- 1School of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
- 2Guangyuan Hospital of Traditional Chinese Medicine, Guangyuan, Sichuan, China
- 3Chengdu First People's Hospital, Chengdu, Sichuan, China
Neurodegenerative diseases are a category of neurological conditions with high prevalence that pose major treatment challenges. Common pathologies involve protein accumulation and mitochondrial damage. Mitophagy maintains cellular homeostasis by removing defective mitochondria, which are associated with the pathogenesis of neurodegenerative diseases. Although the ubiquitin-dependent mitophagy mediated by the PINK1–Parkin pathway has been extensively studied, growing evidence indicates that receptor-mediated mitophagy plays a crucial compensatory role in neurons, particularly when the PINK1–Parkin pathway is impaired. This review focuses on the emerging field of receptor-mediated mitophagy, systematically elaborating its role as a key homeostatic mechanism operating independently of the canonical PINK1/Parkin pathway. It provides a focused analysis of the specific functions and activation mechanisms of key receptors—including BNIP3, NIX, FUNDC1, and AMBRA1—in models of Alzheimer’s disease, Parkinson’s disease, and amyotrophic lateral sclerosis. Furthermore, this review explores the clinical potential of targeting these specific receptors for precise intervention, aiming to provide a new theoretical foundation and direction for developing therapeutic strategies against neurodegenerative diseases.
1 Introduction
Neurodegenerative diseases (NDDs) are a group of disorders characterized by progressive loss of neuronal structure and function in the central nervous system. Pathological features typically include a sustained decrease in neurons, abnormal proliferation of glial cells, and activation of neuroinflammatory responses. As the disease progresses, patients generally experience significant motor dysfunction, cognitive decline, and sensory loss, which are progressive and irreversible (1). It is indisputable that aging, as a major risk factor for most NDDs, is because neurons in the brain are susceptible to aging-induced changes, highlighting one of the key factors in the aging process: mitochondrial function (2). Mitochondria act as the “energy factories” of the cell, producing Adenosine Triphosphate (ATP) through oxidative phosphorylation via the electron transport chain. This is the main energy source for the cell and is required by neurons, which have a higher energy demand (3). Different parts of a neuron have varying energy demands, which require a fully functional mitochondrial network to sustain their complex structures and functions—including synaptic transmission, long-distance axonal transport, and mitochondrial fusion and fission (4). Therefore, any factor that disrupts the balance of the mitochondrial quality control system can trigger neuronal damage or even cell death.
Mitophagy plays a critical role in maintaining neuronal health among the various mitochondrial quality control mechanisms. The focus in this field has traditionally been on the canonical ubiquitin-dependent pathway mediated by PINK1 (PTEN-induced putative kinase 1) and Parkin (Parkinson protein 2). This pathway is collectively recognized as the central mechanism for responding to mitochondrial damage, and its dysfunction is well-established in the pathogenesis of numerous neurodegenerative diseases (5).
However, it has been demonstrated that there exists a form of “receptor-mediated mitophagy” pathways that are not dependent on PINK1-Parkin in the context of pathologies in which PINK1 or Parkin expression is absent or functionally impaired, as well as in neurodegenerative disorders more generally (6). These pathways are directly triggered by specific receptor proteins, which recruit autophagy components with greater directness and superior efficiency. While existing literature has extensively described the general mechanisms of mitophagy, comprehensive summaries of receptor-mediated mitophagy remain inadequate, often relegating it to a secondary or complementary role. This review systematically summarizes recent advances in the role of receptor-mediated mitophagy in neurodegenerative diseases. We will emphasize that this autophagic pathway is not merely a simple backup for the canonical pathway but serves as a crucial and independent clearance mechanism under specific conditions. We aim to provide a novel perspective for understanding NDDs pathogenesis and to establish a theoretical foundation for developing precise therapies that target specific receptor pathways.
2 Quality control of mitochondria
Different endogenous and exogenous stimuli may cause mitochondrial dysfunction, and aggregation of abnormal mitochondria in the neurons can lead to imbalances in energy metabolism, impaired signaling, dysregulation of calcium homeostasis, activation of neuroinflammation, and other pathologies. Fortunately, mitochondria are self-regulating organelles that can sense abnormal states within their bodies and regulate their stabilization through a variety of regulatory mechanisms, including mitophagy, mitochondrial fission and fusion, mitochondria-derived vesicles, proteasomal degradation, and exosome release to remove damaged proteins and mitochondria (7–9). Among these mechanisms, mitophagy has received particular attention in recent years. Mitophagy is a subtype of macroautophagy that selectively removes damaged mitochondria via lysosomal degradation. Mitophagy is initiated when other quality control mechanisms have limited processing capacity or when mitochondria need to be removed due to cellular metabolic demands. Mitophagy plays an important role in normal cellular function and influences disease development, and the molecular mechanisms involved in mitophagy have been characterized in a variety of diseases, including aging (10), neurodegenerative diseases (11), and cancer (12). In neurodegenerative diseases, defective mitophagy is thought to be responsible for the accumulation of damaged mitochondria in neurons and leads to neuronal dysfunction and even death, and thus, mitophagy is being explored as a therapeutic target for neurodegenerative diseases (Figure 1).
Figure 1. The relationship between mitochondria and neurodegenerative diseases (created with BioRender.com).
3 Mitophagy pathways
Mitophagy was first proposed by Lemasters in 2005 (13). With advancing research in this field, the regulatory mechanisms of mitophagy have been increasingly elucidated. The mitophagy pathways mainly consist of ubiquitin (Ub)-dependent pathways and Ub-independent pathways (14). The ubiquitin-dependent pathway includes not only the classical PINK1-Parkin mediated mitophagy but also mitophagy mediated by other E3 ubiquitin ligases such as Siah E3 ubiquitin protein ligase 1 (SIAH1) (15), mitochondrial E3 ubiquitin protein ligase 1 (MUL1) (16), HECT, UBA and WWE domain containing E3 ubiquitin protein ligase 1 (HUWE1) (17), and Glycoprotein 78 (GP78), which promote mitophagy by ubiquitinating damaged mitochondrial proteins (18). Ub-independent pathways can be further categorized into receptor-mediated and lipid-mediated mitophagy. The PINK1-Parkin mitophagy pathway, also called classical mitophagy, is the most widely studied mitochondrial quality control way. Its core mechanism relies on the synergistic action of PINK1 and the E3 ubiquitin ligase Parkin. PINK1, a mitochondria-targeted serine/threonine kinase encoded by the PARK6 gene, triggers subsequent Parkin-dependent light chain 3 (LC3) recruitment (19). Under physiological conditions, PINK1 maintains low protein levels through continuous translocation to the inner mitochondrial membrane (IMM) via the TOM/TIM translocase complex, followed by proteolytic processing and degradation (20). When mitochondria are damaged, PINK1 is unable to translocate to IMM and is activated at the outer mitochondrial membrane (OMM) (21). Accumulated PINK1 recruits and phosphorylates Parkin, which transforms from a self-repressing dormant enzyme to an activated E3 ubiquitin ligase through a series of structural remodeling cascade reactions to ubiquitinate many OMM proteins, such as mitofusins 1/2 (MFN1/2). These ubiquitinated substrates undergo further PINK1-dependent phosphorylation, creating feedforward amplification that recruits additional Parkin to mitochondria. This cascade enhances ubiquitin chain formation and recruits autophagy adaptors. Autophagy adaptors are a class of proteins characterized by the presence of an LC3-interacting regions (LIRs), which mainly includes sequestosome 1(P62) (22), neighbor of BRCA1 gene 1 (NBR1) (23), nuclear dot protein 52 (NDP52), human T-cell leukemia virus type I binding protein 1 (TAX1BP1) (24) and optineurin (OPTN). Next, these adaptors bind to LC3 on autophagophore membranes via LIRs, facilitating engulfment of depolarized mitochondria into autophagosomes. The completed autophagosomes are ultimately degraded by lysosomes (14). Notably, TANK-binding kinase 1 (TBK1) phosphorylates OPTN in this process, serving to amplify the signal and thereby enhance mitophagy (25). (Figure 2A). In addition to ubiquitin-dependent mitophagy, specific mitochondrial proteins act as mitophagy receptors. These receptors directly target dysfunctional mitochondria to the phagophore for degradation via protein–protein interactions, bypassing ubiquitination. Key examples include BCL2-interacting protein 3 (BNIP3), NIP3-like protein X (NIX/BNIP3L), FUN14 domain-containing protein 1 (FUNDC1), as well as Autophagy/Beclin-1 Regulator-1 (AMBRA1), among others. All these receptors contain a conserved LC3-interacting region (LIR) motif that directly binds to Atg8-family proteins (e.g., LC3) on the phagophore membrane, thereby recruiting damaged mitochondria to nascent autophagosomes (26) (Figure 2B). Critically, these receptors operate independently of both PINK1-Parkin recruitment and ubiquitination, they have been considered as potential targets for neurodegenerative diseases. While the mechanisms underlying general mitophagy are well characterized, how these receptor-mediated pathways achieve spatiotemporal coordination with parallel pathways requires further investigation.
Figure 2. Overview of the mitophagy pathway. (A) PINK1-Parkin-mediated mitophagy. Under normal conditions, newly synthesized PINK1 translcates into IMM via the TOM/TIM complexes. PARL cleaves PINK1 at its transmembrane domains, and the cleaved PINK1 is subsequently degraded rapidly by the proteasome, and PINK1 maintains low protein levels. When mitochondria are damaged, PINK1 is unable to translocate to the IMM and is activated at the OMM. Accumulated PINK1 recruits and phosphorylates Parkin, which transforms to an activated E3 ubiquitin ligase through a series of structural remodeling cascade reactions to ubiquitinate many OMM proteins. These ubiquitinated substrates undergo further PINK1-dependent phosphorylation, creating feedforward amplification that recruits additional Parkin to mitochondria. This cascade enhances ubiquitin chain formation and recruits autophagy adaptors, leading to mitophagy. (B) Receptor-mediated mitophagy. Mitophagy receptors such as BNIP3, NIX, FUNDC1, FTMT, AMBRA1, MCL-1, BCL2L13, FKBP8, PHB2, NLRX1, and SAMM50 can directly bind to LC3 and GABARAP-family members at the phagophore membrane (created with BioRender.com).
4 Receptor-mediated mitophagy
4.1 Hypoxia-dependent receptors
These receptors are characterized by their core role in hypoxia-activated mitophagy, directly sensing low oxygen to trigger the process via varied mechanisms for metabolic adaptation (Figure 3). Their activation strategies, though diverse, ultimately converge on a parallel regulatory approach at both the transcriptional and translational levels. This integrated strategy ensures that cells can effectively launch mitochondrial quality control and sustain metabolic stability in response to both acute and prolonged oxygen deprivation.
Figure 3. Schematic diagram of mitophagy receptors classification (created with BioRender.com).
4.1.1 BNIP3 and NIX
BCL2/adenovirus E1B 19-kDa interacting protein 3 (BNIP3) belongs to the BH3-only subfamily of the Bcl-2 protein family, and it regulates the permeability state of OMM. The structure of BNIP3 comprises a large complex N-terminal region. This N-terminal region contains a WXXL motif, which serves as its LIR. Via this motif, BNIP3 binds to LC3 and induces mitophagy (Figure 4) (27).
Figure 4. Schematic representation of the domains of mitophagy receptors (created with BioRender.com).
NIX is a multifunctional BH3-only protein of the Bcl-2 family localized to the OMM. It shares 65% identity with BNIP3 in amino acid sequence (28, 29). The LIR motif of NIX consists of four amino acid motifs that are essential for interaction with LC3 and GABA type A receptor-associated protein (GABARAP), and deletion of the LIR motifs in NIX leads to failure of mitophagy (Figure 4) (30). The role of NIX in mitophagy was initially identified in Nix-knockout mice, which exhibit anemia due to impaired mitophagy, resulting in dysregulated reticulocyte maturation and massive mitochondrial accumulation (31, 32).
Upon the onset of hypoxia, BNIP3 and NIX dimerize and anchor to the OMM through their TM domains; BNIP3 homodimerization is critical for mitophagy, and failure of homodimer formation impairs this process (33). Studies show that besides binding LC3 to induce mitophagy, these receptors enhance Parkin-mediated mitophagy and functionally compensate for Parkin deficiency. Specifically, BNIP3 interacts with PINK1 to inhibit the protein degradation of PINK1, which then participates in ubiquitin-dependent mitophagy by promoting the translocation of Parkin to mitochondria. Moreover, NIX is a Parkin substrate; when ubiquitinated by Parkin, NIX recruits the selective autophagy adaptor NBR1 and LC3/GABARAP to mitochondria to initiate perimitochondrial phagophore assembly (34). NIX also contributes to Parkin accumulation on damaged mitochondria (35). Furthermore, it has been shown that mitophagy clearance in axonal mitochondria requires NIX but not Parkin. This study supports different mechanisms between NIX and Parkin (36).
