Open Access

Beyond self‑eating: Emerging autophagy‑independent functions for the autophagy molecules in cancer (Review)

  • Authors:
    • Giulia Tedesco
    • Manuela Santarosa
    • Roberta Maestro
  • View Affiliations

  • Published online on: April 10, 2024     https://doi.org/10.3892/ijo.2024.5645
  • Article Number: 57
  • Copyright: © Tedesco et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Autophagy is a conserved catabolic process that controls organelle quality, removes misfolded or abnormally aggregated proteins and is part of the defense mechanisms against intracellular pathogens. Autophagy contributes to the suppression of tumor initiation by promoting genome stability, cellular integrity, redox balance and proteostasis. On the other hand, once a tumor is established, autophagy can support cancer cell survival and promote epithelial‑to‑mesenchymal transition. A growing number of molecules involved in autophagy have been identified. In addition to their key canonical activity, several of these molecules, such as ATG5, ATG12 and Beclin‑1, also exert autophagy‑independent functions in a variety of biological processes. The present review aimed to summarize autophagy‑independent functions of molecules of the autophagy machinery and how the activity of these molecules can influence signaling pathways that are deregulated in cancer progression.

1. Introduction

Macroautophagy, hereafter referred to as autophagy, is a catabolic process where cytoplasmic materials are sequestered by double-membrane-bound vesicles, called autophagosomes, to be degraded via fusion with lysosomes. Autophagy, from the Greek autóphagos, meaning 'self-eating', is a process that is highly conserved from yeast to humans and is necessary for maintenance of cellular homeostasis under nutrient deprivation and other stress conditions (1). It is also involved in physiological processes such as defense against intracellular pathogens, organelle quality control and removal of misfolded or aggregated proteins (2).

Numerous studies demonstrate that autophagy acts as a double-edged sword in cancer. Namely, it can inhibit tumor initiation by removing damaged proteins and organelles and preventing genome instability (3-6). Tissue-specific deletion of key autophagic molecules (such as autophagy related genes ATG7 and ATG5) restricts cancer development in several mouse models of inducible cancer such as melanoma and pancreatic cancer (7-12). On the other hand, when a tumor has already developed, autophagy can support cancer cell survival under stressful conditions such as hypoxia and metabolic stress, promoting the persistence of tumor cells in hostile environment (13-16). Autophagy affects anchorage to the extracellular matrix, cytoskeletal remodeling and epithelial-to-mesenchymal transition (EMT), further supporting its involvement in cancer progression (17).

The autophagy machinery consists of a number of molecules, involved in the different steps of autophagy: Initiation, nucleation, elongation, autophagosome-lysosome fusion and degradation of substrates (18). A group of ~20 molecules, called autophagy-related genes, was initially discovered through genetic studies in yeast and found to be necessary for the control of several key phases of the autophagy process (19,20). The group of autophagic molecules has been studied in humans and gradually expanded to include other proteins; The Autophagy Project of the BioGRID repository contains information on ~200 human proteins involved in autophagy (thebiogrid.org/project/6/autophagy.html; accessed on October 2023) (21) (Table SI).

Numerous autophagy machinery molecules (AMMs) have been reported to extend their functions beyond autophagy. Non-canonical autophagy-related processes have been described (22-25). For example, molecules involved in autophagic vesicle elongation are also involved in a form of exocytosis termed secretory autophagy (22). In addition, there is increasing evidence that individual AMMs exert autophagy-independent functions in various types of diseases, including cancer (23-25). The present review aimed to discuss the autophagy-independent functions of AMMs with a particular focus on cancer progression.

2. Autophagic cascade

The steps involving AMMs in the typical autophagic cascade have been explored (26). The present review briefly outlines the autophagic role of those AMMs that have also been reported to have autophagy-independent functions.

Nutrient shortage and stress conditions are the main triggers for the autophagy process (Fig. 1). One of the key sensors of energy, nutrient and redox status is the mTORC1 protein complex, which consists of the Ser/Thr kinase mTOR and other regulatory components (27). Under nutrient-limited conditions, the amount of ATP decreases and increased AMP/ATP ratio triggers the activation of AMPK, which in turn restrains mTORC1 and its inhibitory activity towards Unc-51-like autophagy-activating kinase 1 (ULK1). ULK1 can form a complex with autophagy related proteins ATG101, ATG13 and RB1CC1 (family kinase-interacting protein 200/RB1 Inducible Coiled-Coil 1), which initiates the autophagy cascade (28) by phosphorylating components of PI3K complex I (PIK3C3, Beclin-1, ATG14 and PIK3R4) (29). This complex is essential for the nucleation phase of autophagy. Activated PI3K complex I phosphorylates phosphatidylinositol (PI) to form PI-3-phosphate (PI3P) (29), which binds to the nascent phagophore membrane (30). PI3K complex I is positively regulated by the ultraviolet radiation resistance-associated gene (UVRAG) (31) and autophagy and Beclin-1 regulator 1 (AMBRA1) (32). The process is supported by ATG9, a lipid scramblase that is incorporated into vesicles involved in the nucleation of phagophores and subsequently assists the elongation process (33,34). Two ubiquitin-like conjugation systems are then activated: The phagophore elongation complexes ATG5-ATG12-ATG16L1 and the LC3 system. The ATG5-ATG12-ATG16L1 complex is formed by a reaction cascade involving ATG7 (E1-like enzyme) and ATG10 (E2-like enzyme), which mediate covalent binding between ATG5 and ATG12. Subsequently, the ATG5-ATG12 conjugate binds ATG16L1 and forms a ternary complex located at the autophagosomal membrane (35). Studies propose an alternative model for the formation of the ATG5-ATG12-ATG16L1 complex, which first requires an interaction between ATG5 and ATG16L1. Then, the transient ATG5-ATG16L1 duplet allows recruitment of ATG12 and the formation of a stable trimeric structure via formation of a covalent bond between ATG12 and ATG5 (36-38). The ATG5-ATG12-ATG16L1 complex serves as a scaffold and promotes LC3 lipidation (35). Microtubule-associated protein 1-light chain 3 (MAP1LC3) is the ortholog of Atg8 in yeast. LC3 is first cleaved at its carboxy terminus by ATG4 to form LC3 I. Following ATG4-mediated cleavage, LC3 is activated by ATG7 (E1-like enzyme) and ATG3 (E2-like enzyme) and finally conjugated to phosphatidylethanolamine (PE) to form the active LC3 (LC3-II) (39). Lipidated LC3, together with the ATG5-ATG12-ATG16L1 complex, enables elongation of the autophagic phagophore membrane (40).

The nascent phagophore sequesters specific cargo material via simultaneous interaction between LC3-II molecules and cargo receptors such as sequestrosome-1 (SQSTM1 or p62), toll-interacting Protein), and neighbor Of BRCA1 Gene 1) (41). SQSTM1 oligomerizes via its PB1 domain and forms filaments that interact with polyubiquitinated cargoes and LC3-II via LC3-interacting regions. These interactions enable autophagy-mediated degradation of specific cargo material (42).

In the late stages of the autophagic process, the phagophore closes and fuses with the lysosome to form the autophagolysosome. This phase relies on soluble N-ethylmaleimide-sensitive factor attachment protein receptor (SNARE) proteins, which are found in both membranes (43). The activity of the two known SNARE complexes (TX17-SNAP29-VAMP7/VAMP812 and STX7-SNAP29-YKT6 complexes) is facilitated by the tethering factors such as the homotypic fusion and protein sorting) complex, pleckstrin Homology And RUN Domain Containing M1) and EPG5 (Ectopic P-Granules 5 Autophagy Tethering Factor), which promote close interaction between the membranes. In the autophagolysosome, acidic hydrolases degrade sequestered material and generate metabolites that are released into the cytoplasm (44). The nutrients obtained via the autophagy pathway stimulate mTOR activation. A negative feedback mechanism stops autophagy when availability of nutrients is restored (44).

3. Autophagy-independent role of AMMs and cancer

During tumor transformation and progression, several biological functions are altered (45). Cancer cells acquire genome instability, which gives them selective advantages. The aggressiveness of the transformed clones is characterized by sustained proliferation and death resistance. Cancer cells have invasive behavior determined by the activation of molecular invasion programs and the ability to control the tumor microenvironment by sending signals to surrounding cells, including immune cells (45). Increasing evidence suggests that AMMs may serve functions that are not exclusive to lysosomal degradation of autophagy substrates (22-24). AMMs are involved in the processes of cancer initiation and progression (Table I).

Table I

Autophagy-independent functions of the autophagy machinery.

Table I

Autophagy-independent functions of the autophagy machinery.

