Editorial Open Access
Copyright ©The Author(s) 2024. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Gastroenterol. Jun 21, 2024; 30(23): 2934-2946
Published online Jun 21, 2024. doi: 10.3748/wjg.v30.i23.2934
From macroautophagy to mitophagy: Unveiling the hidden role of mitophagy in gastrointestinal disorders
Duo-Lun Gao, Meng-Ran Lin, Nan Ge, Jin-Tao Guo, Fan Yang, Si-Yu Sun, Department of Gastroenterology, Engineering Research Center of Ministry of Education for Minimally Invasive Gastrointestinal Endoscopic Techniques, Shengjing Hospital of China Medical University, Shenyang 110004, Liaoning Province, China
ORCID number: Duo-Lun Gao (0009-0007-5377-1924); Meng-Ran Lin (0009-0006-4350-7380); Nan Ge (0000-0002-5764-7054); Jin-Tao Guo (0000-0001-5722-6359); Fan Yang (0000-0002-5032-6450); Si-Yu Sun (0000-0002-7308-0473).
Co-first authors: Duo-Lun Gao and Meng-Ran Lin.
Co-corresponding authors: Fan Yang and Si-Yu Sun.
Author contributions: Gao DL and Lin MR contributed equally to this work; Gao DL, Lin MR, Yang F, Ge N, Guo JT, and Sun SY contributed to this study; Gao DL, Lin MR, and Sun SY designed the overall concept and outline of the manuscript; Yang F, Ge N, and Guo JT contributed to the discussion and design of the manuscript; Gao DL, Lin MR, and Yang F contributed to the writing and editing of the manuscript, illustrations, and literature review; Yang F and Sun SY contributed equally to this work as co-corresponding authors.
Supported by the National Natural Science Foundation of China, No. 82100700; Fundamental Scientific Research Project from the Educational Department of Liaoning Province, No. LJKMZ20221191; High-quality Development Fund Project from the Science and Technology of Liaoning Province, No. 2023JH2 and No. 20200063; and 345 Talent Project of Shengjing Hospital, No. 52-30B.
Conflict-of-interest statement: All the authors report no relevant conflicts of interest for this article.
Open-Access: This article is an open-access article that was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution NonCommercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: https://creativecommons.org/Licenses/by-nc/4.0/
Corresponding author: Si-Yu Sun, MD, Professor, Department of Gastroenterology, Engineering Research Center of Ministry of Education for Minimally Invasive Gastrointestinal Endoscopic Techniques, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Shenyang 110004, Liaoning Province, China. sunsy@sj-hospital.org
Received: March 8, 2024
Revised: May 4, 2024
Accepted: May 23, 2024
Published online: June 21, 2024
Processing time: 104 Days and 11.1 Hours

Abstract

In this editorial, we comment on an article titled “Morphological and biochemical characteristics associated with autophagy in gastrointestinal diseases”, which was published in a recent issue of the World Journal of Gastroenterology. We focused on the statement that “autophagy is closely related to the digestion, secretion, and regeneration of gastrointestinal cells”. With advancing research, autophagy, and particularly the pivotal role of the macroautophagy in maintaining cellular equilibrium and stress response in the gastrointestinal system, has garnered extensive study. However, the significance of mitophagy, a unique selective autophagy pathway with ubiquitin-dependent and independent variants, should not be overlooked. In recent decades, mitophagy has been shown to be closely related to the occurrence and development of gastrointestinal diseases, especially inflammatory bowel disease, gastric cancer, and colorectal cancer. The interplay between mitophagy and mitochondrial quality control is crucial for elucidating disease mechanisms, as well as for the development of novel treatment strategies. Exploring the pathogenesis behind gastrointestinal diseases and providing individualized and efficient treatment for patients are subjects we have been exploring. This article reviews the potential mechanism of mitophagy in gastrointestinal diseases with the hope of providing new ideas for diagnosis and treatment.

Key Words: Mitophagy, Gastrointestinal diseases, Parkin, Autophagic receptor, Colorectal cancer, Gastric cancer, Inflammatory bowel disease

Core Tip: Mitochondria are not only the energy factories of eukaryotic cells but are also closely related to apoptosis, and their dysfunction plays an important role in various gastrointestinal diseases. Mitophagy, an important mechanism to remove damaged mitochondria in vivo, has been found to alleviate the severity of inflammatory bowel diseases and plays a dual role in promoting and inhibiting the occurrence and development of gastrointestinal cancer. A complete understanding of the mitophagy pathway in gastrointestinal diseases will be helpful for developing new treatment strategies. Therefore, we investigated the mechanisms underlying mitophagy and its contribution to gastrointestinal diseases.



INTRODUCTION

Cells are continually exposed to various threats, including pathogens[1,2], genetic mutations[3], and oxidative stress[4,5]. These challenges can lead to organelle dysfunction, subsequently inducing autophagy. There are three different types of autophagy in cells: Macroautophagy, micro-autophagy, and chaperone-mediated autophagy. Macroautophagy is usually called autophagy. Autophagy, as the second type of programmed cell death mechanism, helps cells to remove damaged organelles, pathogens or aggregates and then participates in cell growth, development and differentiation[6]. In the last 10 years, the research on the mechanism of autophagy in infection[7], cancer[8], neurodegenerative diseases[9] and other diseases has made breakthrough progress.