Studies primarily focus on hypoxia-inducible factor 1-alpha (HIF-1α) activating BNIP3 transcription during hypoxia, upregulating its expression. Consequently, BNIP3 and NIX are classified as hypoxia-specific mitophagy factors. Besides, some other regulators, such as transcription factors, growth factors, or cellular stresses, are also involved in NIX regulation, while BNIP3 and NIX expression may also be regulated by some microRNAs (35). Phosphorylation represents the most studied mechanism underlying BNIP3/NIX-mediated mitophagy. BNIP3 phosphorylation at Ser17/24 enhances LC3B affinity and increases autophagosome recruitment to mitochondria. c-Jun N-terminal kinase 1/2 (JNK1/2) phosphorylates BNIP3 at Ser60/Thr66 under hypoxia, stabilizing BNIP3 and facilitating direct LC3 binding to drive mitophagy. However, severe hypoxia inactivates JNK1/2, inducing BNIP3 dephosphorylation and suppressing mitophagy (37). NIX phosphorylation at Ser34/35 activates mitophagy via its LIR motif, whereas Ser212 phosphorylation inhibits dimerization, suggesting that phosphorylation of different serine residues would have opposite results. Ubiquitination provides another regulatory mechanism. Ubiquitinated NIX recruits adaptor NBR1 in the autophagosomes for mitophagy (34). Recently, the mitochondrial outer membrane protein FBXL4 has been identified, which recognizes and ubiquitinates BNIP3 and NIX, thereby promoting their degradation and suppressing mitophagy (38). Meanwhile, PPTC7 plays a key role as a connector molecule in assembling the BNIP3/NIX-PPTC7-SCFFBXL4 complex to degrade BNIP3 and NIX and form a homeostatic regulatory loop, which helps to maintain mitochondrial number and function, and prevents cellular energy disruption from excessive mitophagy (39). It is clear that hypoxia is not the only inducer of activation of this pathway, but the specific mechanisms by which BNIP3 and NIX regulate mitophagy have not been fully elucidated and require further follow-up studies.
4.1.2 FUNDC1
FUNDC1 is an OMM-localized mitophagy receptor protein that belongs to the paralogous subfamily of FUN14 domain-containing proteins (40). Its structure comprises a cytoplasmic N-terminus, a C-terminal region, and an OMM transmembrane region, where the N-terminus is exposed to the cytosol and the C-terminus extends into the intermembrane space. The N-terminus contains a typical LIR motif: the Y(18)xxL (21) sequence, which interacts with Atg8 family proteins to induce mitophagy (Figure 4) (41). It has been reported that during hypoxia, FUNDC1 is subject to transcriptional-level gene upregulation to enhance mitophagy (42). The regulation, however, is largely governed by rapid post-translational modifications. Under hypoxic stress, FUNDC1 undergoes dephosphorylation at Ser13 and Tyr18 sites, activating its mediated mitophagy. A key representative regulator is the PGAM family member 5 (PGAM5), which promotes dephosphorylation of FUNDC1 at Ser13 in its multimeric form, thereby serving as a positive regulator of mitophagy (43). Meanwhile, during energy depletion, the mitophagy initiation molecule unc-51-like autophagy activating kinase 1 (ULK1) is activated and recruited to mitochondria, phosphorylates FUNDC1 at Ser17, and promotes mitophagy (44, 45). Studies indicate that there may be crosstalk between ubiquitin-dependent autophagy and mitophagy, and that Membrane-associated ring-CH-Type finger 5 (MARCH5), an E3 ubiquitin ligase, is an important regulator of FUNDC1-mediated mitophagy. During early hypoxia, MARCH5 homo-oligomers dissociate into monomers; subsequently, MARCH5 binds Lys119 of FUNDC1, promoting FUNDC1 ubiquitination and degradation to suppress mitophagy and prevent inappropriate clearance of undamaged mitochondria (46). Whether FUNDC1 has additional modifications remains to be further investigated, and how multiple modifications can be coordinated to regulate the state of FUNDC1 will be the focus of future studies.
4.1.3 FTMT
Mitochondrial ferritin (FTMT) is important for intracellular iron homeostasis, antioxidant defense, inflammatory regulation, and multiple pathological processes. This protein is primarily localized to mitochondria, but under stress conditions such as hypoxia or inflammation, it can be cleaved by thrombin and translocate to the cytoplasm, demonstrating dynamic subcellular distribution (47). Current research indicates that FTMT’s involvement in mitophagy is closely linked to its role in regulating HIF-1α stability and its interaction with nuclear receptor coactivator 4 (NCOA4). Under hypoxic conditions, HIF-1α upregulates FTMT expression, FTMT interacts with NCOA4, and increases the co-localization of FTMT with LC3, thereby promoting mitophagy (48, 49). In addition, the expression of NCOA4 was significantly reduced under hypoxic conditions. This reduction led to the restriction of the ferritin degradation pathway, increasing the accumulation of FTMT, and the excess free iron catalyzed the ROS generation, causing mitochondrial oxidative damage and triggering mitophagy (50). The mechanism by which FTMT mediates mitophagy is not fully understood, and in particular, how changes in its localization in different cell types and stress contexts affect mitophagy initiation remain controversial. For example, the translocation of FTMT to the cytoplasm after thrombin cleavage under hypoxic conditions may alter its original autophagy-related functions.
4.2 Energy stress receptors
The activity of these receptors is regulated by energy stress pathways and primarily responds to cellular energy metabolic imbalances. Under stress conditions, their activity is significantly enhanced, thereby promoting large-scale clearance of severely damaged mitochondria and preventing cellular apoptosis.
4.2.1 AMBRA1
Autophagy and Beclin1 Regulator1 (AMBRA1) is a multifunctional scaffolding protein. It contains three and a half WD40 repeat sequences at both its N-terminal and C-terminal, which are joined and folded into a β-propeller structure. Between the two WD40 structural domains, there are 650 amino acids, as well as 250 amino acids at the end of the protein, constituting an intrinsically disordered region (51). This region is essential for AMBRA1 regulation. The LIR region is located at its C-terminus (Figure 4). In the PINK1-Parkin independent pathway, AMBRA1 binds to LC3B via its LIR motif and mediates the wrapping of mitochondria by autophagosomes. Its LIR motif activity is regulated by CHUK/IKKα kinase phosphorylation (17). A variety of post-translational modifications and signaling pathways regulate AMBRA1 activity. In the resting state, its activity is suppressed by phosphorylation of the Ser52 site by MTOR or by binding to BCL2 (52). Under stress conditions, ULK1 phosphorylates AMBRA1 and translocates to the mitochondria-associated ER membrane (MAM) to activate mitophagy (53). In addition, AMBRA1 is recruited into the OMM during mitochondrial damage, prevents the translocation of PINK1 to the mitochondrial matrix, and aggregates at the OMM by binding to the TOM complex and ATAD3A, and enhances its stability, preventing it from being degraded by mitochondrial proteases, which in turn initiates the Parkin-mediated mitochondrial ubiquitination (54). Recent studies have shown that AMBRA1 stability is regulated by CRL4-mediated ubiquitination degradation as a means of limiting excessive mitophagy, but the exact regulatory mechanism is unclear.
4.2.2 BCL2L13
Bcl2 like 13 (BCL2L13) is a mitochondrial-localized protein belonging to the Bcl-2 protein family. Discovered two decades ago as a functional homolog of yeast’s classical mitophagy receptor Atg32, it anchors to the mitochondrial outer membrane and is expressed in various cells and tissues. BCL2L13 contains four BH domains (BH1-4) and a BHNo region, followed by a transmembrane (TM) domain (55). Uniquely, both the BH and BHNo regions contain a WXXL/I motif (LIR1/2), yet mutation of the motif exclusively in the BHNo region impairs its mitophagic function, indicating that this WXXL/I motif (LIR2, residues 273–276) serves as the LC3-interacting domain (Figure 4). BCL2L13, as a Parkin-independent mitophagy receptor, mediates mitophagy via its unique LIR2 motif, which interacts with LC3, while its mitophagic activity is regulated by Ser272 phosphorylation. PRKAA2/AMPKα2 phosphorylates BCL2L13 at Ser272 (56). In addition, Studies demonstrate that the LIR domain in human BCL2L13 preferentially binds LC3C, GABARAP, and GABARAP-L1 through the ULK1 complex, indicating BCL2L13 exhibits selectivity in mitophagy mediation (57). The physiological significance of this selective binding and the differences in the efficiency and specificity of mitophagy resulting from it in different cell types or tissues are important directions for future research. Despite the functional similarities between BCL2L13 and receptors such as BNIP3, its unique structural basis (e.g., intact BH1-4 domain) and the property of not forming heterodimers with other classical BCL2 family members make it play an irreplaceable role in the regulation of mitochondrial homeostasis, and make it a highly promising target for disease intervention.
4.2.3 MCL-1
Myeloid cell leukemia-1 (MCL-1) is a multifunctional cellular regulatory protein localized to the OMM, notable within the Bcl-2 family for its significant anti-apoptotic activity. MCL-1, like BCL-2, is an anti-apoptotic member of this family and regulates cell viability by maintaining OMM integrity. Early work by Hollville et al. demonstrated that MCL-1 prevents the degradation of damaged mitochondria by suppressing Parkin translocation to OMM (58). Follow-up studies revealed that MCL-1 acts as a mitophagy receptor in a PINK1-Parkin independent pathway, which interacts with LC3A and GABARAP on autophagosomes via the LIR motif to drive mitophagy. Notably, the MCL-1 C-terminus contains three LIR motifs (Figure 4) (59). Co-localization of mitochondria with LC3 is significantly reduced only when these three motifs are simultaneously mutated, but the relative contribution of each LIR under physiological conditions and its regulatory mechanism remain unclear. Moreover, Moyzis et al. demonstrate that MCL-1 exhibits a dual role in autophagy regulation. When nutrients are limited, MCL-1 preserves cellular function by shielding healthy mitochondria from non-selective autophagy; under stress, MCL-1 directly binds to LC3 and promotes the degradation of depolarized mitochondria by synergizing with BNIP3 (59). In addition, MCL-1 also participates in a broader regulatory network, indirectly regulating AMBRA1 activity and the mitophagy process by restricting the recruitment of the E3 ligase HUWE1 to the mitochondria, thus realizing the fine regulation of the mitochondrial mitophagy process (60). These synergistic effects highlight MCL-1 as a key node in the mitophagy network, but how it prioritizes reciprocal partners in different cell types or pathological contexts, and the specific contributions of these reciprocal interactions in physiology and disease, remain gaping points in current research.
4.2.4 FKBP8
FK506-binding protein 8 (FKBP8), also known as FKBP38, is a unique member of the FK506-binding protein (FKBP) family, anchored in the OMM, and a multifunctional junction protein with anti-apoptotic activity. The FKBP8 N-terminal region contains the classical LIR motif, while the C-terminal region contains a TM domain (Figure 4) (61). Under stress conditions, FKBP8 binds to LC3A via its N-terminal LIR sequence to mediate mitophagy, parallel to the PINK1-Parkin pathway. Studies have confirmed that in starvation-induced autophagy, FKBP8 interacts with BECN1 in the VPS34 lipid kinase complex, enhancing the complex’s activity to promote the production of phosphatidylinositol-3-phosphate (PI3P), which initiates mitophagy (62). Interestingly, when autophagosomes encapsulate damaged mitochondria, FKBP8 escapes from acidifying mitochondria to the endoplasmic reticulum (ER) to avoid removal (63). Currently, the precise mechanism underlying FKBP8 escape translocation during mitophagy degradation requires further investigation. Studies demonstrate that abnormally phosphorylated tau protein (p-tau) interferes with this dynamic process of FKBP8, leading to impaired function in autophagy and inhibiting oxidative stress-induced mitophagy by affecting the expression and dynamics of FKBP8 (64). It is noteworthy that FKBP8 also binds to FUNDC1 under hypoxic stress. However, the exact function of this interaction in mitophagy remains elusive.
4.3 Membrane damage-sensing receptor
Membrane damage-sensing receptor serve as sensor that respond to structural damage in mitochondria. Upon irreversible, severe damage to the mitochondrial membrane, they recognize the impairment and initiate the autophagic program to remove the completely dysfunctional organelles.