AMMAutophagic functionCancer-associated function(Refs.)
AMBRA1PI3K complex I positive regulationMYC and cyclin D regulation involved in G1/S transition(55)
Induction of apoptosis via BH3-like domain(66)
ATG3LC3 activationControl of mitochondrial fission and fusion and apoptotic cell death(71)
ATG4BLC3 priming for lipidationmTOR phosphorylation and promotion of G1/S phase transition(56)
Mitochondrial respiration (mitochondrial function impairment and Warburg effect)(73)
ATG4DLC3 activation/inactivationInduction of apoptosis via BH3-like domain(67)
ATG5Phagophore elongationInteraction with ATP6V1E1 causing exosome production(24,92)
Clathrin membrane regulation with endocytic trafficking(37)
Interacts with Mis18α inducing microsatellite instability(53)
Interaction with AuroraB and induction of mitotic catastrophe(54)
Cleaved N-terminal portion inducer of apoptosis(68)
ERK activation(83)
ATG7ATG12 activation for conjugation with ATG5Binding with p53 and DNA damage-driven cell cycle arrest(57)
ATG9BAutophagosomal membrane expansionStabilization of MYH9 and boost of focal adhesion formation(89)
ATG12Phagophore elongationClathrin membrane regulation with endocytic trafficking(37)
Controls mitochondrial fission and fusion and apoptotic cell death(38,69,71)
Block of oncosis(72)
ERK activation(83)
ATG16L1Phagophore elongationInteracts with Rab33A for vesicular release(91)
Beclin-1Autophagic nucleationControls STAT3 downstream signaling(25)
(PI3K complex I component)Controls NHEJ and regulation of centrosome stability(50)
Kinetocore stability by interaction with KMN complex for mitotic progression(58)
Nuclear localization for cytokinesis completion(59)
Cleaved C-terminal portion inducer of apoptosis(61,64,65)
Growth factor receptor endosomal signaling(84)
Controls E-cadherin and α-catenin membrane localization(85)
Regulation of tight junction permeability via endocytosis of occludin(86)
LC3B/LC3Phagophore elongationInteracts with ATP6V1E1 causing exosome production(24,92)
Apoptosis and anoikis induction(76)
ERK activation(83)
Regulates Rho signaling and actin stress fiber formation(88)
LC3-associated phagocytosis(97)
ULK1Autophagy initiation complexTNF-induced cell death, PARP regulation and necrosis(74,75)
UVRAGPI3K complex I positive regulationChromosome stability and DNA repair(48-50,52)

[i] AMM, autophagy machinery molecule; AMBRA1, autophagy and Beclin-1 regulator 1; ATG, autophagy related genes; ULK1, Unc-51-like autophagy-activating kinase 1; UVRAG, ultraviolet radiation resistance-associated gene; ATP6V1E1, ATPase H+ Transporting V1 Subunit E1; NHEJ, Non-Homologous End Joining; KMN, KNL-1/Mis12/Ndc80; Rab33A, Member RAS Oncogene Family 33A; MYH9, myosin heavy chain 9; Mis18α, MIS18 Kinetochore Protein A.

4. Non-autophagic functions of AMMs in genome stability and cell proliferation

Normal cells control DNA integrity to ensure genome stability. Following DNA damage, cell cycle progression is delayed or blocked by a number of cell cycle control mechanisms to allow repair of DNA damage and prevent abnormal cell division (45).

Impaired autophagy has been recognized as a trigger for chromosome instability (46). In addition, numerous AMMs have been shown to play a role in genomic stability independent of autophagy (Fig. 2). Some of these proteins were originally identified because of their function in maintaining genome stability rather than their link to autophagy. For example, UVRAG was originally discovered because of its involvement in nucleotide excision repair (NER) (47). UVRAG interacts with the UV sensor damage Specific DNA Binding Protein 1) and mediates the assembly of the NER complex at damaged DNA foci. Accordingly, UVRAG expression is associated with a lower UV mutation rate in cutaneous melanoma, where UV radiation is the main factor for tumorigenesis and progression (48). UVRAG also promotes repair of DNA double-strand breaks by interacting with the DNA-dependent protein kinase complex DNA-PKcs in the non-homologous end joining (NHEJ) DNA repair system (49). In colon cancer model, DNA double-strand breaks repair requires interaction between UVRAG and Beclin-1 to regulate DNA damage response and centrosome stability (50). UVRAG physically associates with the centrosome component centrosomal protein 63) to control proper chromosome segregation and prevent aneuploidy (49). Truncating UVRAG mutations were detected in a significant proportion of colon carcinoma cases with a defective DNA mismatch repair system (51). The truncated UVRAG protein loses the ability to repair DNA and exhibits autophagy-independent oncogenic properties, although conflicting data on autophagy activation have been reported (51,52).

Figure 2

AMMs affecting genome stability and cell cycle progression. At G1/S checkpoint, AMBRA1 and ATG4B interact with cyclins controlling replication stress and genome instability whereas ATG7 interacts with p53 and promotes CDKN1A expression governing G1/S and G2/M checkpoints. A number of AMMs (UVRAG, ATG5 and Beclin-1) intervene in the processes of DNA repair involved in maintaining genome stability throughout the cell cycle. UVRAG interacts with DDB1, a component of the NER complex, to resolve DNA cross-links. ATG5, together with Mis18α, controls expression of MLH1 and impairs the mismatch repair system. UVRAG is involved in the response to double strand breaks by both NHEJ and HR. Beclin-1 interacts with UVRAG to regulate HR. Certain AMMs (Beclin-1, UVRAG, ATG5) interact with kinetochore structures that affect proper chromosomal segregation. Created with BioRender.com. AMM, autophagy machinery molecule; AMBRA1, autophagy and Beclin-1 regulator 1; ATG, Autophagy related genes; CDKN1A, Cyclin Dependent Kinase Inhibitor 1A; UVRAG, ultraviolet radiation resistance-associated gene; DDB1, Damage Specific DNA Binding Protein 1; NER, Nucleotide Excision Repair; Mis18α, MIS18 kinetochore Protein A; MLH1, MutL Homolog 1; NHEJ, Non-Homologous End Joining; HR, homologous Recombination; CEP63, Centrosomal Protein 63; ERCC2, Excision Repair Cross-Complementing Rodent Repair Deficiency, Complementation Group 2; XPC, Xeroderma Pigmentosum, Complementation Group C; DNA PK, Protein Kinase, DNA-Activated; ATM, Ataxia Telangiectasia Mutated; MMR, Mismatch Mediated Repair.

Following treatment with 5-fluorouracil (5-FU), ATG5 translocates to the nucleus independently of its autophagic function and interacts with Mis18α (MIS18 kinetochore Protein A), a protein localized in the centromere and involved in methylation of the underlying chromatin. This binding increases the levels of promoter methylation of MLH1 (MutL Homolog 1) gene (a component of DNA mismatch repair), thereby downregulating MLH1 expression and enhancing mismatch repair defects and resistance to 5-FU (53). Similarly, following DNA-damaging treatment, ATG5 interacts with survivin in the nucleus, disrupting chromosome segregation and triggering an abnormal mitotic process known as mitotic catastrophe (54). This suggests control of cell cycle progression by ATG5 independent of autophagy.

AMBRA1 is another AMM that controls cell cycle progression by mediating degradation of cyclin D, which regulates the G1/S phase transition. A defective AMBRA1/cyclin D axis leads to premature entry into S phase, resulting in replication stress and genome instability (55). In addition, ATG4B downregulation in colorectal cancer has been shown to decrease expression and activity of cyclin D1 (56). The aforementioned study demonstrated an inhibition of mTOR and induction of autophagy in ATG4B-silenced cells; however it is unclear how this fits with the hypothesis that ATG4B can prime LC3B for lipidation and autophagy induction (56). ATG7 promotes CDKN1A (p21) expression and cell cycle arrest by directly binding to p53, a master keeper of cell cycle, apoptosis and CDKN1A (p21) expression. The effect of ATG7 on this process is enhanced by nutrient starvation, which is known to stimulate autophagy (57). Nevertheless, the E1-like enzymatic activity of ATG7, which is central to its autophagic involvement, is not required for this cell cycle arrest (57).

The component of the PI3K complex Beclin-1 is crucial for mitotic progression as it interacts with the KNL-1/Mis12/Ndc80 complex involved in the precise anchoring of the kinetochore to the mitotic spindle (58). Moreover, a variant of the PI3K complex, which includes Bax-Interacting Factor 1), is involved in cytokinesis, and thus exerts a tumor suppressor function distinct from its role in the early steps of autophagy (59). PI3P generated by the activated PI3K complex mediates contact with proteins of the centrosome and regulates completion of cytokinesis (60).

5. Unconventional role of AMMs in cell death and survival

Autophagy is a multifaceted process that promotes either cell survival or death, depending on the physiological state of the cell and environmental conditions. Autophagy and apoptosis are closely associated processes in which AMMs can be activated by apoptotic factors and vice versa (61). For example, the autophagosome membrane and its associated autophagy machinery serve as a platform for recruitment and activation of the apoptotic caspase cascade (62). On the other hand, proapoptotic caspase-9 has been shown to interact with ATG7 and contribute to autophagy in various human cancer cell lines (63).

Under certain circumstances, autophagy and apoptosis appear to be mutually exclusive processes mediated by common players (Fig. 3). Protease-mediated cleavage of certain AMMs inhibits autophagy and promotes apoptosis. For example, under unfavorable conditions, such as cell starvation and drug treatment, cleavage of Beclin-1 by caspase-3 or caspase-8 results in formation of an autophagy-impaired Beclin-1 fragment that localizes to mitochondria and promotes apoptosis (61,64,65).

AMBRA1 carries a BH3 motif that, after being released by caspase-mediated cleavage, binds and blocks BCL2, one of the key inhibitors of apoptosis (66). Overexpression of ATG4D and a form of ATG4D cleaved by caspase-3 leads to their recruitment to mitochondria, where they contribute to apoptosis. Again, this pro-apoptotic function of ATG4D relies on its C-terminal BH3 domain, which specifically interacts with members of the BCL2 family (67). Similarly, ATG5 is specifically cleaved by calpains 1/2 during apoptosis, independently of cell type and apoptotic stimulus. Truncated ATG5 translocates to the mitochondria, associates with BCL2-like protein 1 (BCL2L1) and triggers caspase activation via an autophagy-independent mechanism (68). The ATG5 partner ATG12 can control apoptosis independently of the other AMMs by interacting with BCL2 protein family via the BH3 motif (69). ATG12 is an unstable protein that, when not conjugated to ATG5, is subject to proteasomal degradation (36). Due to aberrant proteasomal blockade, ATG12 accumulates in osteosarcoma cells, antagonizes BCL2 and activates apoptosis (38). In colorectal cancer cell lines, oncogenic Ras promotes cancer cell survival by decreasing ATG12 levels and thus ATG12-mediated inhibition of apoptosis (70). ATG12 also serves a role in the control of mitochondrial homeostasis. It interacts with ATG3 in a complex that does not affect autophagy, but is critical for controlling mitochondrial fission and fusion and regulating the function of mitochondria-mediated cell death pathways (71).