Mitophagy is a selective autophagy pathway, an important branch of autophagy, and has a unique mechanism. Mitophagy was named when Lemasters et al[10] discovered that damaged mitochondria are engulfed by autophagic vesicles and enveloped in microtubule-associated proteins light chain 3 (LC3) in serum. Mitochondria play an important role in aerobic respiration and adenosine triphosphate (ATP) production via oxidative phosphorylation in all eukaryotic cells, and their abnormal functions are closely related to the occurrence and progression of many diseases. Importantly, the mitochondrial quality control system can eliminate damaged mitochondrial proteins or parts of the mitochondrial network and update their components through mitophagy, maintaining a steady state of the mitochondria[11]. In the last 10 years, significant progress has been made in understanding the molecular mechanism and pathophysiological role of mitophagy in human diseases. Several key mitophagy signaling pathways have been identified, including the ubiquitin-dependent pathway mediated by the PINK1-Parkin pathway or other E3 ubiquitin ligases in the mitochondria and the receptor-mediated ubiquitin-independent pathway. With the gradual clarification of the mitophagy pathway, the pathophysiological role of mitophagy in cardiovascular, lung, liver, gastrointestinal, and other organ-related diseases has been explained, providing a new direction for the treatment of diseases[12].

Gastrointestinal diseases include gastrointestinal peristalsis, infectious inflammation (such as Helicobacter pylori infection, cholera, and intestinal parasites), noninfectious inflammation (such as chronic gastroenteritis and Crohn’s disease), and gastrointestinal cancer[13]. Chang et al[14] summarized the morphological and biochemical characteristics of autophagy in gastrointestinal diseases. However, the integration of the unique mechanism of mitophagy and its role in gastrointestinal diseases is still lacking. Recent studies have indicated that mitophagy is closely related to gastrointestinal diseases (Table 1)[15-24]. In this review, we elaborate on the molecular mechanism of mitophagy, summarize its role in the occurrence and progression of inflammatory bowel diseases (IBD), gastric cancer, and colorectal cancer (CRC), and suggest that mitophagy-related pathways may be important targets for clinical treatment.

Table 1 Research progress of mitophagy in gastrointestinal diseases.
Year of publication
Diseases of concern
Problem solved
Ref.
2017SRMDSRMD leads to intestinal mucosal injury: Defective mitochondria with excess O2- production inhibit mitophagy, ultimately triggering Bax-dependent apoptosis and NF-κB-intervened proinflammatory mucosal injury[15]
2020H. pylori associated gastritisThere was a link between H. pylori infection-promoted mitophagy and inflammation[16]
2022-2024Functional dyspepsiaTraditional Chinese medicine can improve gastrointestinal motility disorders, and the mechanism may be related to the inhibition of mitophagy and mitochondria fission[17-19]
2023I/R injuryIncreased NET formation induces inhibition of mitophagy and lipid peroxidation in IECs, leading to ferroptosis of endothelial cells and microvascular dysfunction[20]
2023Malnutrition enteropathyDysregulation of SIRT1 and mTORC1 pathways leads to disruption of autophagy, mitochondrial homeostasis, which triggers intestinal barrier dysfunction and nutrient malabsorption[21]
2023IBDBergapten treatment alleviated NLRP3 inflammasome activation and pyroptosis by promoting mitophagy, suggesting BeG as a potential anti-inflammatory drug for the treatment of inflammatory diseases[93]
2021-2023IBDPolystyrene nanoplastic induced Crohn’s ileitis-like features are related to mitophagy, while Biogenic selenium nanoparticles can alleviate intestinal epithelial barrier damage by regulating mitophagy, which provides new insights for further evaluating the safety of nanoparticles[22-24]
2023IBDNSAIDs induce mitochondrial stress and mitophagy in IECs, which are related to the pathophysiology of Crohn’s disease[89]
2021CRCMitophagy suppresses CRC growth: PINK1 inhibits CRC growth by reducing acetyl-CoA production and activating P53[70]
2023CRCMitophagy promotes CRC growth: GPR176 activates cAMP/PKA signaling pathway and regulate mitophagy to promote the tumorigenesis and progression of CRC[74]
2018Gastric cancerMitophagy promotes gastric cancer growth: Hippo-Yap promotes tumor progression by activating SIRT1/Mfn2/ mitophagy[63]
2023Gastric cancerMitophagy suppresses gastric cancer growth: 8-paradol promoted PINK1/Parkin-associated mitophagy, mediating cell apoptosis[67]
MITOPHAGY MECHANISMS

According to a known mechanism, mitophagy can be divided into ubiquitin-dependent and-independent mitophagy. Ubiquitin-dependent mitophagy is mainly coordinated by the PINK1 protein kinase and Parkin Ubiquitin E3 ligase[25]. Moreover, research has demonstrated other E3 ubiquitin ligases in the mitochondria, such as Ariadne RBR E3 Ub protein ligase 1 (ARIH1)[26], mitochondrial E3 ubiquitin ligase 1 (MUL1)[27], and Gp78[28], that can mediate ubiquitin-dependent mitophagy without relying on Parkin. Notably, ubiquitin-independent mitophagy is mainly mediated by a direct interaction between LC3 and autophagy receptor proteins (Figure 1).