4.3.1 PHB2
Prohibitin 2 (PHB2), a mitophagy receptor protein, resides primarily in the mitochondrial inner membrane (IMM), with an N-terminal transmembrane region and a C-terminal loop region (Figure 4). PHB2 typically binds to PHB1, which is another member of the PHB family, through the C-terminal loop region to form a heterodimer and promote mitophagy (65, 66). Mitochondrial outer membrane rupture exposes PHB2 to the cytosol, enabling mitophagy mediation through binding between its LIR motif and LC3-II at the IMM. Mutation of this domain blocks PHB2-LC3 interaction and suppresses mitophagy. Moreover, PHB2 plays a key role in mitophagy by regulating the PINK1-Parkin pathway, and its activity is negatively regulated by presenilin-associated rhomboid-like protein (PARL). At homeostasis, newly synthesized PINK1 translocates into IMM via the TOM/TIM complexes. PARL, an inner membrane protease, cleaves PINK1 at its transmembrane domains, and the cleaved PINK1 is subsequently degraded rapidly by the proteasome. This process prevents the accumulation of full-length PINK1 at the OMM to activate mitophagy. The process of PHB2 mediating the stable expression of PINK1 relies on the precise regulation of the PARL-PGAM5-PINK1 axis: the binding of PHB2 to PGAM5 effectively blocks the inhibitory effect of PARL on PGAM5 activity and thus maintains the stable state of PINK1. Notably, mitochondrial depolarization triggers a dynamic shift in the interaction pattern of PHB2, promoting its preferential binding to PARL. This interaction drives the release of PGAM5, thereby relieving the inhibition of PINK1 (67). This indicates that autophagosomes can perform dual recognition of damaged mitochondria by different receptor proteins localized to the OMM and IMM. Recent studies have further revealed that the efficient exposure and functionality of PHB2 is dependent on its specific interaction with the mitochondrial outer membrane protein VDAC1: under depolarizing conditions, VDAC1 forms an oligomerization complex with PHB2, and its degradation creates a highly efficient “window of exposure” at the OMM conducive to PHB2-LC3 binding (68). However, whether VDAC1-PHB2 interactions are widely involved in physiological mitochondrial renewal, whether their regulation is cell-type specific, and whether there are other outer membrane proteins that can perform similar functions remain important directions for future research.
4.4 Immune signaling receptor
Immune signaling receptors are the most functionally unique class of mitophagy receptors. Unlike most receptors that promote mitophagy, their activation can be induced by pathogens in addition to factors such as hypoxia and energetic stress, thereby promoting mitophagy.
4.4.1 NLRX1
NOD-Like Receptor X1 (NLRX1) is a member of the Nod-like receptor (NLR) family. Its structure comprises a central NACHT domain, a C-terminal LRR, and a unique N-terminal mitochondrial targeting sequence (MTS) that directs the protein through the mitochondrial TOM/TIM complexes into the matrix, where the matrix proteasome cleaves off the first 39 amino acids to form the mature protein (Figure 4). This process was verified by experiments with MTS-deficient mutants, in which MTS-deficient NLRX1 is completely retained in the cytoplasm. Studies reveal that NLRX1 recruits LC3 to mitochondria via the LIR motif within its NACHT domain and promotes damaged mitochondria clearance (69). Although NLRX1 typically localizes to the mitochondrial matrix, mitochondrial depolarization leads to mitochondrial protein import stress (MPIS), causing cytoplasmic NLRX1 retention. Cytoplasmic NLRX1 then interacts with the endoplasmic reticulum protein Ribosome Binding Protein 1 (RRBP1); this complex induces mitophagy by promoting recruitment and lipidation of LC3 around damaged mitochondria. It has also been shown that overexpression of NLRX1 reverses FUNDC1 phosphorylation, upregulates NIPSNAP1/2, and enhances FUNDC1-NIPSNAP1/2 interactions, thereby indirectly activating mitophagy, which explains the multiplexed role of NLRX1 in the maintenance of mitochondrial homeostasis (70). Although NLRX1 has been proposed to regulate multiple cellular processes, including antiviral immunity, apoptosis, reactive oxygen species generation, and mitochondrial metabolism, whether its multifunctional roles integrate through autophagy regulation requires further investigation (71, 72).
4.5 Other receptors
This receptor is unique in that it does not mediate typical, wholesale mitophagy but rather operates through an endolysosomal-dependent pathway, functioning only under specific damage conditions.
4.5.1 SAMM50
Sorting and assembly machinery component 50 (SAMM50) is a core component of the β-barrel assembly machinery (SAM complex) on the OMM. SAMM50 is responsible for the proper folding and assembly of β-barrel proteins, which are essential for maintaining normal mitochondrial function and morphology. SAMM50 localizes to the OMM and regulates mitochondrial cristae morphology and membrane stability (73). SAMM50 contains several structural domains, of which the POTRA domain and the C-terminal transmembrane domain are its key features (74). SAMM50 mediates basal mitophagy of the SAM and MICOS components by binding to ATG8-family proteins through its N-terminal LIR motif and interacting with p62 (Figure 4) (75). Notably, SAMM50-mediated mitophagy constitutes a partial autophagic process that targets specific mitochondrial components, such as mitochondrial DNA, components of the SAM and MICOS complexes, for lysosomal degradation, thereby facilitating the renewal of specific components within the mitochondria (76, 77). In addition, SAMM50 controls mitophagy by regulating the stability of PINK1, which accumulates at the OMM when SAMM50 is knocked down, thereby promoting mitophagy (78). The case of SAMM50 strongly demonstrates that mitophagy receptors work synergistically with each other to form a complex regulatory network that is both holistic and locally complementary, laying the groundwork for great potential for future research in this field.
5 Receptors maintain mitochondrial function via other mechanisms
Beyond mediating mitophagy through diverse mechanisms, mitophagy receptors coordinate with other mitochondrial quality control systems—including mitochondrial dynamics, ion homeostasis, and mitochondrial biogenesis—to sustain cellular energy homeostasis. Mitochondrial dynamics involve continuous fission and fusion processes, through which mitochondria regulate their morphology, abundance, distribution, and functionality to meet cellular physiological demands. Mitochondrial fission produces daughter mitochondria, enabling efficient redistribution to energy-demanding cellular zones. Dysfunctional mitochondria require segregation from the network via fission or alternative mechanisms before mitophagic clearance. The most important significance of mitochondrial fusion is that it promotes mixing between the mitochondrial membrane components and matrix contents. During transient fusion events, mitochondria rapidly exchange contents to optimize morphology and functionality (79). BNIP3 inhibits optic atrophy 1 (Opa1)-mediated mitochondrial fusion while facilitating the recruitment of dynein-related protein 1(Drp1) to mitochondria, thereby promoting damaged mitochondrial breakage and segregation (80, 81). BNIP3 and NIX may indirectly affect the mitochondrial fusion process by regulating the activity of MFN1/2 (82). The phosphorylation state of FUNDC1 modulates its interaction with Drp1 or Opa1 to regulate mitochondrial dynamics (83). Notably, BCL2L13—the sole BCL-2 family protein possessing all four BH domains yet excluded from classical BCL-2 subfamilies—is involved not only in the apoptosis signaling pathway and mitophagy, but also in the regulation of mitochondrial fragmentation. The fission-promoting activity of BCL2L13, which is independent of that of the classic mitochondrial fusion protein Drp1 but requires all four of its BH domains, mutations in any of the BH1-4 domains or transmembrane motifs inhibit this process; however, the underlying mechanism remains unknown (84). In maintaining ionic homeostasis, FUNDC1 interacts with the inositol 1,4,5-trisphosphate receptor type 2 (IP3R2), which stabilizes MAMs and promotes ER Ca2 + transfer to mitochondria and cytoplasm (85). Beyond FTMT-mediated intracellular iron regulation, BNIP3 (86), NIX (87), FUNDC1 (88), and BCL2L13 (57) modulate mitochondrial iron homeostasis by altering mitochondrial morphology or outer membrane permeability. As for mitochondrial biogenesis, recent studies indicate that BNIP3 and NIX can indirectly regulate the expression of PGC-1α and its downstream target gene nuclear respiratory factor 1 (NRF1) through multiple mechanisms, thus affecting mitochondrial biogenesis. Although the precise mechanisms remain unelucidated, BNIP3/NIX, as HIF-1α target genes, likely reinforce HIF-1α-mediated PGC-1α suppression via a negative feedback loop.
6 Mitophagy and neurodegenerative diseases
6.1 Alzheimer’s disease
Alzheimer’s disease (AD), the most prevalent neurodegenerative disorder, is characterized by cerebral atrophy in the entorhinal cortex and hippocampus, resulting in synaptic loss, progressive cognitive decline, and mortality. The pathological hallmarks of AD include senile plaques (SPs) formed by extracellular amyloid-β protein (Aβ) deposition and accumulation of intracellular hyperphosphorylated microtubule-associated protein tau inclusions, resulting in neurofibrillary tangles (NFTs) that lead to synaptic degeneration, neuron loss, gliosis, and white matter pathology (89). Pathogenesis involves multiple cellular alterations, including dysfunction of mitochondria and synapses, formation and deposition of Aβ and P-Tau. Dysfunction of mitochondria and synapses often occurs early in the AD process. Aβ and P-Tau-induced defects in mitophagy are important changes in AD pathogenesis (90). The main reason for this may be that elevated levels of Aβ and Tau induce ROS production, leading to excessive mitochondrial defects. Studies confirm that restoring mitophagy improves cognitive function in AD mouse models, enhances microglial clearance of Aβ plaques, and reduces neuroinflammation (91). While the association of PINK1-Parkin-induced mitophagy with AD has been extensively demonstrated by several studies in the past, recent scholars have found that receptor-mediated mitophagy does not share the same characteristics in the pathogenesis of AD - the key receptor is directly and precisely hit by the core pathogenic proteins (Figure 5). Most typically, pathologically phosphorylated Tau was shown to specifically interfere with the function of FKBP8, directly inhibiting its ability to initiate mitophagy under oxidative stress (64). Fang et al. observed decreased AMBRA1 expression in AD patient samples and in vitro models (91). This suggests that Tau pathology contributes directly to the loss of mitochondria within neurons by “hijacking” specific mitophagy receptors. However, Sepe et al. demonstrated abundant Ambra1 content in neurons in a mouse model of AD (92). This may be due to specific “silencing” of the former by pathologic Tau-P and compensatory elevation of the latter. In addition, MCL-1, as the mitophagy receptor, has been shown to trigger mitophagy and effectively improve cognition in a mouse model of AD through the action of UMI-77 (93, 94). FTMT plays an equally important role in AD. Studies have shown that FTMT expression is significantly increased in brain tissue of AD patients, and that this increase may be related to disturbances in iron metabolism (95). Under oxidative stress conditions, FTMT levels rise and exert neuroprotection by reducing oxidative damage through the ERK/P38 MAPK pathway. However, FTMT overexpression may exacerbate Aβ toxicity. Although the specific mechanism of FTMT in AD remains to be elucidated, this dual ability to regulate iron metabolism and mitigate oxidative stress explains the multidimensional phenomenon of the same receptor in a single cell, rather than a generalized enhancement of “overall mitophagy.” Overall, mitophagy receptors may regulate mitochondrial homeostasis and promote mitophagy in response to mitochondrial stress induced by Aβ and P-Tau via the non-PINK1-Parkin pathway. However, excessive receptor activation drives pathological mitochondrial fission and apoptosis, thereby exacerbating neurodegeneration. Further studies are needed to finely regulate their expression and activity to avoid over-activation and cytotoxicity (Table 1).
Figure 5. Mechanisms of mitophagy defects in neurodegenerative diseases (created with BioRender.com).