Not only apoptosis, but also other forms of cell death are influenced by AMMs. Lung and breast cancer cell lines in which ATG12 expression is reduced undergo oncosis, a caspase-independent cell death triggered by energy deficiency (72). In this condition, the imbalance of mitochondrial ions and impaired metabolism cause changes in osmotic pressure, leading to organelle swelling and cytoplasmic blebs that disrupt cellular function. Similarly, Ni et al (73) identified a novel phosphorylation site in ATG4B that, once phosphorylated, allows binding with the soluble catalytic core F1, and the membrane-spanning component, Fo, subunits of ATP synthase. This interaction results in impaired mitochondrial function, which leads to an increase in mitochondrial reactive oxygen species (ROS) and metabolic reprogramming of hepatocellular carcinoma cells towards the Warburg effect (73). Following autophagy-mediated activation, ULK1 can phosphorylate RIPK1, a component of the TNF receptor (TNFR)-mediated cell death complex, thereby improving survival of mouse embryonic fibroblasts (MEFs) (74). However, stress conditions, such as increased ROS species, relocate ULK1 to the nucleus, limiting the autophagic response and allowing nuclear ULK1 to promote PARP1-dependent necrosis (75). Similarly, pharmacological induction of ROS increases LC3B production without activating autophagic flux and enhances both expression of apoptotic molecules and induction of anoikis (76).

6. AMMs acting beyond autophagy in metastasis and immune microenvironment

The majority of cancer deaths are caused by metastasis. The metastatic process begins when cancer cells leave the primary neoplasm, invade the surrounding matrix and colonize other tissue via the bloodstream and lymphatic system. These processes, combined with the uncontrolled proliferative capacity of cancer cells, lead to the destruction of the physiological functions of distant organs (77). At the molecular level, invasive and metastatic behavior is supported by features of EMT, which is an embryonic molecular program that is abnormally activated in tumor cells (78). Several studies have demonstrated the role of autophagy in EMT (79-82). Nevertheless, AMMs have also been reported to have non-autophagic functions in EMT (25,83-85),(Fig. 4).

The metastatic potential of cancer cells is ensured by intracellular processes that enable cells to survive under stress conditions. Autophagic membranes carrying LC3-II and ATG5-ATG12 conjugates serve as scaffolds for ERK pathway activation (83). In colorectal cancer, B1 integrin can promote c-Met internalization and ERK1/2 activation, allowing cancer cells to survive to anoikis (84). c-Met and B1 integrin colocalize on autophagic membranes for their pro-survival signals and require LC3-II, Beclin-1 and ATG5 for this purpose, but not other canonical autophagy mediators. Loss of Beclin-1 impairs endosomal signaling and results in prolonged ERK and AKT activation, leading to migratory and invasive behavior in breast cancer (84). Similarly Beclin-1 suppresses cell migration in colorectal cancer cells by interacting with transcription factor STAT3 (which is abnormally activated in numerous types of cancer) and blocking its phosphorylation by JAK2 (25).

In addition, Beclin-1 and several other AMMs influence cytoskeletal dynamics and cell-cell adhesion. Beclin-1 promotes membrane localization of the adhesion molecules E-cadherin and α-catenin in breast cancer (85). The aforementioned study also suggested a contribution of UVRAG to control of membrane localization of E-cadherin, but the molecular mechanism needs further investigation. Similarly, Beclin-1 localizes to the cell membrane surface and mediates endocytosis of tight junction protein occludin. However, it is not clear whether the downregulation of occludin mediated by Beclin-1 is dependent on autophagy (86). Autophagy mediates the degradation of E-cadherin (87).

LC3 has been reported to regulate cytoskeletal dynamics by interacting with the selective Rho-A exchange factor AKP13 and regulating Rho family of GTPases-dependent reorganization of the actin cytoskeleton (88). ATG9B is involved in regulation of cell-matrix contacts and invasiveness. In colorectal cancer, it serves a non-autophagic function that contributes to the formation of focal adhesions and promotes metastasis (89). In this context, the interaction between ATG9B and myosin heavy chain gene (MYH9) increases the stability of both proteins by preventing their degradative ubiquitination. This favors the interaction between ATG9B, integrin B1 and talin 1, two key molecules of focal adhesions. Immunohistochemical data have confirmed that high expression of ATG9B and MYH9 is associated with poor prognosis in colorectal carcinoma (89).

Endocytosis and exocytosis can be used by tumor cells to create the favorable microenvironment they need for aberrant behavior (90). The ATG5-ATG12 complex is involved in the clathrin membrane trafficking system that affects endocytosis under both normal and starvation conditions in MEFs (37). ATG16L1 has been proposed as a key regulator of several steps of the secretory machinery (especially vesicular release). Its interaction with small GTPase Rab33A has been shown to be key in the process of hormone secretion and may be an hallmark of neuroendocrine tissue (91).

Exosome release and secretory autophagy (SA) are two pathways that mediate secretion and require a number of AMMs. However, the molecular details of these two processes are not yet fully clarified. In the breast cancer cell line MDA-MB-231, ATG5 acts independently of autophagy to sort LC3 into multivesicular bodies, where it binds a component of the vacuolar ATPase H+ Transporting V1 Subunit E1 and causes a decrease in vesicular acidification (92). The increase in pH promotes the fusion of vesicles with plasma membrane and their release as exosomes. These exosomes have been shown to contain invasion mediators (RAS, β-catenin and vimentin) and thus promote invasion and metastasis of breast cancer cells in mice (93). SA involves unconventional release of molecules into the extracellular space to affect the tumor microenvironment and is used for molecules that cannot enter the conventional endoplasmic reticulum-Golgi secretion system because they lack a signal peptide (93). In bladder cancer, cathepsin-B is released into the tumor microenvironment via SA and stimulates endothelial cells to undergo angiogenesis (94). Elevated levels of cathepsin-B are associated with invasiveness, metastasis and poor prognosis in bladder cancer (94).

In melanoma, SA is activated by pharmacological stimuli and mediates secretion of plasminogen activator inhibitor (PAI-1), which is involved in the formation of a pro-tumor immune microenvironment (95). Moreover, cancer-associated fibroblasts in head and neck cancer use part of the autophagic machinery to secrete tumor-promoting cytokines (IL-6 and IL-8) (96). Whether this mechanism of secretion is SA is not clear.

LC3-associated phagocytosis (LAP) is a process that generates anti-inflammatory and immunosuppressive signals that lead to immune tolerance. Studies show that LAP is involved in M2 macrophage polarization and helps to promote an immunosuppressive environment that favors tumor growth (97,98).

Conversely, in patients with colorectal cancer, expression of an ATG16L1 variant (T600A) is responsible for an increase in IFN-I levels (99). Via the mitochondrial antiviral signaling pathway, cancer cells produce IFN-I, which promotes host antitumor immunity and inhibits the proliferation and metastasis of cancer cells (99).

7. microRNAs (miRNAs or miRs) in the control of AMMs

Expression of critical AMMs is regulated by miRNAs, which are also involved in carcinogenesis (100). miRNAs are a class of small non-coding RNAs (20-24 nucleotides) that control gene expression primarily by either inhibiting the translation or promoting decay of target mRNAs (101). Downregulation of several miRNAs has been shown to promote both tumor progression and autophagy by targeting AMMs (102,103). This is the case for a number of miRNAs that directly target core autophagy molecules such as ATG5 (miR-137, miR-153-3p), ATG12 (miR-30a-3p and miR-214), ATG7 (miR-138-5p and miR-375) and Beclin-1 (miR-17-5p, miR-26a, miR-30a, miR-124-3p, miR-216a and miR-409-3p) (104-114). The downregulation of these miRNAs relieves both oncogenic signaling pathways and autophagy that usually are inhibited by them. This suggests a link between autophagy and cancer progression.

8. Diagnostic and prognostic role of AMMs in cancer

As aforementioned, AMMs play a crucial role in cancer, both dependent on autophagy and independent of it. Therefore, changes in the expression of AMMs can be associated with the prognosis of patients with cancer. Molecular AMM signatures with potential diagnostic and prognostic value have been defined in triple-negative breast cancer and sarcoma (115,116).

Moreover, several studies have investigated the prognostic role of the core LC3 family nuclear proteins and shown that their expression is associated with poor prognosis in various cancers such as lung, breast, gastric and other types of carcinomas (117-122).

Similarly, a signature based on high SQSTM1 and LC3 levels has been considered a negative prognostic factor in squamous cell carcinoma (123). It has been frequently observed that SQSTM1 exerts a pro-tumorigenic function in cancer (124,125). In a meta-analysis, SQSTM1 was shown to serve a negative prognostic role in a number of solid tumors (126). However, it is worth noting that these AMMs are typically degraded by active autophagy and their expression is used to monitor autophagic flux. Thus, these studies may reveal the prognostic role of autophagy rather than that of AMMs. At the same time, SQSTM1 functions as a scaffold protein for multiple signaling pathways and its prognostic role may therefore be independent of autophagy (127).

Beclin-1 is considered a haploinsufficient tumor suppressor in a number of cancers. Monoallelic deletion of the BECN1 gene is frequently observed in breast and ovarian cancer (128). However, the BECN1 gene is located in proximity to the known tumor suppressor BRCA1 in both humans (chromosome 17) and mice (chromosome 11) and the two loci are simultaneously deleted in breast and ovarian cancer; therefore, the actual role of Beclin-1 in tumorigenesis has been questioned (129-131). The observation that low expression of the BECN1 transcript, but not BRCA1, is associated with poor prognosis in breast cancer supports the role of Beclin-1 as a tumor suppressor (132). In addition, Beclin-1 enhances the efficacy of chemotherapeutic agents in cervical and gastric cancer cells (133).

High levels of ATG5 are associated with poor prognosis in various solid tumors (134). ATG5 has been identified as a potential prognostic marker in cervical squamous cell carcinoma and endocervical adenocarcinoma cases obtained by The Cancer Genome Atlas (135). The aforementioned study demonstrated the involvement of ATG5 in control of markers of EMT, invasive behavior and the immune effector process of T cell-mediated immunity. Contradictory results have been obtained for AMBRA1 (136). High expression of AMBRA1 is associated with poor prognosis in patients with various malignancies, such as pancreatic ductal adenocarcinoma, cholangiocarcinoma and gastric and prostate cancer (137-140). By contrast, the expression of AMBRA1 in early-stage melanoma is associated with a better prognosis (141).