Figure 1
Figure 1 Major Signaling pathways of mitophagy. Parkin-dependent Ubiquitinated Mitophagy Pathway: Following the depolarization of the mitochondrial membrane, PINK1 recruits and activates the E3 ubiquitin ligase activity of Parkin, leading to the formation of ubiquitin chains. These chains then attract a series of autophagy receptors including P62, NDP52, and OPTN. Subsequently, these receptors bind to light chain 3 (LC3), facilitating the connection of the polyubiquitinated mitochondrial outer membrane to the autophagosome membrane, thereby mediating mitophagy. In this process, TANK-binding kinase 1 kinase enhances the affinity of the autophagy receptor for the Ub chain by phosphorylating the receptor. PTEN-L can reduce Parkin phosphorylation and inhibit its E3 ligase activity, thereby inhibiting mitophagy. Parkin-Independent Ubiquitinated Mitophagy Pathway: E3 ubiquitin ligases such as ARIH1, MUL1, and Gp78 may serve as compensatory pathways for Parkin-mediated mitophagy, although the precise mechanisms are yet to be elucidated. Furthermore, PINK1 has the ability to recruit NDP52 and OPTN to mitochondria, thereby directly initiating mitophagy in a Parkin-independent manner. Receptor-Mediated Ubiquitination-Independent Mitophagy Pathway: Proteins including FK506 binding protein 8, BCL2-interacting protein 3 like/NIP3-like protein X (BNIP3L/NIX), BNIP3, FUN14 domain containing 1 (FUNDC1), and Bcl2-L-13 directly bind to LC3, enabling the mitochondrial membrane to connect to the autophagosome membrane and mediate mitophagy. SCF-FBXL4 mediates the ubiquitination and degradation of BNIP3L/NIX and BNIP3, thereby inhibiting mitophagy. Under hypoxia conditions, phosphoglycerate mutase 5 (PGAM5) promotes the dephosphorylation of FUNDC1, enhancing FUNDC1-mediated mitophagy. Conversely, Src kinase and Casein kinase 2 phosphorylate FUNDC1, inhibiting its mitophagy-promoting activity. Although Bcl-xL positively regulates the binding of BNIP3 to LC3, it inhibits FUNDC1-mediated mitophagy by suppressing PGAM5. LC3: Light chain 3; BNIP3L/NIX: BCL2-interacting protein 3 like/NIP3-like protein X; FUNDC1: FUN14 domain containing 1; TBK1: TANK-binding kinase 1.
PINK1-Parkin-mediated ubiquitin-dependent pathway plays an important role in mitophagy

The activation of PINK1 is one of the most upstream events in mitophagy[29]. Under normal physiological conditions, PINK1 is maintained at an extremely low level of PINK1 in the mitochondria and is almost undetectable through a series of input and degradation cycle mechanisms[30]. When affected by pathological factors, such as mitochondrial damage[31] and increased mitochondrial reactive oxygen species[32], it leads to abnormal mitochondrial membrane potential and depolarization. When mitochondria are depolarized, PINK1 is stabilized on the outer membrane of mitochondria (OMM), where it catalyzes the phosphorylation of S65 in the Ub and Ub-like domains of Parkin, thus activating the E3 ubiquitin ligase activity of Parkin[33]. PS65-Ub can further recruit Parkin from the cytoplasm to the OMM, such that the abundance of pS65-Ub gradually increases, eventually leading to the assembly of Ub chains of about 4400 times, establishing a feed-forward loop and finally wrapping the damaged mitochondria with pS65-Ub chains[33]. Additionally, the pS65-Ub chain further recruits autophagy receptors to the damaged OMM[34], and common autophagic receptors that aggregate in the OMM include OPTN[35], NDP52[36], and P62[37]. The activation of the PINK1-Parkin system immediately activates a fraction of TANK-binding kinase 1 (TBK1), which then binds to and phosphorylates the autophagy receptor upon its binding to the Ub chain. This, in turn, enhances the affinity of the autophagy receptor for the Ub chain, extends the duration of autophagy receptors on the OMM, and facilitates mitophagy[38]. These autophagy receptors aggregated on the OMM bind to ATG8 family proteins through the LC3 interaction region (LIR) motif, and with the help of ATG8 family proteins, ubiquitinated OMM attaches to the autophagy membrane[35,37,39]. The autophagy receptors mentioned above are concentrated in the OMM. Notably, a recent study found that the autophagy receptor PHB2, located on the inner membrane of mitochondria, promotes mitophagy mediated by PINK1-Parkin by stabilizing PINK 1 and increasing mitochondrial recruitment of Parkin[40]. PHB2 is ubiquitinated by Parkin, facilitating its interaction with LC3 and accelerating autophagy clearance in damaged mitochondria (Figure 1)[41].

An in-depth study found that many factors, such as TBK1, Phosphatase and Tensin Homolog (PTEN-L), and DJ-1, regulate ubiquitin-dependent mitophagy. In addition to the function of the phosphorylated autophagy receptors mentioned above, TBK1 can promote the downstream steps of mitophagy by phosphorylating S72 in RAB7A through the Ub chain on the OMM[38]. RAB7AS72 is located in the “switch II” domain, which participates in the exchange of guanosine diphosphate/guanosine triphosphate and its interaction with other proteins, thus regulating mitophagy[38]. PTEN-L, a negative regulatory factor of mitophagy located in the OMM, effectively prevents Parkin mitochondrial translocation, reduces Parkin phosphorylation, inhibits its E3 Ligase activity, decreases the level of pSer65-Ub, blocks the feed-forward mechanism of mitophagy, and ultimately inhibits mitophagy[42]. DJ-1 is a 19.9 kda protein encoded by the PARK7 gene. Its deletion does not interfere with the activation of PINK1 or Parkin after mitochondrial depolarization but blocks downstream mitophagy by inhibiting the recruitment of the selective autophagy receptor OPTN to mitochondrial depolarization[43].