6.2 Parkinson’s disease
Parkinson’s disease (PD) is one of the most prevalent neurodegenerative disorders, featuring dopaminergic neuron degeneration, causing striatal dopamine depletion (96). The pathology is characterized by progressive loss of dopaminergic neurons in the dense portion of the SN and an abnormal accumulation of Lewy bodies containing alpha-synuclein. PD symptoms include motor symptoms such as tremor, bradykinesia, rigidity, and postural instability, as well as non-motor deficits such as dementia, neuropsychiatric disturbances, and autonomic dysfunction. Mitochondrial dysfunction is one of the most important mechanisms in the pathogenesis of PD (Figure 5). It has been found that PD is associated with mutations in PARK6 and PARK2 genes, and that abnormalities in Pink1 and Parkin lead to insufficient mitophagy, accumulation of damaged mitochondria, excessive oxidative stress, and energy deficits, which in turn lead to neuronal death and exacerbate PD progression (97). However, PARK2 gene deletion failed to generalize the PD full phenotypes in mice, indicating the existence of other compensatory mechanisms for mitophagy. Indeed, receptor-mediated mitophagy in PD demonstrates a unique compensatory nature—when the canonical PINK1-Parkin pathway is impaired, multiple mitophagy receptors become specifically activated, forming a robust “molecular backup system.” Recent studies establish NIX-mediated mitophagy as functionally independent of Parkin. In PD patient-derived cells, NIX was found to restore mitophagy by compensating for PARK6/PARK2 mutations (98). In a murine model carrying a glucocerebrosidase β (GBA) mutant, which is a genetic risk variant for PD, NIX dimerization was reduced and accompanied by mitophagy dysfunction. Notably, the GBA mutation did not result in Parkin activity and ubiquitination modifications, suggesting that Parkin is less involved in mitophagy defects, and these data indicate that NIX represents an alternative molecular pathway to restore mitophagy in PD (99). FUNDC1, however, exhibits an even more distinctive compensatory mechanism, the Xu team demonstrated that in Drosophila, FUNDC1 significantly ameliorates the phenotype and mitochondrial dysfunction in PINK1 mutant flies. This effect is independent of its LC3-binding domain or the autophagy-related protein ATG7, but is instead mediated by its interaction with dynamin-related Drp1, which provides a potential new target for PD therapy (100). Additionally, evidence indicates that AMBRA1 levels are reduced in patients harboring GBA mutations (99). Furthermore, mitophagy is impaired at both early and late stages in cells expressing the GBA mutant protein. Notably, reductions in AMBRA1 levels have also been observed in pharmacologically induced PD cells and mouse models (101). Similarly, Di Rita A demonstrated that expression of AMBRA1 increased cell viability and reduced the generation of ROS in an in vitro model of PD (17). Morales et al. demonstrated that damaged mitochondria accumulate within spheroids formed at the axon terminals of dopaminergic neurons in the SN of rat and mouse models of PD induced by 6-hydroxydopamine and exhibited high levels of AMBRA1 and BCL2L13 immunoreactivity (102). These findings suggest that mitophagy mediated by receptors such as AMBRA1 is critically involved in PD pathogenesis. Another hallmark of PD is the particular accumulation of iron in Lewy bodies. Moreover, iron promotes the aggregation of α-synuclein, suggesting that PD and altered iron homeostasis are closely related. Shi et al. demonstrated that FTMT had a significant protective effect against neuronal damage by preventing iron redistribution to maintain iron homeostasis in a 6-hydroxydopamine (6-OHDA)-induced PD model, and that FTMT also exerted its neuroprotective effects by inhibiting oxidative stress, mitochondrial damage, and apoptotic pathways (103).
6.3 Amyotrophic lateral sclerosis
Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disorder characterized by concomitant degeneration of both upper and lower motor neurons. This motor neuron loss leads to progressive impairment of motor function, muscle atrophy, and ultimately, paralysis (104). Patients typically succumb to respiratory failure in the late stages of the disease due to paralysis of the respiratory muscles. A pathologic feature of ALS is the cytoplasmic accumulation of TAR DNA-binding protein 43 (TDP-43) in motor neurons, observed in over 95% of patients. In addition, aberrant aggregation of superoxide dismutase 1 (SOD1) and fused in sarcoma protein (FUS) can occur in motor neurons of ALS patients. The exact pathogenesis of ALS has not yet been clarified, and numerous studies have found structural abnormalities, including mitochondrial swelling and significant intracellular mitochondrial aggregation in neurons of patients with sporadic ALS (105). To date, numerous ALS-related genes have been identified, most of which are functionally linked to mitophagy, such as SOD1, OPTN, and TBK1 (Figure 5). Mutant SOD1 blocks the retrograde transport of mitochondria in neurons. In addition, it aggregates and sequesters OPTN, thereby suppressing the formation of mitophagosomes and inhibiting mitophagy flux (106, 107). Moreover, phosphorylation of OPTN at serine 177 by TBK1 kinase is essential for its binding to LC3. ALS-associated mutations impair the OPTN-TBK1 interaction, thereby suppressing autophagosome maturation (108). These findings collectively indicate a significant impairment of mitophagy in ALS, and the accumulation of damaged mitochondria is strongly suggested to play a critical role in its pathophysiology. Rogers et al. observed reduced expression levels of mitophagy-related proteins—including Parkin, PINK1, and BNIP3—in skeletal muscle tissue from ALS patients (109). However, some studies have reported that abnormal activation of AMPK in motor neurons has been proposed as a pathogenic factor in the early stages of ALS (110). Sharma et al. demonstrated that NIX was detected to be upregulated in the cerebrospinal fluid of ALS patients (111). Consistently, upregulation of NIX was documented in astrocytes derived from SOD1 mutant mouse models (112). Additional research identified FUNDC1 expression was down-regulated in the spinal cord of an ALS mouse model, and FUNDC1 overexpression was found to significantly improve motor function in ALS mice by intraspinal injection of AAV9-FUNDC1 into the spinal cord of the mouse model, demonstrating that FUNDC1-mediated mitophagy has a protective effect in ALS, improving motor function, extending survival, and reducing neuronal apoptosis (113). Furthermore, researchers have uncovered a relationship between TDP-43 and PHB2: the level of TDP-43 directly influences the abundance of PHB2, which may represent an alternative compensatory mechanism for mitophagy (114). While mitophagy has been extensively investigated in a variety of neurodegenerative diseases, with considerable consensus on its neuroprotective role, the precise mechanisms underlying its involvement in ALS remain elusive.
6.4 Huntington’s disease
Huntington’s disease (HD) is a fatal, inherited neurodegenerative disorder. At the molecular level, expansion of the CAG trinucleotide repeat in exon 1 of the gene encoding huntingtin protein (HTT) results in the N-terminal amplification of 36 or more polyglutamine (polyQ) in the mutant Huntingtin protein (mHTT). This mutant protein accumulates at high levels within neurons of the striatum and cerebral cortex, ultimately leading to neuronal death (115–117). Similar to other neurodegenerative diseases, mitochondrial dysfunction occurs in neurons with HD disease (Figure 5). Current evidence indicates that mitochondrial dysfunction induced by mHTT is a key factor in HD pathogenesis, which is manifested in two main mechanisms: first, the interaction of mHTT with autophagy-related proteins (NBR1, OPTN, and NDP52) disrupts the autophagy pathway (118). Second, mHTT impairs the axonal transport of autophagosomes by affecting huntingtin-associated protein 1 (HAP1), which is more tightly associated with mHTT than with HTT. In the presence of mHTT, impaired autophagosome transport leads to the accumulation of autophagosomes and their cargo, resulting in mitophagy dysfunction (119). HD exhibits a unique phenomenon of bidirectional mitophagy dysregulation. Notably, not only is mitophagy impaired, but its paradoxical overactivation can also drive disease progression. mHTT may recruit valosin-containing protein (VCP) to mitochondria, leading to its accumulation. This induces excessive mitophagy, ultimately resulting in mitochondrial depletion and neuronal death. Furthermore, data from Sassone indicate that BNIP3 accumulates and dimerizes in the mitochondria of human HD muscle cells (120). Separately, Francesca Sassone observed that mutant mHTT failed to induce mitochondrial damage in BNIP3-knockout cells (121). These data suggest that BNIP3 has a potential role in mutant mHTT-induced mitochondrial dysfunction. In addition, inhibition of tissue transglutaminase 2 (TG2) has demonstrated therapeutic benefits in animal models of HD, and PHB2 serves as a substrate for TG2, which may be involved in HD pathophysiological (122, 123). However, further exploration is needed. In summary, the mitophagy pathway in HD does not undergo simple enhancement or suppression, but rather exists in an imbalanced state cohabited by both functional impairment and paradoxical hyperactivation. These alterations drive the accumulation of dysfunctional cellular components, which over time exacerbates disease manifestations.
6.5 Multiple sclerosis
Multiple sclerosis (MS) is an immune-mediated neurological disorder characterized by focal demyelination and secondary axonal damage in the central nervous system, representing a leading cause of neurological disability in young adults (124, 125). A substantial body of evidence indicates that mitochondrial dysfunction is closely associated with the pathogenesis and progression of this disease (125, 126) (Figure 5). Pro-inflammatory conditions induce mitochondrial stress, resulting in elevated glucose catabolism and mitophagy. Patergnani et al. observed significantly elevated levels of Parkin and PINK1 in the cerebrospinal fluid of multiple sclerosis patients, which returned to near-baseline levels during disease remission (127). A similar phenomenon was observed in both in vivo and in vitro MS models, indicating activation of a compensatory mitophagy process (128). This finding has also been confirmed by a cohort study from Japan (129). Variants in the 3’UTR microRNA binding sites of BCL2L13 have been identified in multiple sclerosis, which may alter BCL2L13 protein expression levels and consequently disrupt key processes such as mitophagy and apoptosis (130). Furthermore, researchers have found that elevated PHB2 protein levels in multiple sclerosis may represent a compensatory cellular response to mitochondrial damage, aimed at promoting mitophagy and improving neuronal function (131). Notably, NLRX1—a protein known for regulating the innate immune system—exerts immunosuppressive and anti-inflammatory effects in neurons under autoimmune attack. Rare NLRX1 mutations have been identified in MS patients with a family history (132). To evaluate its therapeutic potential, injection of an NLRX1-LRR fusion protein in a mouse model reduced overall disease severity, decreased T-cell infiltration, and lowered inflammatory cytokine levels in spinal cord tissue (133). This reveals the receptor’s indispensable role in the coordinated regulation of mitophagy and innate immune signaling.
7 Receptor-mediated mitophagy as a therapeutic target
Given the crucial role of receptor-mediated mitophagy in neurodegenerative diseases, pharmacological modulation of this pathway may represent an effective therapeutic approach for mitochondria-related disorders. With extensive research, a growing number of compounds (Table 2) have been identified that can restore impaired mitophagy and exert neuroprotective effects in animal or cellular models. Examples include urolithin A (UA), NAD+ precursors, among which certain natural products demonstrate particular promise. However, as most studies have been conducted over relatively short timeframes, research on mitophagy modulators remains in its early stages—though this also implies substantial potential for future applications. To achieve therapeutic targeting, such modulators must specifically localize to mitochondria to regulate mitophagy. Yet, most existing agents exhibit limited specificity toward mitophagy. Enhancing their pharmacological precision for mitophagy targets remains a technical challenge. Moreover, given possible differences in etiology and pathological processes across different diseases, there is a need to develop more personalized treatment strategies tailored to the needs of individual patients. In summary, targeting receptor-mediated mitophagy for neurodegenerative diseases is only the beginning. Continued in-depth research is essential to develop highly specific modulators of mitophagy.
8 Conclusions and future perspectives
Neurodegenerative diseases (NDDs) still lack effective disease therapies, which highlights a huge gap in our understanding of the complex cellular mechanisms involved. Mitophagy is essential for the control of metabolic homeostasis and the removal of damaged or excess mitochondria, and plays an important role in both the initiation and progression of NDDs. This review elucidates that receptor-mediated mitophagy is not merely a fundamental pathway, but functions as a critical compensatory mechanism when canonical pathways such as PINK1/Parkin are impaired, independently initiating mitochondrial clearance to maintain neuronal survival. This discovery reveals its significant potential as both a biomarker and a therapeutic target. Although we have enumerated numerous synthetic and natural compounds that have been shown to promote such mitophagy, however, translating this potential into practical therapeutic strategies still faces a number of key challenges, such as the temporal and spatial interaction patterns of different mitophagy pathways that are regulated during pathological processes, and how to achieve selective receptor activation while avoiding excessive mitophagy process? And whether they are cell-type specific in the broader physiopathological context have yet to be fully elucidated (Figure 6). These challenges, however, chart a course for future investigation. More extensive and in-depth studies on the intrinsic mechanism of dysregulated mitophagy in NDDs will help to unravel the last veil of NDDs and open new therapeutic avenues for NDDs.
Figure 6. Challenges for future research on receptor-mediated mitophagy in neurodegenerative diseases (created with BioRender.com).
Author contributions
JY: Conceptualization, Writing – original draft, Writing – review & editing. FY: Methodology, Writing – original draft. GC: Visualization, Writing – original draft. ML: Writing – review & editing. SY: Writing – review & editing. T-eZ: Project administration, Writing – review & editing.