9. Conclusions and perspectives

AMMs influence tumorigenesis via canonical and non-canonical autophagy functions; however, the dominance of these functions is unclear. Certain AMMs, such as ATG12, Beclin-1 and AMBRA1, have a domain that is activated by caspase and typically prevents activity of anti-apoptotic proteins (65,66,69). These AMMs therefore may link autophagy and apoptosis. Under certain stress conditions, their levels determine the fate of cells towards survival (autophagy) or death (apoptosis). Similarly, high levels of ROS promote autophagy-independent involvement of ULK1 in triggering necrosis (74). Thus, it would be useful to clarify how AMMs are regulated in cancer. For example, the LC3 and GABA type A Receptor-Associated Protein) family consists of at least 7 proteins that have different functions in membrane management for autophagic and non-autophagic purposes (142). Furthermore, they may be regulated by different transcription factors in different tissue, which could explain the activation of tissue-specific molecular programs beyond autophagy (142). As aforementioned, expression of LC3B and LC3A is associated with poor prognosis in gastric, breast and other types of cancer (117-122).

The existence of alternative variants has been demonstrated for several AMMs and may account for novel autophagy-independent functions (143). The autophagy-incompetent ATG7 p.Arg659*, has been proposed as a cholangiocarcinoma-associated gene (144). In addition, an ATG7 splice variant has been described that is unable to lipidate LC3 and is incompetent for autophagy (145), but, it remains to be clarified whether this variant has a function in cancer. As aforementioned, an ATG16L1 variant (T600A) stimulates an anti-tumor immune response and is associated with a good prognosis (99). SQSTM1 is expressed in several variants: N-Ter truncated isoform lacking the domain responsible for SQSTM1 oligomerization and autophagic cargo sorting ability (146); splice variant affecting the p62/Keap1/NRF2 axis (147) and SQSTM1 3' untranslated region-truncated variant associated with aggressiveness and resistance to therapy in patients with breast cancer (148). Therefore, it would be of interest to determine whether these different AMMs isoforms exhibit autophagy-independent functions.

Determining the autophagy-independent function of a single AMM in cancer is challenging because autophagy is a redundant signaling pathway that can find alternative routes to function and influence other cellular processes (5). Therefore, modulation of multiple autophagy markers should be considered before claiming that AMM activity is independent of autophagy. Alternatively, it is advisable to investigate the autophagy-independent role of AMMs in vitro by using autophagy-incompetent mutants, such as the ATG5 variant that cannot bind its autophagic partner ATG5K130R (149).

In summary, the role of AMMs is not limited to canonical autophagy but also involves autophagy-independent functions in various biological processes. Nevertheless, further studies that elucidate the link between autophagy-dependent and -independent pathways will help to clarify the activity of AMMs in cancer progression and response to therapies as well as in the identification of novel therapeutic targets.

Supplementary Data

Availability of data and materials

Not applicable.

Authors' contributions

GT and MS conceived the review, analyzed the literature and wrote the manuscript. GT collected and reviewed the literature and produced the figures. RM critically revised the manuscript.

All authors have read and approved the final manuscript. Data authentication is not applicable.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Abbreviations:

AMM

autophagy machinery molecule

PI3P

phosphatidylinositol-3-phosphate

NER

nucleotide excision repair

NHEJ

non-homologous end joining

MEF

mouse embryonic fibroblast

EMT

epithelial-to-mesenchymal transition

SA

secretory autophagy

LAP

LC-3-associated phagocytosis

ROS

reactive oxygen species

Acknowledgments

Not applicable.

Funding

The present study was supported by the Italian Ministry of Health-Ricerca Corrente.

References

1 

Suzuki K, Kubota Y, Sekito T and Ohsumi Y: Hierarchy of Atg proteins in pre-autophagosomal structure organization. Genes Cells. 12:209–218. 2007. View Article : Google Scholar : PubMed/NCBI

2 

Reggiori F and Klionsky DJ: Autophagic processes in yeast: Mechanism, machinery and regulation. Genetics. 194:341–361. 2013. View Article : Google Scholar : PubMed/NCBI

3 

Pankiv S, Clausen TH, Lamark T, Brech A, Bruun JA, Outzen H, Øvervatn A, Bjørkøy G and Johansen T: p62/SQSTM1 binds directly to Atg8/LC3 to facilitate degradation of ubiquitinated protein aggregates by autophagy. J Biol Chem. 282:24131–24145. 2007. View Article : Google Scholar : PubMed/NCBI

4 

Elmore SP, Qian T, Grissom SF and Lemasters JJ: The mitochondrial permeability transition initiates autophagy in rat hepatocytes. FASEB J. 15:2286–2287. 2001. View Article : Google Scholar : PubMed/NCBI

5 

Mizushima N and Levine B: Autophagy in human diseases. N Engl J Med. 383:1564–1576. 2020. View Article : Google Scholar : PubMed/NCBI

6 

Bustos SO, Antunes F, Rangel MC and Chammas R: Emerging autophagy functions shape the tumor microenvironment and play a role in cancer progression-implications for cancer therapy. Front Oncol. 10:6064362020. View Article : Google Scholar

7 

Xie X, Koh JY, Price S, White E and Mehnert JM: Atg7 overcomes senescence and promotes growth of BrafV600E-Driven Melanoma. Cancer Discov. 5:410–423. 2015. View Article : Google Scholar : PubMed/NCBI

8 

Yang A, Herter-Sprie G, Zhang H, Lin EY, Biancur D, Wang X, Deng J, Hai J, Yang S, Wong KK and Kimmelman AC: Autophagy sustains pancreatic cancer growth through both cell-autonomous and nonautonomous mechanisms. Cancer Discov. 8:276–287. 2018. View Article : Google Scholar : PubMed/NCBI

9 

Shchors K, Massaras A and Hanahan D: Dual targeting of the autophagic regulatory circuitry in gliomas with repurposed drugs elicits cell-lethal autophagy and therapeutic benefit. Cancer Cell. 28:456–471. 2015. View Article : Google Scholar : PubMed/NCBI

10 

Santanam U, Banach-Petrosky W, Abate-Shen C, Shen MM, White E and DiPaola RS: Atg7 cooperates with Pten loss to drive prostate cancer tumor growth. Genes Dev. 30:399–407. 2016. View Article : Google Scholar : PubMed/NCBI

11 

Karsli-Uzunbas G, Guo JY, Price S, Teng X, Laddha SV, Khor S, Kalaany NY, Jacks T, Chan CS, Rabinowitz JD and White E: Autophagy is required for glucose homeostasis and lung tumor maintenance. Cancer Discov. 4:914–927. 2014. View Article : Google Scholar : PubMed/NCBI

12 

Huo Y, Cai H, Teplova I, Bowman-Colin C, Chen G, Price S, Barnard N, Ganesan S, Karantza V, White E and Xia B: Autophagy opposes p53-mediated tumor barrier to facilitate tumorigenesis in a model of PALB2-associated hereditary breast cancer. Cancer Discov. 3:894–907. 2013. View Article : Google Scholar : PubMed/NCBI

13 

Degenhardt K, Mathew R, Beaudoin B, Bray K, Anderson D, Chen G, Mukherjee C, Shi Y, Gélinas C, Fan Y, et al: Autophagy promotes tumor cell survival and restricts necrosis, inflammation, and tumorigenesis. Cancer Cell. 10:51–64. 2006. View Article : Google Scholar : PubMed/NCBI

14 

Karantza-Wadsworth V, Patel S, Kravchuk O, Chen G, Mathew R, Jin S and White E: Autophagy mitigates metabolic stress and genome damage in mammary tumorigenesis. Genes Dev. 21:1621–1635. 2007. View Article : Google Scholar : PubMed/NCBI

15 

Moussay E, Kaoma T, Baginska J, Muller A, Van Moer K, Nicot N, Nazarov PV, Vallar L, Chouaib S, Berchem G and Janji B: The acquisition of resistance to TNFα in breast cancer cells is associated with constitutive activation of autophagy as revealed by a transcriptome analysis using a custom microarray. Autophagy. 7:760–770. 2011. View Article : Google Scholar : PubMed/NCBI

16 

Bildik G, Liang X, Sutton MN, Bast RC Jr and Lu Z: DIRAS3: An Imprinted tumor suppressor gene that regulates RAS and PI3K-driven cancer growth, motility, autophagy and tumor dormancy. Mol Cancer Ther. 21:25–37. 2021. View Article : Google Scholar

17 

Dower CM, Wills CA, Frisch SM and Wang HG: Mechanisms and context underlying the role of autophagy in cancer metastasis. Autophagy. 14:1110–1128. 2018. View Article : Google Scholar : PubMed/NCBI

18 

Mizushima N, Yoshimori T and Ohsumi Y: The role of Atg proteins in autophagosome formation. Annu Rev Cell Dev Biol. 27:107–132. 2011. View Article : Google Scholar : PubMed/NCBI

19 

Kuma A, Hatano M, Matsui M, Yamamoto A, Nakaya H, Yoshimori T, Ohsumi Y, Tokuhisa T and Mizushima N: The role of autophagy during the early neonatal starvation period. Nature. 432:1032–1036. 2004. View Article : Google Scholar : PubMed/NCBI

20 

Levine B and Kroemer G: Biological functions of autophagy genes: A disease perspective. Cell. 176:11–42. 2019. View Article : Google Scholar : PubMed/NCBI

21 

Oughtred R, Rust J, Chang C, Breitkreutz BJ, Stark C, Willems A, Boucher L, Leung G, Kolas N, Zhang F, et al: The BioGRID database: A comprehensive biomedical resource of curated protein, genetic, and chemical interactions. Protein Sci. 30:187–200. 2021. View Article : Google Scholar

22 

Leidal AM and Debnath J: Emerging roles for the autophagy machinery in extracellular vesicle biogenesis and secretion. FASEB Bioadv. 3:377–386. 2021. View Article : Google Scholar : PubMed/NCBI