Parkin-independent ubiquitin-dependent mitophagy

ARIH1 and Parkin belong to the same RING-in-between-RING family and are widely expressed in cancer cells. Villa et al[26] found that they can ubiquitinate damaged mitochondria in a PINK1-dependent manner, leading to their elimination via autophagy. In addition, Yun et al[27] found that increasing the protein level of MUL1 in Drosophila can counteract the harmful effects caused by the deletion of PINK1 or Parkin, whereas removing MUL1 from PINK1 or Parkin mutants aggravates symptoms, suggesting that MUL1 may participate in the compensatory pathway of the PINK1/Parkin pathway. Furthermore, GP78 is a key E3 ubiquitin ligase involved in endoplasmic reticulum-mediated degradation. In HEK293 cells subjected to Parkin knockdown via siRNA, mitophagy triggered by GP78 remained unaffected, indicating that it operates in Parkin-induced mitophagy independently[28]. In addition, the ubiquitin-binding protein Vps13D has been found in D. melanogaster. Studies have shown that this protein plays a role downstream of PINK1, which is parallel to Parkin in mitophagy, and regulates the localization of ubiquitin and ATG8 around the mitochondria[44]. However, the specific working mechanisms of these proteins are not clear, and further research is needed. Another study showed that PINK1 could recruit the autophagy receptors NDP52 and OPTN into the mitochondria and directly activate mitophagy without relying on Parkin[45].

Receptor-mediated ubiquitin-independent pathway mediates mitophagy

The non-ubiquitin-dependent mitophagy pathway is mediated by the direct interactions between LC3 and mitophagy receptor proteins. These mitophagy receptor proteins include NIP3-like protein X/BCL2-interacting protein 3 like (NIX/BNIP3L), BCL2-interacting protein 3 (BNIP3), FUN14 domain containing 1 (FUNDC1), Bcl2-like protein 13 (Bcl2-L-13), and FK506 binding protein 8 (FKBP8)[46]. They directly bind to LC3 via the LIR region, skip ubiquitination, and directly initiate mitophagy. BNIP3 and BNIP3L/NIX were similar to some extent, indicating that they all contained an atypical BH3 domain. Under the condition of moderate hypoxia (apparent 1%-3% oxygen), hypoxia-inducible factor-1a activates the up-regulation of both transcription factors[47]. Posttranslational modifications regulate mitophagy mediated by BNIP3L/NIX and BNIP3. SCF-FBXL 4 (SkP1/Cul1/F-box protein ubiquitin ligase complex), located in the OMM, mediates the ubiquitination and degradation of BNIP3L/NIX and BNIP3, thus inhibiting mitophagy[48]. However, phosphorylation of serine residues in BNIP3 LIR and BNIP3L LIR promoted mitophagy[49,50]. In addition, the homodimeric form of BNIP3L recruits autophagosomes more robustly than the monomeric form does[51]. Endogenous FUNDC1 is located only in mitochondria. Under hypoxia, phosphoglycerate mutase 5 (PGAM5) promotes FUNDC1 dephosphorylation and FUNDC1-mediated mitophagy, while Src kinase and Casein kinase 2 phosphorylate FUNDC1 and inhibit FUNDC1-mediated mitophagy[52,53]. Notably, Bcl-xL plays different roles in the regulation of mitophagy mediated by BNIP3 and FUNDC1; moreover, it positively regulates the binding between BNIP3 and LC3[49] but inhibits FUNDC1-mediated mitophagy by inhibiting PGAM5[54]. Moreover, iron deficiency can trigger mitophagy mediated by FUNDC1, which is, in turn, mediated by the activation of PGAM5[55]. Bcl2-L-13, a mammalian functional homolog of ATG32, mediates mitophagy by binding to LC3B through the WXXI motif of LIR[56], and the Unc-51-like Kinase (ULK1) complex is necessary for this process[57]. FKBP8 is a significant anti-apoptotic protein featuring a characteristic LIR motif at its N-terminus, which promotes non-ubiquitination of mitophagy via interaction with LC3A. During this process, FKBP8 can exit the mitochondria to evade degradation[46]. In addition to the LIR sequence, FKBP8 contains an LIR motif-like sequence that binds to optical atrophy1 to mediate mitochondrial fragmentation, thus inducing mitophagy[58].

THE ROLE OF MITOPHAGY IN GASTROINTESTINAL DISORDERS

Mitophagy plays an important role in gastric cancer, CRC and IBD. Numerous studies have supported the double-edged sword effect of autophagy in cancer. Specifically, mitophagy plays an inhibitory role in the initial stage of tumorigenesis or cancerous transformation; in contrast, mitophagy provides survival advantages for established and metastatic tumors and can prevent cell death induced by chemotherapy drugs[40,59]. In addition, the anti-inflammatory role of mitophagy in IBD has been extensively studied. The subsequent section will focus on the role of mitophagy in gastric cancer, CRC, and IBD and its potential as a therapeutic target (Tables 2 and 3).