Funding
The author(s) declare that financial support was received for the research and/or publication of this article. This study is funded by the Key Research and Development Program of Science and Technology Department of Sichuan Province (Grant, No.2023YFS0333); Innovative Research Team Project from Chengdu University of TCM (2023), the Special Research Project of Sichuan Provincial Administration of Traditional Chinese Medicine (Grant, No. 2024MS041), University & Hospital Joint Innovation Fund Project (Grant, No. LH202402017).
Acknowledgments
The images in this article were drawn by BioRender (www.biorender.com).
Conflict of interest
The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.
Generative AI statement
The authors declare that no Gen AI was used in the creation of this manuscript.
Any alternative text (alt text) provided alongside figures in this article has been generated by Frontiers with the support of artificial intelligence and reasonable efforts have been made to ensure accuracy, including review by the authors wherever possible. If you identify any issues, please contact us.
Publisher’s note
All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.
References
1. Wilson, DM 3rd, Cookson, MR, Van Den Bosch, L, Zetterberg, H, Holtzman, DM, Dewachter, I, et al. Hallmarks of neurodegenerative diseases. Cell. (2023) 186:693–714. doi: 10.1016/j.cell.2022.12.032
2. Lima, T, Li, TY, Mottis, A, and Auwerx, J. Pleiotropic effects of mitochondria in aging. Nat Aging. (2022) 2:199–213. doi: 10.1038/s43587-022-00191-2
3. Grimm, A, and Eckert, A. Brain aging and neurodegeneration: from a mitochondrial point of view. J Neurochem. (2017) 143:418–31. doi: 10.1111/jnc.14037
4. Harbauer, AB. Mitochondrial health maintenance in axons. Biochem Soc Trans. (2017) 45:1045–52. doi: 10.1042/bst20170023
5. Fivenson, EM, Lautrup, S, Sun, N, Scheibye-Knudsen, M, Stevnsner, T, Nilsen, H, et al. Mitophagy in neurodegeneration and aging. Neurochem Int. (2017) 109:202–9. doi: 10.1016/j.neuint.2017.02.007
6. Ordureau, A, Kraus, F, Zhang, J, An, H, Park, S, Ahfeldt, T, et al. Temporal proteomics during neurogenesis reveals large-scale proteome and organelle remodeling via selective autophagy. Mol Cell. (2021) 81:5082–98.e11. doi: 10.1016/j.molcel.2021.10.001
7. Chan, DC. Mitochondrial dynamics and its involvement in disease. Annu Rev Pathol. (2020) 15:235–59. doi: 10.1146/annurev-pathmechdis-012419-032711
8. Jiao, H, Jiang, D, Hu, X, Du, W, Ji, L, Yang, Y, et al. Mitocytosis, a Migrasome-mediated mitochondrial quality-control process. Cell. (2021) 184:2896–910.e13. doi: 10.1016/j.cell.2021.04.027
9. Roca-Portoles, A, and Tait, SWG. Mitochondrial quality control: from molecule to organelle. Cell Mol Life Sci. (2021) 78:3853–66. doi: 10.1007/s00018-021-03775-0
10. Bakula, D, and Scheibye-Knudsen, M. Mitophaging: mitophagy in aging and disease. Front Cell Dev Biol. (2020) 8:239. doi: 10.3389/fcell.2020.00239
11. Ozgen, S, Krigman, J, Zhang, R, and Sun, N. Significance of mitochondrial activity in neurogenesis and neurodegenerative diseases. Neural Regen Res. (2022) 17:741–7. doi: 10.4103/1673-5374.322429
12. Bernardini, JP, Lazarou, M, and Dewson, G. Parkin and Mitophagy in Cancer. Oncogene. (2017) 36:1315–27. doi: 10.1038/onc.2016.302
13. Lemasters, JJ. Selective mitochondrial autophagy, or Mitophagy, as a targeted defense against oxidative stress, mitochondrial dysfunction, and aging. Rejuvenation Res. (2005) 8:3–5. doi: 10.1089/rej.2005.8.3
14. Picca, A, Faitg, J, Auwerx, J, Ferrucci, L, and D'Amico, D. Mitophagy in human health, ageing and disease. Nat Metab. (2023) 5:2047–61. doi: 10.1038/s42255-023-00930-8
15. Szargel, R, Shani, V, Abd Elghani, F, Mekies, LN, Liani, E, Rott, R, et al. The Pink1, Synphilin-1 and Siah-1 complex constitutes a novel Mitophagy pathway. Hum Mol Genet. (2016) 25:3476–90. doi: 10.1093/hmg/ddw189
16. Puri, R, Cheng, XT, Lin, MY, Huang, N, and Sheng, ZH. Defending stressed mitochondria: uncovering the role of Mul1 in suppressing neuronal Mitophagy. Autophagy. (2020) 16:176–8. doi: 10.1080/15548627.2019.1687216
17. Di Rita, A, Peschiaroli, A, D Acunzo, P, Strobbe, D, Hu, Z, Gruber, J, et al. Huwe1 E3 ligase promotes Pink1/Parkin-independent mitophagy by regulating Ambra1 activation via Ikkα. Nat Commun. (2018) 9:3755. doi: 10.1038/s41467-018-05722-3
18. 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) 8:304. doi: 10.1038/s41392-023-01503-7
19. Rasool, S, Soya, N, Truong, L, Croteau, N, Lukacs, GL, and Trempe, JF. Pink1 autophosphorylation is required for ubiquitin recognition. EMBO Rep. (2018) 19:e44981. doi: 10.15252/embr.201744981
20. Han, R, Liu, Y, Li, S, Li, XJ, and Yang, W. Pink1-Prkn mediated Mitophagy: differences between in vitro and in vivo models. Autophagy. (2023) 19:1396–405. doi: 10.1080/15548627.2022.2139080
21. Sekine, S. Pink1 import regulation at a crossroad of mitochondrial fate: the molecular mechanisms of Pink1 import. J Biochem. (2020) 167:217–24. doi: 10.1093/jb/mvz069
22. Matsumoto, G, Wada, K, Okuno, M, Kurosawa, M, and Nukina, N. Serine 403 phosphorylation of P62/Sqstm1 regulates selective Autophagic clearance of Ubiquitinated proteins. Mol Cell. (2011) 44:279–89. doi: 10.1016/j.molcel.2011.07.039
23. Rasmussen, NL, Kournoutis, A, Lamark, T, and Johansen, T. Nbr1: the archetypal selective autophagy receptor. J Cell Biol. (2022) 221:e202208092. doi: 10.1083/jcb.202208092
24. 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) 12:5212. doi: 10.1038/s41467-021-25572-w
25. Richter, B, Sliter, DA, Herhaus, L, Stolz, A, Wang, C, Beli, P, et al. Phosphorylation of Optn by Tbk1 enhances its binding to Ub chains and promotes selective autophagy of damaged mitochondria. Proc Natl Acad Sci USA. (2016) 113:4039–44. doi: 10.1073/pnas.1523926113
26. Terešak, P, Lapao, A, Subic, N, Boya, P, Elazar, Z, and Simonsen, A. Regulation of Prkn-independent Mitophagy. Autophagy. (2022) 18:24–39. doi: 10.1080/15548627.2021.1888244
27. Hanna, RA, Quinsay, MN, Orogo, AM, Giang, K, Rikka, S, and Gustafsson, ÅB. Microtubule-associated protein 1 light chain 3 (Lc3) interacts with Bnip3 protein to selectively remove endoplasmic reticulum and mitochondria via autophagy. J Biol Chem. (2012) 287:19094–104. doi: 10.1074/jbc.M111.322933
28. Chen, G, Cizeau, J, Vande Velde, C, Park, JH, Bozek, G, Bolton, J, et al. Nix and Nip3 form a subfamily of pro-apoptotic mitochondrial proteins. J Biol Chem. (1999) 274:7–10. doi: 10.1074/jbc.274.1.7
29. Gao, A, Jiang, J, Xie, F, and Chen, L. Bnip3 in Mitophagy: novel insights and potential therapeutic target for diseases of secondary mitochondrial dysfunction. Clin Chim Acta. (2020) 506:72–83. doi: 10.1016/j.cca.2020.02.024
30. Novak, I, Kirkin, V, McEwan, DG, Zhang, J, Wild, P, Rozenknop, A, et al. Nix is a selective autophagy receptor for mitochondrial clearance. EMBO Rep. (2010) 11:45–51. doi: 10.1038/embor.2009.256
31. Schweers, RL, Zhang, J, Randall, MS, Loyd, MR, Li, W, Dorsey, FC, et al. Nix is required for programmed mitochondrial clearance during reticulocyte maturation. Proc Natl Acad Sci USA. (2007) 104:19500–5. doi: 10.1073/pnas.0708818104
32. Sandoval, H, Thiagarajan, P, Dasgupta, SK, Schumacher, A, Prchal, JT, Chen, M, et al. Essential role for nix in Autophagic maturation of erythroid cells. Nature. (2008) 454:232–5. doi: 10.1038/nature07006
33. Marinković, M, Šprung, M, and Novak, I. Dimerization of Mitophagy receptor Bnip3l/nix is essential for recruitment of Autophagic machinery. Autophagy. (2021) 17:1232–43. doi: 10.1080/15548627.2020.1755120
34. Gao, F, Chen, D, Si, J, Hu, Q, Qin, Z, Fang, M, et al. The mitochondrial protein Bnip3l is the substrate of Park2 and mediates Mitophagy in Pink1/Park2 pathway. Hum Mol Genet. (2015) 24:2528–38. doi: 10.1093/hmg/ddv017
35. Li, Y, Zheng, W, Lu, Y, Zheng, Y, Pan, L, Wu, X, et al. Bnip3l/nix-mediated mitophagy: molecular mechanisms and implications for human disease. Cell Death Dis. (2021) 13:14. doi: 10.1038/s41419-021-04469-y
36. Han, S, Jeong, YY, Sheshadri, P, Su, X, and Cai, Q. Mitophagy regulates integrity of mitochondria at synapses and is critical for synaptic maintenance. EMBO Rep. (2020) 21:e49801. doi: 10.15252/embr.201949801
37. He, YL, Li, J, Gong, SH, Cheng, X, Zhao, M, Cao, Y, et al. Bnip3 phosphorylation by Jnk1/2 promotes Mitophagy via enhancing its stability under hypoxia. Cell Death Dis. (2022) 13:966. doi: 10.1038/s41419-022-05418-z
38. Cao, Y, Zheng, J, Wan, H, Sun, Y, Fu, S, Liu, S, et al. A mitochondrial Scf-Fbxl4 ubiquitin E3 ligase complex degrades Bnip3 and nix to restrain Mitophagy and prevent mitochondrial disease. EMBO J. (2023) 42:e113033. doi: 10.15252/embj.2022113033
39. Sun, Y, Cao, Y, Wan, H, Memetimin, A, Cao, Y, Li, L, et al. A Mitophagy sensor Pptc7 controls Bnip3 and nix degradation to regulate mitochondrial mass. Mol Cell. (2024) 84:327–44.e9. doi: 10.1016/j.molcel.2023.11.038
40. Wu, X, Wu, FH, Wu, Q, Zhang, S, Chen, S, and Sima, M. Phylogenetic and molecular evolutionary analysis of Mitophagy receptors under hypoxic conditions. Front Physiol. (2017) 8:539. doi: 10.3389/fphys.2017.00539
41. Liu, L, Feng, D, Chen, G, Chen, M, Zheng, Q, Song, P, et al. Mitochondrial outer-membrane protein Fundc1 mediates hypoxia-induced Mitophagy in mammalian cells. Nat Cell Biol. (2012) 14:177–85. doi: 10.1038/ncb2422
42. Liu, L, Li, Y, Wang, J, Zhang, D, Wu, H, Li, W, et al. Mitophagy receptor Fundc1 is regulated by Pgc-1α/Nrf1 to fine tune mitochondrial homeostasis. EMBO Rep. (2021) 22:e50629. doi: 10.15252/embr.202050629