23 

Okamoto T, Yeo SK, Hao M, Copley MR, Haas MA, Chen S and Guan JL: FIP200 suppresses immune checkpoint therapy responses in breast cancers by limiting AZI2/TBK1/IRF signaling independent of its canonical autophagy function. Cancer Res. 80:3580–3592. 2020. View Article : Google Scholar : PubMed/NCBI

24 

Guo H, Sadoul R and Gibbings D: Autophagy-independent effects of autophagy-related-5 (Atg5) on exosome production and metastasis. Mol Cell Oncol. 5:e14459412018. View Article : Google Scholar : PubMed/NCBI

25 

Hu F, Li G, Huang C, Hou Z, Yang X, Luo X, Feng Y, Wang G, Hu J and Cao Z: The autophagy-independent role of BECN1 in colorectal cancer metastasis through regulating STAT3 signaling pathway activation. Cell Death Dis. 11:3042020. View Article : Google Scholar : PubMed/NCBI

26 

Yamamoto H, Zhang S and Mizushima N: Autophagy genes in biology and disease. Nat Rev Genet. 24:382–400. 2023. View Article : Google Scholar : PubMed/NCBI

27 

Agarwal S, Bell CM, Rothbart SB and Moran RG: AMP-activated Protein Kinase (AMPK) Control of mTORC1 Is p53-and TSC2-independent in pemetrexed-treated carcinoma cells. J Biol Chem. 290:27473–27486. 2015. View Article : Google Scholar : PubMed/NCBI

28 

Hosokawa N, Hara T, Kaizuka T, Kishi C, Takamura A, Miura Y, Iemura S, Natsume T, Takehana K, Yamada N, et al: Nutrient-dependent mTORC1 association with the ULK1-Atg13-FIP200 complex required for autophagy. Mol Biol Cell. 20:1981–1991. 2009. View Article : Google Scholar : PubMed/NCBI

29 

Mercer TJ, Gubas A and Tooze SA: A molecular perspective of mammalian autophagosome biogenesis. J Biol Chem. 293:5386–5395. 2018. View Article : Google Scholar : PubMed/NCBI

30 

Zachari M and Ganley IG: The mammalian ULK1 complex and autophagy initiation. Essays Biochem. 61:585–596. 2017. View Article : Google Scholar : PubMed/NCBI

31 

Liang C, Feng P, Ku B, Dotan I, Canaani D, Oh BH and Jung JU: Autophagic and tumour suppressor activity of a novel Beclin1-binding protein UVRAG. Nat Cell Biol. 8:688–699. 2006. View Article : Google Scholar : PubMed/NCBI

32 

Fimia GM, Stoykova A, Romagnoli A, Giunta L, Di Bartolomeo S, Nardacci R, Corazzari M, Fuoco C, Ucar A, Schwartz P, et al: Ambra1 regulates autophagy and development of the nervous system. Nature. 447:1121–1125. 2007. View Article : Google Scholar : PubMed/NCBI

33 

Sawa-Makarska J, Baumann V, Coudevylle N, von Bülow S, Nogellova V, Abert C, Schuschnig M, Graef M, Hummer G and Martens S: Reconstitution of autophagosome nucleation defines Atg9 vesicles as seeds for membrane formation. Science. 369:eaaz77142020. View Article : Google Scholar : PubMed/NCBI

34 

Matoba K, Kotani T, Tsutsumi A, Tsuji T, Mori T, Noshiro D, Sugita Y, Nomura N, Iwata S, Ohsumi Y, et al: Atg9 is a lipid scramblase that mediates autophagosomal membrane expansion. Nat Struct Mol Biol. 27:1185–1193. 2020. View Article : Google Scholar : PubMed/NCBI

35 

Dooley HC, Razi M, Polson HE, Girardin SE, Wilson MI and Tooze SA: WIPI2 links LC3 conjugation with PI3P, autophagosome formation, and pathogen clearance by recruiting Atg12-5-16L1. Mol Cell. 55:238–252. 2014. View Article : Google Scholar : PubMed/NCBI

36 

Wible DJ, Chao HP, Tang DG and Bratton SB: ATG5 cancer mutations and alternative mRNA splicing reveal a conjugation switch that regulates ATG12-ATG5-ATG16L1 complex assembly and autophagy. Cell Discov. 5:422019. View Article : Google Scholar : PubMed/NCBI

37 

Baines K, Yoshioka K, Takuwa Y and Lane JD: The ATG5 interactome links clathrin-mediated vesicular trafficking with the autophagosome assembly machinery. Autophagy Rep. 1:88–118. 2022. View Article : Google Scholar : PubMed/NCBI

38 

Haller M, Hock AK, Giampazolias E, Oberst A, Green DR, Debnath J, Ryan KM, Vousden KH and Tait SW: Ubiquitination and proteasomal degradation of ATG12 regulates its proapoptotic activity. Autophagy. 10:2269–2278. 2014. View Article : Google Scholar

39 

Tanida I, Ueno T and Kominami E: LC3 conjugation system in mammalian autophagy. Int J Biochem Cell Biol. 36:2503–2518. 2004. View Article : Google Scholar : PubMed/NCBI

40 

Melia TJ, Lystad AH and Simonsen A: Autophagosome biogenesis: From membrane growth to closure. J Cell Biol. 219:e2020020852020. View Article : Google Scholar : PubMed/NCBI

41 

Gatica D, Lahiri V and Klionsky DJ: Cargo recognition and degradation by selective autophagy. Nat Cell Biol. 20:233–242. 2018. View Article : Google Scholar : PubMed/NCBI

42 

Liu WJ, Ye L, Huang WF, Guo LJ, Xu ZG, Wu HL, Yang C and Liu HF: p62 links the autophagy pathway and the ubiqutin-proteasome system upon ubiquitinated protein degradation. Cell Mol Biol Lett. 21:292016. View Article : Google Scholar

43 

Yim WW and Mizushima N: Lysosome biology in autophagy. Cell Discov. 6:62020. View Article : Google Scholar : PubMed/NCBI

44 

Dikic I and Elazar Z: Mechanism and medical implications of mammalian autophagy. Nat Rev Mol Cell Biol. 19:349–364. 2018. View Article : Google Scholar : PubMed/NCBI

45 

Hanahan D: Hallmarks of cancer: New Dimensions. Cancer Discov. 12:31–46. 2022. View Article : Google Scholar : PubMed/NCBI

46 

Mathew R, Kongara S, Beaudoin B, Karp CM, Bray K, Degenhardt K, Chen G, Jin S and White E: Autophagy suppresses tumor progression by limiting chromosomal instability. Genes Dev. 21:1367–1381. 2007. View Article : Google Scholar : PubMed/NCBI

47 

Marteijn JA, Lans H, Vermeulen W and Hoeijmakers JH: Understanding nucleotide excision repair and its roles in cancer and ageing. Nat Rev Mol Cell Biol. 15:465–481. 2014. View Article : Google Scholar : PubMed/NCBI

48 

Yang Y, He S, Wang Q, Li F, Kwak MJ, Chen S, O'Connell D, Zhang T, Pirooz SD, Jeon YH, et al: Autophagic UVRAG Promotes UV-Induced Photolesion Repair by Activation of the CRL4(DDB2) E3 Ligase. Mol Cell. 62:507–519. 2016. View Article : Google Scholar : PubMed/NCBI

49 

Zhao Z, Oh S, Li D, Ni D, Pirooz SD, Lee JH, Yang S, Lee JY, Ghozalli I, Costanzo V, et al: A dual role for UVRAG in maintaining chromosomal stability independent of autophagy. Dev Cell. 22:1001–1016. 2012. View Article : Google Scholar : PubMed/NCBI

50 

Park JM, Tougeron D, Huang S, Okamoto K and Sinicrope FA: Beclin 1 and UVRAG confer protection from radiation-induced DNA damage and maintain centrosome stability in colorectal cancer cells. PLoS One. 9:e1008192014. View Article : Google Scholar : PubMed/NCBI

51 

Knævelsrud H, Ahlquist T, Merok MA, Nesbakken A, Stenmark H, Lothe RA and Simonsen A: UVRAG mutations associated with microsatellite unstable colon cancer do not affect autophagy. Autophagy. 6:863–870. 2010. View Article : Google Scholar : PubMed/NCBI

52 

He S, Zhao Z, Yang Y, O'Connell D, Zhang X, Oh S, Ma B, Lee JH, Zhang T, Varghese B, et al: Truncating mutation in the autophagy gene UVRAG confers oncogenic properties and chemosensitivity in colorectal cancers. Nat Commun. 6:78392015. View Article : Google Scholar : PubMed/NCBI

53 

Sun SY, Hu XT, Yu XF, Zhang YY, Liu XH, Liu YH, Wu SH, Li YY, Cui SX and Qu XJ: Nuclear translocation of ATG5 induces DNA mismatch repair deficiency (MMR-D)/microsatellite instability (MSI) via interacting with Mis18α in colorectal cancer. Br J Pharmacol. 178:2351–2369. 2021. View Article : Google Scholar : PubMed/NCBI

54 

Maskey D, Yousefi S, Schmid I, Zlobec I, Perren A, Friis R and Simon HU: ATG5 is induced by DNA-damaging agents and promotes mitotic catastrophe independent of autophagy. Nat Commun. 4:21302013. View Article : Google Scholar : PubMed/NCBI

55 

Maiani E, Milletti G, Nazio F, Holdgaard SG, Bartkova J, Rizza S, Cianfanelli V, Lorente M, Simoneschi D, Di Marco M, et al: AMBRA1 regulates cyclin D to guard S-phase entry and genomic integrity. Nature. 592:799–803. 2021. View Article : Google Scholar : PubMed/NCBI

56 

Liu PF, Leung CM, Chang YH, Cheng JS, Chen JJ, Weng CJ, Tsai KW, Hsu CJ, Liu YC, Hsu PC, et al: ATG4B promotes colorectal cancer growth independent of autophagic flux. Autophagy. 10:1454–1465. 2014. View Article : Google Scholar : PubMed/NCBI