Table 2 Pathways regulating mitophagy in gastric cancer, colorectal cancer, and inflammatory bowel diseases.
Diseases
Molecules
Effects/mechanisms
Significance
Ref.
Gastric cancerYapActivates the SIRT1/Mfn2/mitophagy axis. Knockdown of Yap impairs the expression of adhesive proteins, reduces F-actin expression, and inhibits lamellipodium formationTumor-promoting effects: It contributes to the migration and survival of gastric cancer cells[63]
GGT7Binds with the mitophagy regulator RAB7 to induce mitophagy. GGT7 inhibits ROS production and MAPK cascadesTumor-suppressing effect: It inhibits the growth, G1-S phase transition and migration of gastric cancer cells[64]
CRCpiR823Promotes ubiquitination and proteasome-dependent degradation of PINK1, thereby inhibiting mitophagyTumor-promoting
effects: It is involved in CRC tumorigenesis
[71]
MST1Inhibits mitophagy through the JNK/p53/BNIP3 pathway, leading to oxidative stress and initiating mitochondria-mediated apoptosisTumor-suppressing effect: It inhibits tumor proliferation[73]
GPR176Inhibits mitophagy through the cAMP/PKA/BNIP3L axisTumor-promoting effects: It promotes the development of CRC[74]
IBDNR1D1Acts as a positive regulator of BNIP3 expression, promoting mitophagy and maintaining the immune homeostasis of IECsInhibitory effect on colitis: It reduces the severity and progression of colitis[88]
Table 3 Drugs affecting mitophagy in gastric cancer, colorectal cancer, and inflammatory bowel diseases.
Diseases
Drugs
Effects/mechanisms
Significance
Ref.
Gastric cancerTNFαActivates Parkin-dependent mitophagy, and excessive mitophagy blocks mitochondrial apoptosisRelates to the resistance of gastric cancer cells to TNFα[65]
MetforminActivates AMPK signaling pathway and up-regulates the expression of mitophagy-related proteins PINK1, Parkin, and LC3BPromotes the resistance of gastric cancer cells to cisplatin[66]
CRCMito-CP, mito-metforminInduces the release of ULK1, which promotes mitophagyTumor-suppressing effect: It inhibits tumor proliferation[78]
Aloe gel glucomannanActivates PINK1/Parkin pathway to promote mitophagy; it activates the transcription factor EB to induce mitochondrial damage and ROS generationTumor-suppressing effect: It Inhibits tumor proliferation[79]
δ-valbetaine Activates mitophagy through the PINK1/Parkin pathwayTumor-suppressing effect: Inducing apoptosis of CRC cells[80]
OxymatrineInduces mitophagy and reduces NLRP3 inflammasome activation in CRC cellsTumor-suppressing effect: Inhibit the growth and migration of CRC cells[81]
Small molecule andrographolide Inactivates the NLRP3 inflammasome induced by mitophagy in macrophagesAlleviates colitis progression and reduces the risk of colitis-related cancers[82]
IBDSodium butyrateActivates Pink1/Parkin expression to promote mitophagy; it inhibits phosphorylation of NF-κB and activation of the NLRP3 inflammasomeHas an inhibitory effect on ulcerative colitis[92]
NSAIDsInduces mitochondrial stress which leads to impaired mitophagyProinflammatory effects[89]
BergaptenPromotes mitophagy and maintains mitochondrial homeostasis to inhibit NLRP3 inflammasome activation and pyroptosisAnti-inflammatory activity[93]
Ginsenoside RdActivates AMPK/ULK1/p62 signaling pathway to trigger mitophagy, thereby inhibiting NLRP3 inflammasomeAnti-inflammatory activity[94]
Gastric cancer and mitophagy

Gastric cancer is the fifth most common cancer worldwide, with a high mortality rate[60]. The use of traditional endoscopy and ultrasonic endoscopy (EUS) facilitates the diagnosis of gastric cancer in the early stages and the evaluation of invasion depth, which is beneficial for improving, to some extent, its outcomes[61]. The inclusion of therapeutic EUS in the treatment of complex hepatobiliary, pancreatic, and gastrointestinal diseases has significantly enhanced the quality of life for tumor patients[62]. Defining the survival, migration, treatment, and drug resistance mechanisms of gastric cancer has always been a topic of great concern (Figure 2). In view of the survival and migration of gastric cancer cells, experiments have demonstrated that Sirtuin 1 (SIRT1), a Yes-associated protein (Yap) signal, activates mitophagy and promotes mitochondrial homeostasis[63]. The Yap-STRT1 mitophagy pathway blocks the caspase-9-related apoptosis axis, enhances cell migration based on F-actin, and participates in the migration and survival of gastric cancer cells[63]. Furthermore, an additional study demonstrated that gamma-glutamyltransferase 7 (GGT7) was significantly downregulated in gastric cancer cells, markedly inhibiting their growth, G1-S transition, and migration ability. This inhibition may be associated with the occurrence of GGT7-induced mitophagy[64].

Figure 2
Figure 2 Pathways regulating mitophagy in colorectal cancer, gastric cancer, and inflammatory bowel disease. A: Colorectal cancer cells: GPR176 recruits GNAS to inhibit BCL2-interacting protein 3 like through the AC/cAMP/PKA pathway, thereby suppressing mitophagy. Additionally, MST1 activates the c-Jun N-terminal kinase (JNK) pathway, up-regulating P53 expression, which in turn inhibits BNIP3 transcription and activity, leading to mitophagy arrest; B: Gastric cancer cells: Interactions between GGT7 and Rab7 promote mitophagy. Yap activates sirtuin 1, enhancing Mfn2 expression and sustaining mitophagy. JNK upregulates Parkin to activate mitophagy; however, overexpression of NR4A1 inhibits mitophagy by suppressing JNK. Furthermore, the AMPK/ULK1/Parkin axis also supports mitophagy; C: Macrophages in inflammatory bowel disease: Damaged or stressed mitochondria in macrophages release mtDNA and reactive oxygen species, which directly contribute to NOD-like receptor thermal protein domain associated protein 3 (NLRP3) inflammasome formation and activate NF-κB via the TLR9 pathway, triggering NLRP3 inflammasome activation and subsequent Caspase-1 activity. This results in the production of IL-1β and IL-18. Enhancing the AMPK-ULK1-P62 axis-driven mitophagy efficiently removes damaged mitochondria, inhibiting NLRP3 inflammasome activation and exerting anti-inflammatory effects. CRC: Colorectal cancer cell; NLRP3: NOD-like receptor thermal protein domain associated protein 3; SIRT1: Sirtuin 1; JNK: c-Jun N-terminal kinase.