43. Ma, K, Zhang, Z, Chang, R, Cheng, H, Mu, C, Zhao, T, et al. Dynamic Pgam5 Multimers dephosphorylate Bcl-xl or Fundc1 to regulate mitochondrial and cellular fate. Cell Death Differ. (2020) 27:1036–51. doi: 10.1038/s41418-019-0396-4
44. Lv, M, Wang, C, Li, F, Peng, J, Wen, B, Gong, Q, et al. Structural insights into the recognition of phosphorylated Fundc1 by Lc3b in Mitophagy. Protein Cell. (2017) 8:25–38. doi: 10.1007/s13238-016-0328-8
45. Zhang, W. The Mitophagy receptor Fun14 domain-containing 1 (Fundc1): a promising biomarker and potential therapeutic target of human diseases. Genes Dis. (2021) 8:640–54. doi: 10.1016/j.gendis.2020.08.011
46. Chen, Z, Liu, L, Cheng, Q, Li, Y, Wu, H, Zhang, W, et al. Mitochondrial E3 ligase March5 regulates Fundc1 to fine-tune hypoxic Mitophagy. EMBO Rep. (2017) 18:495–509. doi: 10.15252/embr.201643309
47. Yanatori, I, Nishina, S, Kishi, F, and Hino, K. Newly uncovered biochemical and functional aspects of ferritin. FASEB J. (2023) 37:e23095. doi: 10.1096/fj.202300918R
48. Wu, Q, Wu, WS, Su, L, Zheng, X, Wu, WY, Santambrogio, P, et al. Mitochondrial ferritin is a hypoxia-inducible factor 1α-inducible gene that protects from hypoxia-induced cell death in brain. Antioxid Redox Signal. (2019) 30:198–212. doi: 10.1089/ars.2017.7063
49. Hara, Y, Yanatori, I, Tanaka, A, Kishi, F, Lemasters, JJ, Nishina, S, et al. Iron loss triggers Mitophagy through induction of mitochondrial ferritin. EMBO Rep. (2020) 21:e50202. doi: 10.15252/embr.202050202
50. Fuhrmann, DC, Mondorf, A, Beifuß, J, Jung, M, and Brüne, B. Hypoxia inhibits Ferritinophagy, increases mitochondrial ferritin, and protects from Ferroptosis. Redox Biol. (2020) 36:101670. doi: 10.1016/j.redox.2020.101670
51. Liu, M, Wang, Y, Teng, F, Mai, X, Wang, X, Su, MY, et al. Structure of the Ddb1-Ambra1 E3 ligase receptor complex linked to cell cycle regulation. Nat Commun. (2023) 14:7631. doi: 10.1038/s41467-023-43174-6
52. Nazio, F, Strappazzon, F, Antonioli, M, Bielli, P, Cianfanelli, V, Bordi, M, et al. Mtor inhibits autophagy by controlling Ulk1 Ubiquitylation, self-association and function through Ambra1 and Traf6. Nat Cell Biol. (2013) 15:406–16. doi: 10.1038/ncb2708
53. Sankar, DS, Kaeser-Pebernard, S, Vionnet, C, Favre, S, de Oliveira Marchioro, L, Pillet, B, et al. The Ulk1 effector Bag2 regulates autophagy initiation by modulating Ambra1 localization. Cell Rep. (2024) 43:114689. doi: 10.1016/j.celrep.2024.114689
54. Di Rienzo, M, Romagnoli, A, Ciccosanti, F, Refolo, G, Consalvi, V, Arena, G, et al. Ambra1 regulates Mitophagy by interacting with Atad3a and promoting Pink1 stability. Autophagy. (2022) 18:1752–62. doi: 10.1080/15548627.2021.1997052
55. Kataoka, T, Holler, N, Micheau, O, Martinon, F, Tinel, A, Hofmann, K, et al. Bcl-Rambo, a novel Bcl-2 homologue that induces apoptosis via its unique C-terminal extension. J Biol Chem. (2001) 276:19548–54. doi: 10.1074/jbc.M010520200
56. Murakawa, T, and Otsu, K. Phosphorylation of Bcl2l13 by Prkaa2/Ampkα2 activates Mitophagy in pressure-overloaded heart. Autophagy. (2025) 21:1382–3. doi: 10.1080/15548627.2025.2465408
57. Meng, F, Sun, N, Liu, D, Jia, J, Xiao, J, and Dai, H. Bcl2l13: physiological and pathological meanings. Cell Mol Life Sci. (2021) 78:2419–28. doi: 10.1007/s00018-020-03702-9
58. Hollville, E, Carroll, RG, Cullen, SP, and Martin, SJ. Bcl-2 family proteins participate in mitochondrial quality control by regulating Parkin/Pink1-dependent Mitophagy. Mol Cell. (2014) 55:451–66. doi: 10.1016/j.molcel.2014.06.001
59. Moyzis, AG, Lally, NS, Liang, W, Najor, RH, and Gustafsson, ÅB. Mcl-1 differentially regulates autophagy in response to changes in energy status and mitochondrial damage. Cells. (2022) 11:1469. doi: 10.3390/cells11091469
60. Strappazzon, F, Di Rita, A, Peschiaroli, A, Leoncini, PP, Locatelli, F, Melino, G, et al. Huwe1 controls Mcl1 stability to unleash Ambra1-induced Mitophagy. Cell Death Differ. (2020) 27:1155–68. doi: 10.1038/s41418-019-0404-8
61. Shirane, M, and Nakayama, KI. Inherent Calcineurin inhibitor Fkbp38 targets Bcl-2 to mitochondria and inhibits apoptosis. Nat Cell Biol. (2003) 5:28–37. doi: 10.1038/ncb894
62. Aguilera, MO, Robledo, E, Melani, M, Wappner, P, and Colombo, MI. Fkbp8 is a novel molecule that participates in the regulation of the Autophagic pathway. Biochim Biophys Acta, Mol Cell Res. (2022) 1869:119212. doi: 10.1016/j.bbamcr.2022.119212
63. Bhujabal, Z, Birgisdottir, ÅB, Sjøttem, E, Brenne, HB, Øvervatn, A, Habisov, S, et al. Fkbp8 recruits Lc3a to mediate Parkin-independent Mitophagy. EMBO Rep. (2017) 18:947–61. doi: 10.15252/embr.201643147
64. Isei, MO, Crockett, M, Chen, E, Rodwell-Bullock, J, Carroll, T, Girardi, PA, et al. Tau phosphorylation suppresses oxidative stress-induced Mitophagy via Fkbp8 receptor modulation. PLoS One. (2025) 20:e0307358. doi: 10.1371/journal.pone.0307358
65. Osman, C, Merkwirth, C, and Langer, T. Prohibitins and the functional compartmentalization of mitochondrial membranes. J Cell Sci. (2009) 122:3823–30. doi: 10.1242/jcs.037655
66. García-Chávez, D, Domínguez-Martín, E, Kawasaki, L, Ongay-Larios, L, Ruelas-Ramírez, H, Mendoza-Martinez, AE, et al. Prohibitins, Phb1 and Phb2, function as Atg8 receptors to support yeast Mitophagy and also play a negative regulatory role in Atg32 processing. Autophagy. (2024) 20:2478–89. doi: 10.1080/15548627.2024.2371717
67. Yan, C, Gong, L, Chen, L, Xu, M, Abou-Hamdan, H, Tang, M, et al. Phb2 (Prohibitin 2) promotes Pink1-Prkn/Parkin-dependent Mitophagy by the Parl-Pgam5-Pink1 Axis. Autophagy. (2020) 16:419–34. doi: 10.1080/15548627.2019.1628520
68. Roy, M, Nandy, S, Marchesan, E, Banerjee, C, Mondal, R, Caicci, F, et al. Efficient Phb2 (Prohibitin 2) exposure during Mitophagy depends on Vdac1 (voltage dependent Anion Channel 1). Autophagy. (2025) 21:897–909. doi: 10.1080/15548627.2024.2426116
69. Killackey, SA, Bi, Y, Soares, F, Hammi, I, Winsor, NJ, Abdul-Sater, AA, et al. Mitochondrial protein import stress regulates the Lc3 Lipidation step of Mitophagy through Nlrx1 and Rrbp1. Mol Cell. (2022) 82:e5:2815–31. doi: 10.1016/j.molcel.2022.06.004
70. Li, S, Zhou, Y, Gu, X, Zhang, X, and Jia, Z. Nlrx1/Fundc1/Nipsnap1-2 Axis regulates Mitophagy and alleviates intestinal Ischaemia/reperfusion injury. Cell Prolif. (2021) 54:e12986. doi: 10.1111/cpr.12986
71. Bi, PY, Killackey, SA, Schweizer, L, and Girardin, SE. Nlrx1: versatile functions of a mitochondrial Nlr protein that controls Mitophagy. Biom J. (2024) 47:100635. doi: 10.1016/j.bj.2023.100635
72. Zhang, Y, Yao, Y, Qiu, X, Wang, G, Hu, Z, Chen, S, et al. Listeria hijacks host Mitophagy through a novel Mitophagy receptor to evade killing. Nat Immunol. (2019) 20:433–46. doi: 10.1038/s41590-019-0324-2
73. Chen, L, Dong, J, Liao, S, Wang, S, Wu, Z, Zuo, M, et al. Loss of Sam50 in hepatocytes induces Cardiolipin-dependent mitochondrial membrane remodeling to trigger Mtdna release and liver injury. Hepatology. (2022) 76:1389–408. doi: 10.1002/hep.32471
74. Abudu, YP, Mouilleron, S, Tooze, SA, Lamark, T, and Johansen, T. Samm50 is a receptor for basal piecemeal Mitophagy and acts with Sqstm1/P62 in Oxphos-induced Mitophagy. Autophagy. (2021) 17:2656–8. doi: 10.1080/15548627.2021.1953846
75. Abudu, YP, Shrestha, BK, Zhang, W, Palara, A, Brenne, HB, Larsen, KB, et al. Samm50 acts with P62 in piecemeal basal- and Oxphos-induced Mitophagy of Sam and Micos components. J Cell Biol. (2021) 220:e202009092. doi: 10.1083/jcb.202009092
76. Le Guerroué, F, Eck, F, Jung, J, Starzetz, T, Mittelbronn, M, Kaulich, M, et al. Autophagosomal content profiling reveals an Lc3c-dependent piecemeal Mitophagy pathway. Mol Cell. (2017) 68:786–96.e6. doi: 10.1016/j.molcel.2017.10.029
77. Sen, A, Boix, J, and Pla-Martín, D. Endosomal-dependent Mitophagy coordinates mitochondrial nucleoid and Mtdna elimination. Autophagy. (2023) 19:2609–10. doi: 10.1080/15548627.2023.2170959
78. Jian, F, Chen, D, Chen, L, Yan, C, Lu, B, Zhu, Y, et al. Sam50 regulates Pink1-Parkin-mediated Mitophagy by controlling Pink1 stability and mitochondrial morphology. Cell Rep. (2018) 23:2989–3005. doi: 10.1016/j.celrep.2018.05.015
79. Tábara, LC, Segawa, M, and Prudent, J. Molecular mechanisms of mitochondrial dynamics. Nat Rev Mol Cell Biol. (2025) 26:123–46. doi: 10.1038/s41580-024-00785-1
80. Landes, T, Emorine, LJ, Courilleau, D, Rojo, M, Belenguer, P, and Arnauné-Pelloquin, L. The Bh3-only Bnip3 binds to the dynamin Opa1 to promote mitochondrial fragmentation and apoptosis by distinct mechanisms. EMBO Rep. (2010) 11:459–65. doi: 10.1038/embor.2010.50
81. Lee, Y, Lee, HY, Hanna, RA, and Gustafsson, ÅB. Mitochondrial autophagy by Bnip3 involves Drp1-mediated mitochondrial fission and recruitment of Parkin in cardiac myocytes. Am J Physiol Heart Circ Physiol. (2011) 301:H1924–31. doi: 10.1152/ajpheart.00368.2011
82. Chen, H, and Chan, DC. Mitochondrial dynamics--fusion, fission, movement, and mitophagy--in neurodegenerative diseases. Hum Mol Genet. (2009) 18:R169–76. doi: 10.1093/hmg/ddp326
83. Chen, M, Chen, Z, Wang, Y, Tan, Z, Zhu, C, Li, Y, et al. Mitophagy receptor Fundc1 regulates mitochondrial dynamics and Mitophagy. Autophagy. (2016) 12:689–702. doi: 10.1080/15548627.2016.1151580
84. Murakawa, T, Yamaguchi, O, Hashimoto, A, Hikoso, S, Takeda, T, Oka, T, et al. Bcl-2-like protein 13 is a mammalian Atg32 homologue that mediates Mitophagy and mitochondrial fragmentation. Nat Commun. (2015) 6:7527. doi: 10.1038/ncomms8527
85. Wu, S, Lu, Q, Wang, Q, Ding, Y, Ma, Z, Mao, X, et al. Binding of Fun14 domain containing 1 with inositol 1,4,5-trisphosphate receptor in mitochondria-associated endoplasmic reticulum membranes maintains mitochondrial dynamics and function in hearts in vivo. Circulation. (2017) 136:2248–66. doi: 10.1161/circulationaha.117.030235