57 

Lee IH, Kawai Y, Fergusson MM, Rovira II, Bishop AJ, Motoyama N, Cao L and Finkel T: Atg7 modulates p53 activity to regulate cell cycle and survival during metabolic stress. Science. 336:225–228. 2012. View Article : Google Scholar : PubMed/NCBI

58 

Frémont S, Gérard A, Galloux M, Janvier K, Karess RE and Berlioz-Torrent C: Beclin-1 is required for chromosome congression and proper outer kinetochore assembly. EMBO Rep. 14:364–372. 2013. View Article : Google Scholar : PubMed/NCBI

59 

Thoresen SB, Pedersen NM, Liestøl K and Stenmark H: A phosphatidylinositol 3-kinase class III sub-complex containing VPS15, VPS34, Beclin 1, UVRAG and BIF-1 regulates cytokinesis and degradative endocytic traffic. Exp Cell Res. 316:3368–3378. 2010. View Article : Google Scholar : PubMed/NCBI

60 

Sagona AP, Nezis IP, Pedersen NM, Liestøl K, Poulton J, Rusten TE, Skotheim RI, Raiborg C and Stenmark H: PtdIns(3) P controls cytokinesis through KIF13A-mediated recruitment of FYVE-CENT to the midbody. Nat Cell Biol. 12:362–371. 2010. View Article : Google Scholar : PubMed/NCBI

61 

Zhu Y, Zhao L, Liu L, Gao P, Tian W, Wang X, Jin H, Xu H and Chen Q: Beclin 1 cleavage by caspase-3 inactivates autophagy and promotes apoptosis. Protein Cell. 1:468–477. 2010. View Article : Google Scholar

62 

Young MM, Takahashi Y, Khan O, Park S, Hori T, Yun J, Sharma AK, Amin S, Hu CD, Zhang J, et al: Autophagosomal membrane serves as platform for intracellular death-inducing signaling complex (iDISC)-mediated caspase-8 activation and apoptosis. J Biol Chem. 287:12455–12468. 2012. View Article : Google Scholar : PubMed/NCBI

63 

Han J, Hou W, Goldstein LA, Stolz DB, Watkins SC and Rabinowich H: A Complex between Atg7 and Caspase-9: A novel mechanism of cross-regulation between autophagy and apoptosis. J Biol Chem. 289:6485–6497. 2014. View Article : Google Scholar :

64 

Wirawan E, Vande Walle L, Kersse K, Cornelis S, Claerhout S, Vanoverberghe I, Roelandt R, De Rycke R, Verspurten J, Declercq W, et al: Caspase-mediated cleavage of Beclin-1 inactivates Beclin-1-induced autophagy and enhances apoptosis by promoting the release of proapoptotic factors from mitochondria. Cell Death Dis. 1:e182010. View Article : Google Scholar : PubMed/NCBI

65 

Li X, Su J, Xia M, Li H, Xu Y, Ma C, Ma L, Kang J, Yu H, Zhang Z and Sun L: Caspase-mediated cleavage of Beclin1 inhibits autophagy and promotes apoptosis induced by S1 in human ovarian cancer SKOV3 cells. Apoptosis. 21:225–238. 2016. View Article : Google Scholar

66 

Strappazzon F, Di Rita A, Cianfanelli V, D'Orazio M, Nazio F, Fimia GM and Cecconi F: Prosurvival AMBRA1 turns into a proapoptotic BH3-like protein during mitochondrial apoptosis. Autophagy. 12:963–975. 2016. View Article : Google Scholar : PubMed/NCBI

67 

Betin VM and Lane JD: Caspase cleavage of Atg4D stimulates GABARAP-L1 processing and triggers mitochondrial targeting and apoptosis. J Cell Sci. 122(Pt 14): 2554–2566. 2009. View Article : Google Scholar : PubMed/NCBI

68 

Yousefi S, Perozzo R, Schmid I, Ziemiecki A, Schaffner T, Scapozza L, Brunner T and Simon HU: Calpain-mediated cleavage of Atg5 switches autophagy to apoptosis. Nat Cell Biol. 8:1124–1132. 2006. View Article : Google Scholar : PubMed/NCBI

69 

Rubinstein AD, Eisenstein M, Ber Y, Bialik S and Kimchi A: The autophagy protein Atg12 associates with antiapoptotic Bcl-2 family members to promote mitochondrial apoptosis. Mol Cell. 44:698–709. 2011. View Article : Google Scholar : PubMed/NCBI

70 

Yoo BH, Khan IA, Koomson A, Gowda P, Sasazuki T, Shirasawa S, Gujar S and Rosen KV: Oncogenic RAS-induced downregulation of ATG12 is required for survival of malignant intestinal epithelial cells. Autophagy. 14:134–151. 2018. View Article : Google Scholar :

71 

Radoshevich L, Murrow L, Chen N, Fernandez E, Roy S, Fung C and Debnath J: ATG12 conjugation to ATG3 regulates mitochondrial homeostasis and cell death. Cell. 142:590–600. 2010. View Article : Google Scholar : PubMed/NCBI

72 

Liu H, He Z, Germič N, Ademi H, Frangež Ž, Felser A, Peng S, Riether C, Djonov V, Nuoffer JM, et al: ATG12 deficiency leads to tumor cell oncosis owing to diminished mitochondrial biogenesis and reduced cellular bioenergetics. Cell Death Differ. 27:1965–1980. 2020. View Article : Google Scholar :

73 

Ni Z, He J, Wu Y, Hu C, Dai X, Yan X, Li B, Li X, Xiong H, Li Y, et al: AKT-mediated phosphorylation of ATG4B impairs mitochondrial activity and enhances the Warburg effect in hepatocellular carcinoma cells. Autophagy. 14:685–701. 2018. View Article : Google Scholar :

74 

Wu W, Wang X, Berleth N, Deitersen J, Wallot-Hieke N, Böhler P, Schlütermann D, Stuhldreier F, Cox J, Schmitz K, et al: The autophagy-initiating kinase ULK1 Controls RIPK1-mediated cell death. Cell Rep. 31:1075472020. View Article : Google Scholar : PubMed/NCBI

75 

Joshi A, Iyengar R, Joo JH, Li-Harms XJ, Wright C, Marino R, Winborn BJ, Phillips A, Temirov J, Sciarretta S, et al: Nuclear ULK1 promotes cell death in response to oxidative stress through PARP1. Cell Death Differ. 23:216–230. 2016. View Article : Google Scholar :

76 

Satyavarapu EM, Das R and Mandal C, Mukhopadhyay A and Mandal C: Autophagy-independent induction of LC3B through oxidative stress reveals its non-canonical role in anoikis of ovarian cancer cells. Cell Death Dis. 9:9342018. View Article : Google Scholar

77 

Hanahan D and Weinberg RA: Hallmarks of cancer: the next generation. Cell. 144:646–674. 2011. View Article : Google Scholar : PubMed/NCBI

78 

Aiello NM, Maddipati R, Norgard RJ, Balli D, Li J, Yuan S, Yamazoe T, Black T, Sahmoud A, Furth EE, et al: EMT subtype influences epithelial plasticity and mode of cell migration. Dev Cell. 45:681–695.e4. 2018. View Article : Google Scholar : PubMed/NCBI

79 

Zada S, Hwang JS, Ahmed M, Lai TH, Pham TM and Kim DR: Control of the epithelial-to-mesenchymal transition and cancer metastasis by autophagy-dependent SNAI1 degradation. Cells. 8:1292019. View Article : Google Scholar : PubMed/NCBI

80 

Han JH, Kim YK, Kim H, Lee J, Oh MJ, Kim SB, Kim M, Kim KH, Yoon HJ, Lee MS, et al: Snail acetylation by autophagy-derived acetyl-coenzyme A promotes invasion and metastasis of KRAS-LKB1 co-mutated lung cancer cells. Cancer Commun (Lond). 42:716–749. 2022. View Article : Google Scholar : PubMed/NCBI

81 

Sharifi MN, Mowers EE, Drake LE, Collier C, Chen H, Zamora M, Mui S and Macleod KF: Autophagy promotes focal adhesion disassembly and cell motility of metastatic tumor cells through the direct interaction of paxillin with LC3. Cell Rep. 15:1660–1672. 2016. View Article : Google Scholar : PubMed/NCBI

82 

Santarosa M and Maestro R: The autophagic route of E-Cadherin and cell adhesion molecules in cancer progression. Cancers (Basel). 13:63282021. View Article : Google Scholar : PubMed/NCBI

83 

Martinez-Lopez N, Athonvarangkul D, Mishall P, Sahu S and Singh R: Autophagy proteins regulate ERK phosphorylation. Nat Commun. 4:27992013. View Article : Google Scholar : PubMed/NCBI

84 

Rohatgi RA, Janusis J, Leonard D, Bellvé KD, Fogarty KE, Baehrecke EH, Corvera S and Shaw LM: Beclin 1 regulates growth factor receptor signaling in breast cancer. Oncogene. 34:5352–5362. 2015. View Article : Google Scholar : PubMed/NCBI

85 

Wijshake T, Zou Z, Chen B, Zhong L, Xiao G, Xie Y, Doench JG, Bennett L and Levine B: Tumor-suppressor function of Beclin 1 in breast cancer cells requires E-cadherin. Proc Natl Acad Sci USA. 118:e20204781182021. View Article : Google Scholar : PubMed/NCBI

86 

Wong M, Ganapathy AS, Suchanec E, Laidler L, Ma T and Nighot P: Intestinal epithelial tight junction barrier regulation by autophagy-related protein ATG6/beclin 1. Am J Physiol, Cell Physiol. 316:C753–C765. 2019. View Article : Google Scholar : PubMed/NCBI

87 

Damiano V, Spessotto P, Vanin G, Perin T, Maestro R and Santarosa M: The autophagy machinery contributes to E-cadherin turnover in breast cancer. Front Cell Dev Biol. 8:5452020. View Article : Google Scholar : PubMed/NCBI

