scholar 

  126. Feinkohl I, Winterer G, Spies CD, Pischon T. Cognitive reserve and the risk of postoperative cognitive dysfunction. Dtsch Arztebl Int. 2017;114(7):110–7.

    PubMed  PubMed Central  Google Scholar 

  127. Billings LM, Green KN, McGaugh JL, LaFerla FM. Learning decreases A beta*56 and tau pathology and ameliorates behavioral decline in 3xTg-AD mice. J Neurosci. 2007;27(4):751–61.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  128. Ji Z, Feng T, Wang H. The effects of 12-week physical exercise tapping high-level cognitive functions. Adv Cogn Psychol. 2020;16(1):59–66.

    Article  PubMed  PubMed Central  Google Scholar 

  129. Lee SS, Lo Y, Verghese J. Physical activity and risk of postoperative delirium. J Am Geriatr Soc. 2019;67(11):2260–6.

    Article  PubMed  PubMed Central  Google Scholar 

  130. Trubnikova OA, Tarasova IV, Moskin EG, Kupriyanova DS, Argunova YA, Pomeshkina SA, et al. Beneficial effects of a short course of physical prehabilitation on neurophysiological functioning and neurovascular biomarkers in patients undergoing coronary artery bypass grafting. Front Aging Neurosci. 2021;13: 699259.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  131. Erickson KI, Voss MW, Prakash RS, Basak C, Szabo A, Chaddock L, et al. Exercise training increases size of hippocampus and improves memory. Proc Natl Acad Sci U S A. 2011;108(7):3017–22.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  132. Hu J, Huang B, Chen K. The impact of physical exercise on neuroinflammation mechanism in Alzheimer’s disease. Front Aging Neurosci. 2024;16:1444716.

    Article  PubMed  PubMed Central  Google Scholar 

  133. Cui K, Li C, Fang G. Aerobic exercise delays Alzheimer’s disease by regulating mitochondrial proteostasis in the cerebral cortex and hippocampus. Life (Basel). 2023;13(5):1204.

    PubMed  CAS  Google Scholar 

  134. Zhao N, Zhang X, Li B, Wang J, Zhang C, Xu B. Treadmill exercise improves PINK1/Parkin-Mediated mitophagy activity against Alzheimer’s disease pathologies by upregulated SIRT1-FOXO1/3 Axis in APP/PS1 mice. Mol Neurobiol. 2023;60(1):277–91.

    Article  PubMed  CAS  Google Scholar 

Download references

Funding

Chongqing Natural Science Foundation (General Program) in 2024 (CSTB2024NSCQ-MSX0633) 2024 School-level Postgraduate Supervisor Team Projects of the First Clinical College of Chongqing Medical University (CYYY-DSTDXM-202419).

Author information

Authors and Affiliations

Authors

Contributions

First author Zhen Feng wrote the main text of the manuscript, and co-first author Yan Hou made substantial contributions to its conception. The second author Chang Yu and the third author Ting Li acquired, analysed, and interpreted the data. Fourth author Haoyang Fu prepared Table 1. Corresponding author Feng Lv critically revised the manuscript for important intellectual content, and corresponding author Ping Ling approved the publication of the version. All authors revised the manuscript.

Corresponding authors

Correspondence to Feng Lv or Ping Li.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Competing interests

The authors declare 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-NonCommercial-NoDerivatives 4.0 International License, which permits any non-commercial use, sharing, 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 you modified the licensed material. You do not have permission under this licence to share adapted material derived from this article or parts of it. 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-nc-nd/4.0/.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Feng, Z., Hou, Y., Yu, C. et al. Mitophagy in perioperative neurocognitive disorder: mechanisms and therapeutic strategies. Eur J Med Res 30, 270 (2025). https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s40001-025-02400-1

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s40001-025-02400-1

Keywords