86. Wang, XX, Li, M, Xu, XW, Zhao, WB, Jin, YM, Li, LL, et al. Bnip3-mediated Mitophagy attenuates hypoxic-ischemic brain damage in neonatal rats by inhibiting Ferroptosis through P62-Keap1-Nrf2 pathway activation to maintain Iron and redox homeostasis. Acta Pharmacol Sin. (2025) 46:33–51. doi: 10.1038/s41401-024-01365-x
87. Yamashita, SI, Sugiura, Y, Matsuoka, Y, Maeda, R, Inoue, K, Furukawa, K, et al. Mitophagy mediated by Bnip3 and nix protects against Ferroptosis by downregulating mitochondrial reactive oxygen species. Cell Death Differ. (2024) 31:651–61. doi: 10.1038/s41418-024-01280-y
88. Pei, Z, Liu, Y, Liu, S, Jin, W, Luo, Y, Sun, M, et al. Fundc1 insufficiency sensitizes high fat diet intake-induced cardiac remodeling and contractile anomaly through Acsl4-mediated Ferroptosis. Metabolism. (2021) 122:154840. doi: 10.1016/j.metabol.2021.154840
89. Zheng, Q, and Wang, X. Alzheimer's disease: insights into pathology, molecular mechanisms, and therapy. Protein Cell. (2025) 16:83–120. doi: 10.1093/procel/pwae026
90. Reddy, PH, and Oliver, DM. Amyloid beta and phosphorylated tau-induced defective autophagy and mitophagy in Alzheimer's disease. Cells. (2019) 8:488. doi: 10.3390/cells8050488
91. 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) 22:401–12. doi: 10.1038/s41593-018-0332-9
92. Sepe, S, Nardacci, R, Fanelli, F, Rosso, P, Bernardi, C, Cecconi, F, et al. Expression of Ambra1 in mouse brain during physiological and Alzheimer type aging. Neurobiol Aging. (2014) 35:96–108. doi: 10.1016/j.neurobiolaging.2013.07.001
93. Cen, X, Chen, Y, Xu, X, Wu, R, He, F, Zhao, Q, et al. Pharmacological targeting of mcl-1 promotes Mitophagy and improves disease pathologies in an Alzheimer's disease mouse model. Nat Commun. (2020) 11:5731. doi: 10.1038/s41467-020-19547-6
94. Cen, X, Xu, X, and Xia, H. Targeting Mcl1 to induce Mitophagy is a potential therapeutic strategy for Alzheimer disease. Autophagy. (2021) 17:818–9. doi: 10.1080/15548627.2020.1860542
95. Liu, JL, Fan, YG, Yang, ZS, Wang, ZY, and Guo, C. Iron and Alzheimer's disease: from pathogenesis to therapeutic implications. Front Neurosci. (2018) 12:632. doi: 10.3389/fnins.2018.00632
96. Bloem, BR, Okun, MS, and Klein, C. Parkinson's disease. Lancet. (2021) 397:2284–303. doi: 10.1016/s0140-6736(21)00218-x
97. Zilocchi, M, Finzi, G, Lualdi, M, Sessa, F, Fasano, M, and Alberio, T. Mitochondrial alterations in Parkinson's disease human samples and cellular models. Neurochem Int. (2018) 118:61–72. doi: 10.1016/j.neuint.2018.04.013
98. Koentjoro, B, Park, JS, and Sue, CM. Nix restores Mitophagy and mitochondrial function to protect against Pink1/Parkin-related Parkinson's disease. Sci Rep. (2017) 7:44373. doi: 10.1038/srep44373
99. Li, H, Ham, A, Ma, TC, Kuo, SH, Kanter, E, Kim, D, et al. Mitochondrial dysfunction and Mitophagy defect triggered by heterozygous Gba mutations. Autophagy. (2019) 15:113–30. doi: 10.1080/15548627.2018.1509818
100. Xu, J, Deng, Z, Shang, S, Wang, C, and Han, H. Fundc1 collaborates with Pink1 in regulating mitochondrial fission and compensating for Pink1 deficiency. Biochem Biophys Res Commun. (2023) 687:149210. doi: 10.1016/j.bbrc.2023.149210
101. Zhou, J, Zhao, Y, Li, Z, Zhu, M, Wang, Z, Li, Y, et al. Mir-103a-3p regulates Mitophagy in Parkinson's disease through Parkin/Ambra1 signaling. Pharmacol Res. (2020) 160:105197. doi: 10.1016/j.phrs.2020.105197
102. Morales, I, Sanchez, A, Puertas-Avendaño, R, Rodriguez-Sabate, C, Perez-Barreto, A, and Rodriguez, M. Neuroglial Transmitophagy and Parkinson's disease. Glia. (2020) 68:2277–99. doi: 10.1002/glia.23839
103. Shi, ZH, Nie, G, Duan, XL, Rouault, T, Wu, WS, Ning, B, et al. Neuroprotective mechanism of mitochondrial ferritin on 6-Hydroxydopamine-induced dopaminergic cell damage: implication for neuroprotection in Parkinson's disease. Antioxid Redox Signal. (2010) 13:783–96. doi: 10.1089/ars.2009.3018
104. Feldman, EL, Goutman, SA, Petri, S, Mazzini, L, Savelieff, MG, Shaw, PJ, et al. Amyotrophic lateral sclerosis. Lancet. (2022) 400:1363–80. doi: 10.1016/s0140-6736(22)01272-7
105. Moore, AS, and Holzbaur, EL. Dynamic recruitment and activation of Als-associated Tbk1 with its target Optineurin are required for efficient Mitophagy. Proc Natl Acad Sci USA. (2016) 113:E3349–58. doi: 10.1073/pnas.1523810113
106. Tak, YJ, Park, JH, Rhim, H, and Kang, S. Als-related mutant Sod1 aggregates interfere with Mitophagy by sequestering the autophagy receptor Optineurin. Int J Mol Sci. (2020) 21:7525. doi: 10.3390/ijms21207525
107. Wen, D, Ji, Y, Li, Y, Duan, W, Wang, Y, Li, Z, et al. Optn gene therapy increases autophagy and protects mitochondria in Sod1-G93a-expressing transgenic mice and cells. FEBS J. (2024) 291:795–813. doi: 10.1111/febs.17009
108. He, Y, Liu, Y, Zheng, M, Zou, Y, Huang, M, Wang, L, et al. Targeting Atad3a phosphorylation mediated by Tbk1 ameliorates senescence-associated pathologies. Adv Sci (Weinh). (2025) 12:e2404109. doi: 10.1002/advs.202404109
109. Rogers, RS, Tungtur, S, Tanaka, T, Nadeau, LL, Badawi, Y, Wang, H, et al. Impaired Mitophagy plays a role in denervation of neuromuscular junctions in Als mice. Front Neurosci. (2017) 11:473. doi: 10.3389/fnins.2017.00473
110. Liu, YJ, Kuo, HC, and Chern, Y. A system-wide Mislocalization of Rna-binding proteins in motor neurons is a new feature of Als. Neurobiol Dis. (2021) 160:105531. doi: 10.1016/j.nbd.2021.105531
111. Sharma, A, Varghese, AM, Vijaylakshmi, K, Sumitha, R, Prasanna, VK, Shruthi, S, et al. Cerebrospinal fluid from sporadic amyotrophic lateral sclerosis patients induces mitochondrial and lysosomal dysfunction. Neurochem Res. (2016) 41:965–84. doi: 10.1007/s11064-015-1779-7
112. Duval, N, Sumner, WA, Andrianakos, AG, Gray, JJ, Bouchard, RJ, Wilkins, HM, et al. The Bcl-2 Homology-3 domain (Bh3)-only proteins, bid, Dp5/Hrk, and Bnip3l, are upregulated in reactive astrocytes of end-stage mutant Sod1 mouse spinal cord. Front Cell Neurosci. (2018) 12:15. doi: 10.3389/fncel.2018.00015
113. Guo, X, Zhang, Z, Gu, J, Ke, P, Liu, J, Meng, Y, et al. Fudnc1-dependent Mitophagy ameliorate motor neuron death in an amyotrophic lateral sclerosis mouse model. Neurobiol Dis. (2024) 197:106534. doi: 10.1016/j.nbd.2024.106534
114. Davis, SA, Itaman, S, Khalid-Janney, CM, Sherard, JA, Dowell, JA, Cairns, NJ, et al. Tdp-43 interacts with mitochondrial proteins critical for Mitophagy and mitochondrial dynamics. Neurosci Lett. (2018) 678:8–15. doi: 10.1016/j.neulet.2018.04.053
115. Jurcau, A. Molecular pathophysiological mechanisms in Huntington's disease. Biomedicine. (2022) 10:1432. doi: 10.3390/biomedicines10061432
116. Legleiter, J. Membranes as targets and modifiers of mutant huntingtin aggregation. Trends Biochem Sci. (2025) S0968–4. doi: 10.1016/j.tibs.2025.08.005
117. Chang, CP, Wu, CW, and Chern, Y. Metabolic dysregulation in Huntington's disease: neuronal and glial perspectives. Neurobiol Dis. (2024) 201:106672. doi: 10.1016/j.nbd.2024.106672
118. Franco-Iborra, S, Plaza-Zabala, A, Montpeyo, M, Sebastian, D, Vila, M, and Martinez-Vicente, M. Mutant Htt (huntingtin) impairs Mitophagy in a cellular model of Huntington disease. Autophagy. (2021) 17:672–89. doi: 10.1080/15548627.2020.1728096
119. Šonský, I, Vodička, P, Vodičková Kepková, K, and Hansíková, H. Mitophagy in Huntington's disease. Neurochem Int. (2021) 149:105147. doi: 10.1016/j.neuint.2021.105147
120. Sassone, J, Colciago, C, Marchi, P, Ascardi, C, Alberti, L, di Pardo, A, et al. Mutant huntingtin induces activation of the Bcl-2/adenovirus E1b 19-Kda interacting protein (Bnip3). Cell Death Dis. (2010) 1:e7. doi: 10.1038/cddis.2009.6
121. Sassone, F, Margulets, V, Maraschi, A, Rodighiero, S, Passafaro, M, Silani, V, et al. Bcl-2/adenovirus E1b 19-Kda interacting protein (Bnip3) has a key role in the mitochondrial dysfunction induced by mutant huntingtin. Hum Mol Genet. (2015) 24:6530–9. doi: 10.1093/hmg/ddv362
122. Signorile, A, Sgaramella, G, Bellomo, F, and De Rasmo, D. Prohibitins: a critical role in mitochondrial functions and implication in diseases. Cells. (2019) 8:71. doi: 10.3390/cells8010071
123. Liu, J, and Mouradian, MM. Pathogenetic contributions and therapeutic implications of transglutaminase 2 in neurodegenerative diseases. Int J Mol Sci. (2024) 25:2364. doi: 10.3390/ijms25042364
124. Charabati, M, Wheeler, MA, Weiner, HL, and Quintana, FJ. Multiple sclerosis: Neuroimmune crosstalk and therapeutic targeting. Cell. (2023) 186:1309–27. doi: 10.1016/j.cell.2023.03.008
125. Al-Kuraishy, HM, Jabir, MS, Al-Gareeb, AI, Saad, HM, Batiha, GE, and Klionsky, DJ. The beneficial role of autophagy in multiple sclerosis: yes or no? Autophagy. (2024) 20:259–74. doi: 10.1080/15548627.2023.2259281
126. Barcelos, IP, Troxell, RM, and Graves, JS. Mitochondrial dysfunction and multiple sclerosis. Biology. (2019) 8:37. doi: 10.3390/biology8020037
127. Patergnani, S, Fossati, V, Bonora, M, Giorgi, C, Marchi, S, Missiroli, S, et al. Mitochondria in multiple sclerosis: molecular mechanisms of pathogenesis. Int Rev Cell Mol Biol. (2017) 328:49–103. doi: 10.1016/bs.ircmb.2016.08.003
128. Patergnani, S, Bonora, M, Ingusci, S, Previati, M, Marchi, S, Zucchini, S, et al. Antipsychotic drugs counteract autophagy and Mitophagy in multiple sclerosis. Proc Natl Acad Sci USA. (2021) 118:e2020078118. doi: 10.1073/pnas.2020078118
129. Cossu, D, Yokoyama, K, Sechi, LA, and Hattori, N. Potential of Pink1 and Parkin proteins as biomarkers for active multiple sclerosis: a Japanese cohort study. Front Immunol. (2021) 12:681386. doi: 10.3389/fimmu.2021.681386