88 

Baisamy L, Cavin S, Jurisch N and Diviani D: The ubiquitin-like protein LC3 regulates the Rho-GEF activity of AKAP-Lbc. J Biol Chem. 284:28232–28242. 2009. View Article : Google Scholar : PubMed/NCBI

89 

Zhong Y, Long T, Gu CS, Tang JY, Gao LF, Zhu JX, Hu ZY, Wang X, Ma YD, Ding YQ, et al: MYH9-dependent polarization of ATG9B promotes colorectal cancer metastasis by accelerating focal adhesion assembly. Cell Death Differ. 28:3251–3269. 2021. View Article : Google Scholar : PubMed/NCBI

90 

Galluzzi L and Green DR: Autophagy-Independent functions of the autophagy machinery. Cell. 177:1682–1699. 2019. View Article : Google Scholar : PubMed/NCBI

91 

Ishibashi K, Uemura T, Waguri S and Fukuda M: Atg16L1, an essential factor for canonical autophagy, participates in hormone secretion from PC12 cells independently of autophagic activity. Mol Biol Cell. 23:3193–3202. 2012. View Article : Google Scholar : PubMed/NCBI

92 

Guo H, Chitiprolu M, Roncevic L, Javalet C, Hemming FJ, Trung MT, Meng L, Latreille E, Tanese de Souza C, McCulloch D, et al: Atg5 Disassociates the V1V0-ATPase to promote exosome production and tumor metastasis independent of canonical macroautophagy. Dev Cell. 43:716–730.e7. 2017. View Article : Google Scholar

93 

Ponpuak M, Mandell MA, Kimura T, Chauhan S, Cleyrat C and Deretic V: Secretory autophagy. Curr Opin Cell Biol. 35:106–116. 2015. View Article : Google Scholar : PubMed/NCBI

94 

Li X, Wei Z, Yu H, Xu Y, He W, Zhou X and Gou X: Secretory autophagy-induced bladder tumour-derived extracellular vesicle secretion promotes angiogenesis by activating the TPX2-mediated phosphorylation of the AURKA-PI3K-AKT axis. Cancer Lett. 523:10–28. 2021. View Article : Google Scholar : PubMed/NCBI

95 

Tzeng HT, Yang JL, Tseng YJ, Lee CH, Chen WJ and Chyuan IT: Plasminogen activator inhibitor-1 secretion by autophagy contributes to melanoma resistance to chemotherapy through tumor microenvironment modulation. Cancers (Basel). 13:12532021. View Article : Google Scholar : PubMed/NCBI

96 

New J, Arnold L, Ananth M, Alvi S, Thornton M, Werner L, Tawfik O, Dai H, Shnayder Y, Kakarala K, et al: Secretory autophagy in cancer-associated fibroblasts promotes head and neck cancer progression and offers a novel therapeutic target. Cancer Res. 77:6679–6691. 2017. View Article : Google Scholar : PubMed/NCBI

97 

Cunha LD, Yang M, Carter R, Guy C, Harris L, Crawford JC, Quarato G, Boada-Romero E, Kalkavan H, Johnson MDL, et al: LC3-Associated phagocytosis in myeloid cells promotes tumor immune tolerance. Cell. 175:429–441.e16. 2018. View Article : Google Scholar : PubMed/NCBI

98 

Liu X, Zhang W, Xu Y, Xu X, Jiang Q, Ruan J, Wu Y, Zhou Y, Saw PE and Luo B: Targeting PI3Kγ/AKT Pathway Remodels LC3-Associated phagocytosis induced immunosuppression after radiofrequency ablation. Adv Sci (Weinh). 9:e21021822022. View Article : Google Scholar

99 

Grimm WA, Messer JS, Murphy SF, Nero T, Lodolce JP, Weber CR, Logsdon MF, Bartulis S, Sylvester BE, Springer A, et al: The Thr300Ala variant in ATG16L1 is associated with improved survival in human colorectal cancer and enhanced production of type I interferon. Gut. 65:456–464. 2016. View Article : Google Scholar

100 

Peng Y and Croce CM: The role of MicroRNAs in human cancer. Signal Transduct Target Ther. 1:150042016. View Article : Google Scholar : PubMed/NCBI

101 

Chipman LB and Pasquinelli AE: miRNA Targeting: Growing beyond the Seed. Trends Genet. 35:215–222. 2019. View Article : Google Scholar : PubMed/NCBI

102 

de la Cruz-Ojeda P, Flores-Campos R, Navarro-Villarán E and Muntané J: The role of non-coding RNAs in autophagy during carcinogenesis. Front Cell Dev Biol. 10:7993922022. View Article : Google Scholar : PubMed/NCBI

103 

Shan C, Chen X, Cai H, Hao X, Li J, Zhang Y, Gao J, Zhou Z, Li X, Liu C, et al: The emerging roles of autophagy-related MicroRNAs in cancer. Int J Biol Sci. 17:134–150. 2021. View Article : Google Scholar : PubMed/NCBI

104 

Zhu H, Wu H, Liu X, Li B, Chen Y, Ren X, Liu CG and Yang JM: Regulation of autophagy by a beclin 1-targeted microRNA, miR-30a, in cancer cells. Autophagy. 5:816–823. 2009. View Article : Google Scholar : PubMed/NCBI

105 

Wang ZC, Huang FZ, Xu HB, Sun JC and Wang CF: MicroRNA-137 inhibits autophagy and chemosensitizes pancreatic cancer cells by targeting ATG5. Int J Biochem Cell Biol. 111:63–71. 2019. View Article : Google Scholar : PubMed/NCBI

106 

Chen Y, Zhou J, Wu X, Huang J, Chen W, Liu D, Zhang J, Huang Y and Xue W: miR-30a-3p inhibits renal cancer cell invasion and metastasis through targeting ATG12. Transl Androl Urol. 9:646–653. 2020. View Article : Google Scholar : PubMed/NCBI

107 

Hu JL, He GY, Lan XL, Zeng ZC, Guan J, Ding Y, Qian XL, Liao WT, Ding YQ and Liang L: Inhibition of ATG12-mediated autophagy by miR-214 enhances radiosensitivity in colorectal cancer. Oncogenesis. 7:162018. View Article : Google Scholar : PubMed/NCBI

108 

Pan X, Chen Y, Shen Y and Tantai J: Knockdown of TRIM65 inhibits autophagy and cisplatin resistance in A549/DDP cells by regulating miR-138-5p/ATG7. Cell Death Dis. 10:4292019. View Article : Google Scholar : PubMed/NCBI

109 

Chang Y, Yan W, He X, Zhang L, Li C, Huang H, Nace G, Geller DA, Lin J and Tsung A: miR-375 inhibits autophagy and reduces viability of hepatocellular carcinoma cells under hypoxic conditions. Gastroenterology. 143:177–187.e8. 2012. View Article : Google Scholar : PubMed/NCBI

110 

Zhang X, Shi H, Lin S, Ba M and Cui S: MicroRNA-216a enhances the radiosensitivity of pancreatic cancer cells by inhibiting beclin-1-mediated autophagy. Oncol Rep. 34:1557–1564. 2015. View Article : Google Scholar : PubMed/NCBI

111 

Li M, Chen XM, Wang DM, Gan L and Qiao Y: Effects of miR-26a on the expression of Beclin 1 in retinoblastoma cells. Genet Mol Res. 15:2016.

112 

Hou W, Song L, Zhao Y, Liu Q and Zhang S: Inhibition of beclin-1-mediated autophagy by MicroRNA-17-5p enhanced the radiosensitivity of glioma cells. Oncol Res. 25:43–53. 2017. View Article : Google Scholar : PubMed/NCBI

113 

Zhang F, Wang B, Long H, Yu J, Li F, Hou H and Yang Q: Decreased miR-124-3p expression prompted breast cancer cell progression mainly by targeting Beclin-1. Clin Lab. 62:1139–1145. 2016. View Article : Google Scholar : PubMed/NCBI

114 

Tan S, Shi H, Ba M, Lin S, Tang H, Zeng X and Zhang X: miR-409-3p sensitizes colon cancer cells to oxaliplatin by inhibiting Beclin-1-mediated autophagy. Int J Mol Med. 37:1030–1038. 2016. View Article : Google Scholar : PubMed/NCBI

115 

Wang Y, Li J, Shao C, Tang X, Du Y, Xu T, Zhao Z, Hu H, Sheng Y, Hu C and Xi Y: Systematic profiling of diagnostic and prognostic value of autophagy-related genes for sarcoma patients. BMC Cancer. 21:582021. View Article : Google Scholar : PubMed/NCBI

116 

Yang Q, Sun K, Xia W, Li Y, Zhong M and Lei K: Autophagy-related prognostic signature for survival prediction of triple negative breast cancer. PeerJ. 10:e128782022. View Article : Google Scholar : PubMed/NCBI

117 

Sivridis E, Koukourakis MI, Zois CE, Ledaki I, Ferguson DJ, Harris AL, Gatter KC and Giatromanolaki A: LC3A-positive light microscopy detected patterns of autophagy and prognosis in operable breast carcinomas. Am J Pathol. 176:2477–2489. 2010. View Article : Google Scholar : PubMed/NCBI

118 

Gachechiladze M, Uberall I, Skanderova D, Matchavariani J, Ibrahim M, Shani I, Smickova P, Kolek V, Cierna L, Klein J, et al: LC3A positive 'stone like structures' are differentially associated with survival outcomes and CD68 macrophage infiltration in patients with lung adenocarcinoma and squamous cell carcinoma. Lung Cancer. 156:129–135. 2021. View Article : Google Scholar : PubMed/NCBI

119 

Terabe T, Uchida F, Nagai H, Omori S, Ishibashi-Kanno N, Hasegawa S, Yamagata K, Gosho M, Yanagawa T and Bukawa H: Expression of autophagy-related markers at the surgical margin of oral squamous cell carcinoma correlates with poor prognosis and tumor recurrence. Hum Pathol. 73:156–163. 2018. View Article : Google Scholar