Drug resistance in gastric cancer cells is primarily manifested through tumor necrosis factor α (TNFα) and chemotherapy drugs. As a pro-inflammatory and pro-apoptotic cytokine, TNFα is an important host defense system against the progress of gastric cancer; however, its therapeutic effect is limited by drug resistance[65]. Experiments indicate that TNFα treatment initiates Parkin-dependent mitophagy, and excessive mitophagy prevents mitochondrial apoptosis, mitigating the toxic effect of TNFα on cancer cells[65]. Furthermore, the suppression of mitophagy to enhance the responsiveness of gastric cancer cells to TNFα might present a novel approach to treating gastric cancer. Cisplatin remains the principal medication for managing gastric cancer; however, it demonstrates significant drug resistance, posing a crucial challenge that necessitates immediate attention in clinical settings. Moreover, research has indicated that metformin, an antidiabetic medication, may reduce the sensitivity of cancer cells to cisplatin[66]. Metformin stimulates the phosphorylation of AMPK (Thr172) and increases the expression of mitophagy markers, including Parkin and PINK1, in an AMPK signal-dependent manner, significantly increasing the mitophagy of cancer cells, reducing ATP production, and protecting gastric cancer cells from the therapeutic toxicity of cisplatin[66]. To explore the mechanism behind metformin reducing the sensitivity of gastric cancer cells to cisplatin and provide new possibilities for solving the cisplatin resistance of gastric cancer patients.

Some studies have suggested potential drugs and strategies for treating gastric cancer associated with mitophagy: 8-paradol, a phenolic compound derived from ginger, can induce cell apoptosis by enhancing mitophagy via the PINK1-Parkin pathway. Furthermore, the suppression of mitophagy using chloroquine ameliorates mitochondrial dysfunction and apoptosis triggered by 8-paradol. This observation underscores the pivotal role of mitophagy in the anticancer activity elicited by 8-paradol[67]. The Newcastle disease virus, a paramyxovirus, is utilized in cancer treatment. It can induce mitochondrial damage, elevate mitochondrial reactive oxygen species, and disrupt electron transport chain function. Consequently, this leads to the activation of the PINK1-Parkin pathway and the formation of a ubiquitin chain with Mitofusin 2. Furthermore, the molecular receptor p62 recognizes damaged mitochondria, mediates mitophagy, and regulates cancer cells[68].

CRC and mitophagy

According to the 2020 global cancer data, CRC is now the second leading cause of cancer-related mortality globally[60]. While surgical intervention and adjuvant chemotherapy can effectively treat early-stage CRC, a significant proportion (25%-50%) of patients progress to metastasis, resulting in a dismal 5-year survival rate of approximately 14%[69]. Hence, delving deeper into the molecular mechanisms underlying CRC tumorigenesis and progression is imperative to develop new therapeutic strategies. Several studies have underscored the critical role of mitophagy in these processes (Figure 2). Thus, this subsection aims to comprehensively review how the mitophagy pathway contributes to CRC development, drug resistance, and treatment.

The role of PINK1 in inhibiting tumor growth within CRC has been elucidated. Yin et al[70] explored mouse colon cancer cells and found that PINK1 overexpression not only promoted mitophagy and decreased glycolysis through the activation of the p53 signaling pathway but also inhibited acetyl-CoA production within tumor cells, thus impeding tumor growth. The non-coding RNA piR823 interacts with PINK1, promoting its ubiquitination and proteasome-dependent degradation, thereby hindering mitophagy[71]. However, evidence suggests that PINK1 promotes survival in CRC. Chen et al[72] demonstrated that disruption of the mitophagy pathway due to PINK1 KD leads to a cytosolic iron imbalance, which can be rescued by ferritophagy activation through nuclear receptor coactivator 4 overexpression. These findings suggest that PINK1 regulates intracellular iron availability in conjunction with mitophagy and ferroautophagy, maintaining intracellular iron homeostasis, which is vital for supporting CRC cell survival and growth.

The receptor-mediated mitophagy pathway assumes a dual role in CRC development. Mammalian Ste20-like kinase 1, found to be down-regulated in CRC, inhibits mitophagy via the c-Jun N-terminal kinase (JNK)/p53/BNIP3 pathway, thereby inducing oxidative stress and initiating mitochondrial-mediated apoptosis, which contributes to the inhibition of tumor growth[73]. In contrast, the GPR176/GNAS complex inhibits mitophagy via the cAMP/PKA/BNIP3L axis, thereby promoting CRC development[74]. In addition to promoting or inhibiting tumor cells, Ziegler et al[75] found that elevated levels of mitophagy in intestinal epithelial cells induced adaptive immune responses in CD8+ T cells, providing a therapeutic target for tumor immunity.

Further, mitophagy has been implicated in drug resistance and radioresistance of CRC. Yan et al[76] observed that the level of mitophagy and the expression of BNIP3L were significantly increased in cancer stem cells (CSCs) after treatment with doxorubicin (DXR); however, silencing BNIP3L significantly inhibited mitophagy and enhanced the sensitivity of CSCs to DXR, suggesting that mitophagy is involved in DXR resistance in CSCs. Wei et al[77] proposed a mechanism by which mitophagy contributes to CRC radioresistance. Notably, excessive activation of mitophagy leads to decreased RING1b expression, which culminates in the deubiquitination of histone H2A at K119, thereby facilitating enhanced repair of radiation-induced DNA damage.