130. Fashina, IA, McCoy, CE, and Furney, SJ. In silico prioritisation of microrna-associated common variants in multiple sclerosis. Hum Genomics. (2023) 17:31. doi: 10.1186/s40246-023-00478-4
131. De Rasmo, D, Ferretta, A, Russo, S, Ruggieri, M, Lasorella, P, Paolicelli, D, et al. Pbmc of multiple sclerosis patients show deregulation of Opa1 processing associated with increased Ros and Phb2 protein levels. Biomedicine. (2020) 8:85. doi: 10.3390/biomedicines8040085
132. Gharagozloo, M, Mahmoud, S, Simard, C, Yamamoto, K, Bobbala, D, Ilangumaran, S, et al. Nlrx1 inhibits the early stages of Cns inflammation and prevents the onset of spontaneous autoimmunity. PLoS Biol. (2019) 17:e3000451. doi: 10.1371/journal.pbio.3000451
133. Koo, JH, Kim, DH, Cha, D, Kang, MJ, and Choi, JM. Lrr domain of Nlrx1 protein delivery by Dnp2 inhibits T cell functions and alleviates autoimmune encephalomyelitis. Theranostics. (2020) 10:3138–50. doi: 10.7150/thno.43441
134. He, YL, Li, J, Cao, Y, Wu, HT, and Wu, LY. A crosstalk between phosphorylation and ubiquitination of Bnip3 regulates Mitophagy under hypoxia. Autophagy Rep. (2023) 2:2197637. doi: 10.1080/27694127.2023.2197637
135. Li, H, Bai, X, Yang, T, Wu, C, Liu, Y, Shan, C, et al. Hif-1α activates Mitophagy through Bnip3 to inhibit Ipec-J2 apoptosis under heat stress. J Agric Food Chem. (2025) 73:26621–32. doi: 10.1021/acs.jafc.5c09057
136. Adriaenssens, E, Schaar, S, Cook, ASI, Stuke, JFM, Sawa-Makarska, J, Nguyen, TN, et al. Reconstitution of Bnip3/nix-Mitophagy initiation reveals hierarchical flexibility of the autophagy machinery. Nat Cell Biol. (2025) 27:1272–87. doi: 10.1038/s41556-025-01712-y
137. Liu, R, Xu, C, Zhang, W, Cao, Y, Ye, J, Li, B, et al. Fundc1-mediated Mitophagy and Hif1α activation drives pulmonary hypertension during hypoxia. Cell Death Dis. (2022) 13:634. doi: 10.1038/s41419-022-05091-2
138. Chen, D, Zhou, L, Chen, G, Lin, T, Lin, J, Zhao, X, et al. Fundc1-induced Mitophagy protects spinal cord neurons against ischemic injury. Cell Death Discov. (2024) 10:4. doi: 10.1038/s41420-023-01780-9
139. Guan, H, Yang, H, Yang, M, Yanagisawa, D, Bellier, JP, Mori, M, et al. Mitochondrial ferritin protects sh-sy5y cells against H2O2-induced oxidative stress and modulates Α-synuclein expression. Exp Neurol. (2017) 291:51–61. doi: 10.1016/j.expneurol.2017.02.001
140. Yuan, Y, He, Q, Yang, X, Flores, JJ, Huang, L, Luo, X, et al. Mitochondrial ferritin upregulation reduced oxidative stress and blood-brain-barrier disruption by maintaining cellular Iron homeostasis in a neonatal rat model of germinal matrix hemorrhage. Exp Neurol. (2024) 374:114703. doi: 10.1016/j.expneurol.2024.114703
141. Gao, G, and Chang, YZ. Mitochondrial ferritin in the regulation of brain Iron homeostasis and neurodegenerative diseases. Front Pharmacol. (2014) 5:19. doi: 10.3389/fphar.2014.00019
142. Di Rienzo, M, Romagnoli, A, Refolo, G, Vescovo, T, Ciccosanti, F, Zuchegna, C, et al. Role of Ambra1 in Mitophagy regulation: emerging evidence in aging-related diseases. Autophagy. (2024) 20:2602–15. doi: 10.1080/15548627.2024.2389474
143. Wang, J, Chen, A, Xue, Z, Liu, J, He, Y, Liu, G, et al. Bcl2l13 promotes Mitophagy through Dnm1l-mediated mitochondrial fission in glioblastoma. Cell Death Dis. (2023) 14:585. doi: 10.1038/s41419-023-06112-4
144. D'Arcy, MS. Mitophagy in health and disease. Molecular mechanisms, regulatory pathways, and therapeutic implications. Apoptosis. (2024) 29:1415–28. doi: 10.1007/s10495-024-01977-y
145. Nikhil, K, and Shah, K. The Cdk5-mcl-1 axis promotes mitochondrial dysfunction and neurodegeneration in a model of Alzheimer's disease. J Cell Sci. (2017) 130:3023–39. doi: 10.1242/jcs.205666
146. Isei, MO, Crockett, M, Chen, E, Rodwell-Bullock, J, Caroll, T, Girardi, PA, et al. Tau phosphorylation suppresses oxidative stress-induced mitophagy via Fkbp8 receptor modulation. bioRxiv. (2024). doi: 10.1101/2024.07.05.602170
147. Yoo, SM, Yamashita, SI, Kim, H, Na, D, Lee, H, Kim, SJ, et al. Fkbp8 Lirl-dependent mitochondrial fragmentation facilitates Mitophagy under stress conditions. FASEB J. (2020) 34:2944–57. doi: 10.1096/fj.201901735R
148. Wei, Y, Chiang, WC, Sumpter, R Jr, Mishra, P, and Levine, B. Prohibitin 2 is an inner mitochondrial membrane Mitophagy receptor. Cell. (2017) 168:224–38.e10. doi: 10.1016/j.cell.2016.11.042
149. Bernstein, HG, Smalla, KH, Keilhoff, G, Dobrowolny, H, Kreutz, MR, and Steiner, J. The many "Neurofaces" of Prohibitins 1 and 2: crucial for the healthy brain, dysregulated in numerous brain disorders. J Chem Neuroanat. (2023) 132:102321. doi: 10.1016/j.jchemneu.2023.102321
150. Stokman, G, Kors, L, Bakker, PJ, Rampanelli, E, Claessen, N, Teske, GJD, et al. Nlrx1 dampens oxidative stress and apoptosis in tissue injury via control of mitochondrial activity. J Exp Med. (2017) 214:2405–20. doi: 10.1084/jem.20161031
151. Rios, KE, Zhou, M, Lott, NM, Beauregard, CR, McDaniel, DP, Conrads, TP, et al. Card19 interacts with mitochondrial contact site and cristae organizing system constituent proteins and regulates cristae morphology. Cells. (2022) 11:1175. doi: 10.3390/cells11071175
152. 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) 6:76–93. doi: 10.1038/s41551-021-00819-5
153. Chen, M, Peng, L, Gong, P, Zheng, X, Sun, T, Zhang, X, et al. Baicalein mediates mitochondrial autophagy via Mir-30b and the nix/Bnip3 signaling pathway in Parkinson's disease. Biochem Res Int. (2021) 2021:2319412. doi: 10.1155/2021/2319412
154. Lin, MW, Lin, CC, Chen, YH, Yang, HB, and Hung, SY. Celastrol inhibits dopaminergic neuronal death of Parkinson's disease through activating Mitophagy. Antioxidants (Basel). (2019) 9:37. doi: 10.3390/antiox9010037
155. Wang, SY, Li, MM, Wang, L, Pan, J, Sun, Y, Wu, JT, et al. Schisandra Chinensis (Turcz.) Baill neutral polysaccharides alleviate Parkinson's disease via effectively activating mcl-1 expression regulation of autophagy signaling. Int J Biol Macromol. (2024) 279:134952. doi: 10.1016/j.ijbiomac.2024.134952
156. Tao, G, Wang, X, Wang, J, Ye, Y, Zhang, M, Lang, Y, et al. Dihydro-resveratrol ameliorates Nlrp3 Inflammasome-mediated Neuroinflammation via Bnip3-dependent Mitophagy in Alzheimer's disease. Br J Pharmacol. (2025) 182:1005–24. doi: 10.1111/bph.17373
157. Xie, Y, Ke, X, Ye, Z, Li, X, Chen, Z, Liu, J, et al. Se-Methylselenocysteine ameliorates mitochondrial function by targeting both Mitophagy and autophagy in the mouse model of Alzheimer's disease. Food Funct. (2024) 15:4310–22. doi: 10.1039/d4fo00520a
158. Naeem, A, Waseem, A, Khan, MA, Robertson, AA, and Raza, SS. Therapeutic potential of Mcc950 in restoring autophagy and cognitive function in Stz-induced rat model of Alzheimer's disease. Mol Neurobiol. (2025) 62:6041–58. doi: 10.1007/s12035-024-04662-y
159. Chen, Y, Wang, Y, Qin, Q, Zhang, Y, Xie, L, Xiao, J, et al. Carnosic acid ameliorated aβ-mediated (amyloid-Β peptide) toxicity, cholinergic dysfunction and mitochondrial defect in Caenorhabditis elegans of Alzheimer's model. Food Funct. (2022) 13:4624–40. doi: 10.1039/d1fo02965g
160. Lai, X, Zhang, Y, Wu, J, Shen, M, Yin, S, and Yan, J. Rutin attenuates oxidative stress via Phb2-mediated Mitophagy in Mpp(+)-induced Sh-Sy5y cells. Neurotox Res. (2023) 41:242–55. doi: 10.1007/s12640-023-00636-5
161. Yu, W, Wang, L, Ren, WY, Xu, HX, Wu, NN, Yu, DH, et al. Sglt2 inhibitor Empagliflozin alleviates cardiac remodeling and contractile anomalies in a Fundc1-dependent manner in experimental Parkinson's disease. Acta Pharmacol Sin. (2024) 45:87–97. doi: 10.1038/s41401-023-01144-0
162. Wang, XL, Feng, ST, Wang, YT, Zhang, NN, Guo, ZY, Yan, X, et al. Mangiferin, a natural Glucoxilxanthone, inhibits mitochondrial dynamin-related protein 1 and relieves aberrant Mitophagic proteins in mice model of Parkinson's disease. Phytomedicine. (2022) 104:154281. doi: 10.1016/j.phymed.2022.154281
163. Wang, DX, Yang, Y, Huang, XS, Tang, JY, Zhang, X, Huang, HX, et al. Pramipexole attenuates neuronal injury in Parkinson's disease by targeting Mir-96 to activate Bnip3-mediated Mitophagy. Neurochem Int. (2021) 146:104972. doi: 10.1016/j.neuint.2021.104972
164. Yan, J, Sun, W, Shen, M, Zhang, Y, Jiang, M, Liu, A, et al. Idebenone improves motor dysfunction, learning and memory by regulating mitophagy in MPTP-treated mice. Cell Death Discov. (2022) 8:28. doi: 10.1038/s41420-022-00826-8
165. Prados, ME, Correa-Sáez, A, Unciti-Broceta, JD, Garrido-Rodríguez, M, Jimenez-Jimenez, C, Mazzone, M, et al. Betulinic acid Hydroxamate is neuroprotective and induces protein phosphatase 2a-dependent Hif-1α stabilization and post-transcriptional Dephosphorylation of prolyl hydrolase 2. Neurotherapeutics. (2021) 18:1849–61. doi: 10.1007/s13311-021-01089-4
Keywords: neurodegenerative diseases, PINK1/Parkin-independent mitophagy, autophagy receptors, mitochondria, mitophagy, mitochondrial dysfunction
Citation: Yang J, Yang F, Chen G, Liu M, Yuan S and Zhang T-e (2025) Receptor-mediated mitophagy: a new target of neurodegenerative diseases. Front. Neurol. 16:1665315. doi: 10.3389/fneur.2025.1665315
Edited by:
Murali Vijayan, Texas Tech University College of Education, United StatesReviewed by:
Simone Patergnani, University of Ferrara, ItalyPiao Zhao, Sichuan Agricultural University, China
Copyright © 2025 Yang, Yang, Chen, Liu, Yuan and Zhang. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.
*Correspondence: Shiqing Yuan, eXVhbnNoaXFpbmdAY2R1dGNtLmVkdS5jbg==; Tian-e Zhang, emh0ZTIwMDNAY2R1dGNtLmVkdS5jbg==
Fuquan Yang2