120 

Giatromanolaki A, Koukourakis MI, Georgiou I, Kouroupi M and Sivridis E: LC3A, LC3B and Beclin-1 Expression in gastric cancer. Anticancer Res. 38:6827–6833. 2018. View Article : Google Scholar : PubMed/NCBI

121 

Bortnik S, Tessier-Cloutier B, Leung S, Xu J, Asleh K, Burugu S, Magrill J, Greening K, Derakhshan F, Yip S, et al: Differential expression and prognostic relevance of autophagy-related markers ATG4B, GABARAP, and LC3B in breast cancer. Breast Cancer Res Treat. 183:525–547. 2020. View Article : Google Scholar : PubMed/NCBI

122 

Kim JW, Jun SY, Kim JM, Oh YH, Yoon G, Hong SM and Chung JY: Prognostic value of LC3B and p62 expression in small intestinal adenocarcinoma. J Clin Med. 10:53982021. View Article : Google Scholar : PubMed/NCBI

123 

Langer R, Neppl C, Keller MD, Schmid RA, Tschan MP and Berezowska S: Expression analysis of autophagy related markers LC3B, p62 and HMGB1 indicate an autophagy-independent negative prognostic impact of High p62 expression in pulmonary squamous cell carcinomas. Cancers (Basel). 10:2812018. View Article : Google Scholar : PubMed/NCBI

124 

Mathew R, Karp CM, Beaudoin B, Vuong N, Chen G, Chen HY, Bray K, Reddy A, Bhanot G, Gelinas C, et al: Autophagy suppresses tumorigenesis through elimination of p62. Cell. 137:1062–1075. 2009. View Article : Google Scholar : PubMed/NCBI

125 

Islam MA, Sooro MA and Zhang P: Autophagic regulation of p62 is critical for cancer therapy. Int J Mol Sci. 19:14052018. View Article : Google Scholar : PubMed/NCBI

126 

Ruan H, Xu J, Wang L, Zhao Z, Kong L, Lan B and Li X: The prognostic value of p62 in solid tumor patients: A meta-analysis. Oncotarget. 9:4258–4266. 2018. View Article : Google Scholar : PubMed/NCBI

127 

Sánchez-Martín P, Saito T and Komatsu M: p62/SQSTM1: 'Jack of all trades' in health and cancer. FEBS J. 286:8–23. 2019. View Article : Google Scholar

128 

Laddha SV, Ganesan S, Chan CS and White E: Mutational landscape of the essential autophagy gene BECN1 in human cancers. Mol Cancer Res. 12:485–490. 2014. View Article : Google Scholar : PubMed/NCBI

129 

Delaney JR, Patel CB, Bapat J, Jones CM, Ramos-Zapatero M, Ortell KK, Tanios R, Haghighiabyaneh M, Axelrod J, DeStefano JW, et al: Autophagy gene haploinsufficiency drives chromosome instability, increases migration, and promotes early ovarian tumors. PLoS Genet. 16:e10085582020. View Article : Google Scholar : PubMed/NCBI

130 

Qu X, Yu J, Bhagat G, Furuya N, Hibshoosh H, Troxel A, Rosen J, Eskelinen EL, Mizushima N, Ohsumi Y, et al: Promotion of tumorigenesis by heterozygous disruption of the beclin 1 autophagy gene. J Clin Invest. 112:1809–1820. 2003. View Article : Google Scholar : PubMed/NCBI

131 

Ajazi A and Foiani M: Vps30/Atg6/BECN1 at the crossroads between cell metabolism and DNA damage response. Autophagy. 18:1202–1204. 2022. View Article : Google Scholar : PubMed/NCBI

132 

Tang H, Sebti S, Titone R, Zhou Y, Isidoro C, Ross TS, Hibshoosh H, Xiao G, Packer M, Xie Y and Levine B: Decreased BECN1 mRNA expression in human breast cancer is associated with estrogen receptor-negative subtypes and poor prognosis. EBioMedicine. 2:255–263. 2015. View Article : Google Scholar : PubMed/NCBI

133 

Liu C, Yan X, Wang HQ, Gao YY, Liu J, Hu Z, Liu D, Gao J and Lin B: Autophagy-independent enhancing effects of Beclin 1 on cytotoxicity of ovarian cancer cells mediated by proteasome inhibitors. BMC Cancer. 12:6222012. View Article : Google Scholar : PubMed/NCBI

134 

Xu C, Zang Y, Zhao Y, Cui W, Zhang H, Zhu Y and Xu M: comprehensive pan-cancer analysis confirmed that ATG5 Promoted the maintenance of tumor metabolism and the occurrence of tumor immune escape. Front Oncol. 11:6522112021. View Article : Google Scholar : PubMed/NCBI

135 

Zhou S, Wang X, Ding J, Yang H and Xie Y: Increased ATG5 expression predicts poor prognosis and promotes EMT in cervical carcinoma. Front Cell Dev Biol. 9:7571842021. View Article : Google Scholar : PubMed/NCBI

136 

Qin YQ, Liu SY, Lv ML and Sun WL: Ambra1 in cancer: Implications for clinical oncology. Apoptosis. 27:720–729. 2022. View Article : Google Scholar : PubMed/NCBI

137 

Nitta T, Sato Y, Ren XS, Harada K, Sasaki M, Hirano S and Nakanuma Y: Autophagy may promote carcinoma cell invasion and correlate with poor prognosis in cholangiocarcinoma. Int J Clin Exp Pathol. 7:4913–4921. 2014.PubMed/NCBI

138 

Ko YH, Cho YS, Won HS, Jeon EK, An HJ, Hong SU, Park JH and Lee MA: Prognostic significance of autophagy-related protein expression in resected pancreatic ductal adenocarcinoma. Pancreas. 42:829–835. 2013. View Article : Google Scholar : PubMed/NCBI

139 

Falasca L, Torino F, Marconi M, Costantini M, Pompeo V, Sentinelli S, De Salvo L, Patrizio M, Padula C, Gallucci M, et al: AMBRA1 and SQSTM1 expression pattern in prostate cancer. Apoptosis. 20:1577–1586. 2015. View Article : Google Scholar : PubMed/NCBI

140 

Ieni A, Cardia R, Giuffrè G, Rigoli L, Caruso RA and Tuccari G: Immunohistochemical expression of autophagy-related proteins in advanced tubular gastric adenocarcinomas and its implications. Cancers (Basel). 11:3892019. View Article : Google Scholar : PubMed/NCBI

141 

Tang DY, Ellis RA and Lovat PE: Prognostic impact of autophagy biomarkers for cutaneous melanoma. Front Oncol. 6:2362016. View Article : Google Scholar : PubMed/NCBI

142 

Schaaf MB, Keulers TG, Vooijs MA and Rouschop KM: LC3/GABARAP family proteins: Autophagy-(un)related functions. FASEB J. 30:3961–3978. 2016. View Article : Google Scholar : PubMed/NCBI

143 

González-Rodríguez P, Klionsky DJ and Joseph B: Autophagy regulation by RNA alternative splicing and implications in human diseases. Nat Commun. 13:27352022. View Article : Google Scholar : PubMed/NCBI

144 

Greer SU, Ogmundsdottir MH, Chen J, Lau BT, Delacruz RGC, Sandoval IT, Kristjansdottir S, Jones DA, Haslem DS, Romero R, et al: Genetic risk of cholangiocarcinoma is linked to the autophagy gene ATG7. BioRxiv. 2019.

145 

Ogmundsdottir MH, Fock V, Sooman L, Pogenberg V, Dilshat R, Bindesbøll C, Ogmundsdottir HM, Simonsen A, Wilmanns M and Steingrimsson E: A short isoform of ATG7 fails to lipidate LC3/GABARAP. Sci Rep. 8:143912018. View Article : Google Scholar : PubMed/NCBI

146 

Somlapura M, Gottschalk B, Lahiri P, Kufferath I, Pabst D, Rülicke T, Graier WF, Denk H and Zatloukal K: Different Roles of p62 (SQSTM1) isoforms in keratin-related protein aggregation. Int J Mol Sci. 22:62272021. View Article : Google Scholar : PubMed/NCBI

147 

Kageyama S, Saito T, Obata M, Koide RH, Ichimura Y and Komatsu M: Negative Regulation of the Keap1-Nrf2 Pathway by a p62/Sqstm1 Splicing Variant. Mol Cell Biol. 38:e006422018. View Article : Google Scholar : PubMed/NCBI

148 

Guo Q, Wang H, Duan J, Luo W, Zhao R, Shen Y, Wang B, Tao S, Sun Y, Ye Q, et al: An alternatively spliced p62 isoform confers resistance to chemotherapy in breast cancer. Cancer Res. 82:4001–4015. 2022. View Article : Google Scholar : PubMed/NCBI

149 

Otomo C, Metlagel Z, Takaesu G and Otomo T: Structure of the human ATG12~ATG5 conjugate required for LC3 lipidation in autophagy. Nat Struct Mol Biol. 20:59–66. 2013. View Article : Google Scholar :

Related Articles

Journal Cover

June-2024
Volume 64 Issue 6

Print ISSN: 1019-6439
Online ISSN:1791-2423

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Tedesco G, Santarosa M and Maestro R: Beyond self‑eating: Emerging autophagy‑independent functions for the autophagy molecules in cancer (Review). Int J Oncol 64: 57, 2024
APA
Tedesco, G., Santarosa, M., & Maestro, R. (2024). Beyond self‑eating: Emerging autophagy‑independent functions for the autophagy molecules in cancer (Review). International Journal of Oncology, 64, 57. https://doi.org/10.3892/ijo.2024.5645
MLA
Tedesco, G., Santarosa, M., Maestro, R."Beyond self‑eating: Emerging autophagy‑independent functions for the autophagy molecules in cancer (Review)". International Journal of Oncology 64.6 (2024): 57.
Chicago
Tedesco, G., Santarosa, M., Maestro, R."Beyond self‑eating: Emerging autophagy‑independent functions for the autophagy molecules in cancer (Review)". International Journal of Oncology 64, no. 6 (2024): 57. https://doi.org/10.3892/ijo.2024.5645