Given the significant role of mitophagy in both tumorigenesis and the progression of CRC, researchers have identified mitophagy as a promising therapeutic target for CRC. Specifically, in KRAS-mutant CRC, Mito-CP and Mito-metformin induce the release of ULK1, which promotes mitophagy and serves an anti-proliferative function[78]. Aloe gel glucomannan was found to induce mitochondrial damage and reactive oxygen species (ROS) generation, thereby inducing cytotoxic mitophagy in colon cancer cells through the PINK1/Parkin pathway and activation of the transcription factor EB[79]. Similarly, δ-valbetaine induces apoptosis in colon cancer cells by activating mitophagy through the PINK1/Parkin pathway[80]. Additionally, traditional Chinese medicine extracts have been shown to play an important role in the treatment of CRC. Oxymatrine treatment induces mitophagy in CRC cells and reduces NOD-like receptor thermal protein domain associated protein 3 (NLRP3) inflammasome activation, inhibiting the growth and migration of CRC cells in vitro and in vivo[81]. However, the small molecule andrographolide (Andro) was shown to inactivate the NLRP3 inflammasome induced by mitophagy in macrophages, helping to mitigate colitis progression and tumor burden, thereby reducing the risk of colitis-associated cancer[82].

IBD and mitophagy

IBD is a group of autoimmune diseases characterized by gastrointestinal inflammation, primarily ulcerative colitis (UC) and Crohn’s disease. In recent years, the incidence and prevalence of IBD in Asian populations have gradually increased[83]. Several studies have reported that mitochondria are related to the inflammatory response (Figure 2). When mitochondria are damaged, mtDNA and mtROS are released. mtDNA can not only activate NLRP3 inflammasome[84] but also trigger the toll-like receptor 9 pathway to induce an NF-κB and MAPK inflammatory cascade[85]. Moreover, ROS affect the secretion of inflammatory cytokines[86]. Finally, mitophagy removes damaged mitochondria, suggesting that it may be a protective factor for IBD.

NIX is an important receptor protein that mediates mitophagy. Vincent et al[87] found that NIX expression was upregulated in the intestinal epithelial cells (IECs) of patients with UC, and compared to wild-type mice, NIX-/- mice exhibited stronger inflammatory characteristics and loss of mucosal integrity when experimental colitis occurred. Moreover, research has demonstrated that the expression level of the circadian clock gene NR1D1 is reduced in patients with UC, and NR1D1 knockout results in a disruption of IECs immune homeostasis and a diminished mitophagy. Subsequent studies have identified that NR1D1 positively influences the expression of the autophagy receptor BNIP3, thereby enhancing mitophagy[88]. The use of non-steroidal anti-inflammatory drugs (NSAIDs) has been identified as a risk factor for IBD. This condition induces mitochondrial stress in IECs, resulting in impaired mitophagy. Such impairment leads to the release of mitochondrial damage-associated molecular patterns with pro-inflammatory potential. These mitochondrial components then act as pro-inflammatory molecules[89].

The maintenance of mitochondrial homeostasis mediated by mitophagy limits the excessive activation of the NLRP3 inflammasome[90], which plays a key role in colitis[91].

Several substances have been demonstrated to harness this process and improve outcomes in IBD. For example, sodium butyrate is effective in suppressing UC because it inhibits the phosphorylation of NF-κB and activates the NLRP3 inflammasome. Moreover, it also enhances mitophagy through the activation of PINK1/Parkin expression[92]. Similarly, Bergapten, a plant-derived hormone with anti-inflammatory properties, has been shown to inhibit NLRP3 inflammasome activation and pyroptosis in a mouse model of intestinal inflammation, further supporting mitochondrial health by facilitating mitophagy[93]. In addition, Ginsenoside Rd initiates mitophagy by activating the AMPK/ULK1/p62 signaling pathway, which in turn inhibits the NLRP3 inflammasome[94]. Furthermore, probiotics have been demonstrated to ameliorate UC. Specifically, Lacidophilus acidophilus enhances the levels of short-chain fatty acids, thereby stimulating the mitophagy/NLRP3 inflammasome pathway. This activation helps to maintain inflammatory homeostasis both in vivo and in vitro and contributes to the improvement of intestinal barrier function[95]. However, mitophagy-related pathway proteins may also play a role in IBD. Parkin, an E3 ubiquitin ligase, has been identified by Ma et al[96] as playing a significant role in the context of IBD, with the vitamin D receptor (VDR) acting as a crucial inhibitory regulator. Specifically, Parkin escalates the incidence of colitis and severe inflammation by facilitating VDR degradation via the p62-related autophagy-lysosomal pathway.

CONCLUSION
Mitochondria and mitophagy

Along with acting as the energy factories of cells, mitochondria play crucial roles in cell signal transduction, calcium regulation, reactive oxygen species production, cellular protein homeostasis, anti-inflammatory responses, apoptosis, and intercellular mitochondrial transfer[97-100]. The diverse and important functions of mitochondria are the basis for maintaining cell homeostasis. Under the supervision of quality control system, mitochondria will undergo continuous fission and fusion cycles in cells to maintain their shape, network and inheritance[101]. When mitochondrial function is irreparably damaged or under specific stress conditions such as hypoxia or nutritional deprivation, mitophagy is activated. This process selectively promotes the degradation of mitochondria via the autophagy-lysosome pathway[102]. Mitophagy, as a mechanism to maintain the quality and quantity of mitochondria, is involved in the pathophysiological processes such as cell growth, cell differentiation, cell aging and apoptosis.

Advances in mitophagy

Autophagy is an energy-intensive process. Excessive regulation of autophagy can result in cellular homeostasis imbalance, leading to unnecessary degradation and damage of organelles[103]. Therefore, it appears to be a more sensible strategy to develop selective autophagy modulators for the treatment of gastrointestinal diseases. Mitophagy regulators developed over the past decade have shown a certain efficacy in gastrointestinal disease models. In the context of gastric cancer and CRC, mitophagy serves dual functions: It not only maintains mitochondrial homeostasis to prevent cancer but also confers survival advantages and enhances drug resistance in cancer cells, influenced by the complex tumor microenvironment. Regulating mitophagy can be an effective strategy to prevent cancer, halt its progression, and enhance treatment efficacy. In IBD, mitochondrial damage can prompt the release of a series of inflammatory factors, which in turn exacerbate intestinal tissue damage. Moreover, mitophagy plays a crucial role in inhibiting the progression of IBD by removing damaged mitochondria. Thus, developing mitophagy inducers may represent a novel therapeutic approach for IBD.

Problems to be solved

However, the research to date has demonstrated a correlation between mitophagy and several conditions, such as gastric cancer, intestinal cancer, and IBD, thus offering a new and viable direction for treatment. Nevertheless, numerous challenges remain. The signaling mechanisms of mitophagy are intricate and vary across different tissues, developmental stages, and states of stress or metabolism. Therefore, what are the temporal and spatial regulations of mitophagy under various pathophysiological conditions? Numerous experiments have demonstrated the dual role of mitophagy in cancer; however, the critical question remains: What is the threshold between its inhibitory and promotive effects? Currently, many mitophagy inducers are mitochondrial decoupling agents or mitochondrial toxins developed from in vitro experiments. Are these agents clinically effective? Is the pharmacological activity of these known regulators solely attributed to mitophagy regulation? Thus, identifying biomarkers and developing detection methods that can reliably and specifically measure mitophagy flux are essential. This will ensure precise regulation of mitophagy and facilitate the practical evaluation of therapeutic effects. Furthermore, the mechanism of mitophagy in additional gastrointestinal diseases, including gastrointestinal peristalsis, infectious inflammation, and chronic inflammation of the gastrointestinal tract, remains largely unexplored and warrants further investigation.

Future application fields

In summary, further analysis of the molecular mechanisms of mitophagy and its role in regulating the onset and progression of gastrointestinal diseases is warranted. Developing small-molecule drugs that target mitophagy for the treatment of gastrointestinal diseases represents a novel approach. With the appearance of an in vivo mitophagy imaging system[12] and a mitophagy modulator characterization system[103], it has gradually become possible to study and verify mitophagy modulators in disease animal models and to characterize drugs. Although there are many problems that need to be solved urgently in mitophagy, it is undeniable that targeted mitophagy is a promising treatment for gastrointestinal diseases.

ACKNOWLEDGEMENTS

The authors extend the deepest appreciation to Dr. Si-Yu Sun and Dr. Fan Yang, who have made genuine contributions to the manuscript and endorsed the conclusion.

Footnotes

Provenance and peer review: Invited article; Externally peer reviewed.

Peer-review model: Single blind

Specialty type: Gastroenterology and hepatology

Country of origin: China

Peer-review report’s classification

Scientific Quality: Grade B, Grade C, Grade C

Novelty: Grade B, Grade B, Grade B

Creativity or Innovation: Grade B, Grade B, Grade C

Scientific Significance: Grade B, Grade B, Grade B

P-Reviewer: Elkady N, Egypt; Emran TB, Bangladesh S-Editor: Chen YL L-Editor: A P-Editor: Zheng XM

References
1.  Lesa KN, Khandaker MU, Mohammad Rashed Iqbal F, Sharma R, Islam F, Mitra S, Emran TB. Nutritional Value, Medicinal Importance, and Health-Promoting Effects of Dietary Mushroom (Pleurotus ostreatus). J Food Quality. 2022;2022:1-9.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in F6Publishing: 3]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
2.  Mitra S, Paul S, Roy S, Sutradhar H, Bin Emran T, Nainu F, Khandaker MU, Almalki M, Wilairatana P, Mubarak MS. Exploring the Immune-Boosting Functions of Vitamins and Minerals as Nutritional Food Bioactive Compounds: A Comprehensive Review. Molecules. 2022;27.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 19]  [Article Influence: 9.5]  [Reference Citation Analysis (0)]
3.  Mitra S, Emran TB, Chandran D, Zidan BMRM, Das R, Mamada SS, Masyita A, Salampe M, Nainu F, Khandaker MU, Idris AM, Simal-Gandara J. Cruciferous vegetables as a treasure of functional foods bioactive compounds: Targeting p53 family in gastrointestinal tract and associated cancers. Front Nutr. 2022;9:951935.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
4.  Shi M, Gu J, Wu H, Rauf A, Emran TB, Khan Z, Mitra S, Aljohani ASM, Alhumaydhi FA, Al-Awthan YS, Bahattab O, Thiruvengadam M, Suleria HAR. Phytochemicals, Nutrition, Metabolism, Bioavailability, and Health Benefits in Lettuce-A Comprehensive Review. Antioxidants (Basel). 2022;11.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 26]  [Article Influence: 13.0]  [Reference Citation Analysis (0)]
5.  Wang C, Li M, Duan X, Abu-izneid T, Rauf A, Khan Z, Mitra S, Emran TB, Aljohani ASM, Alhumaydhi FA, Thiruvengadam M, Suleria HAR. Phytochemical and Nutritional Profiling of Tomatoes; Impact of Processing on Bioavailability - A Comprehensive Review. Food Res Int. 2022;39:5986-6010.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
6.  Li X, He S, Ma B. Autophagy and autophagy-related proteins in cancer. Mol Cancer. 2020;19:12.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 304]  [Cited by in F6Publishing: 755]  [Article Influence: 188.8]  [Reference Citation Analysis (0)]
7.  Debnath J, Gammoh N, Ryan KM. Autophagy and autophagy-related pathways in cancer. Nat Rev Mol Cell Biol. 2023;24:560-575.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 105]  [Cited by in F6Publishing: 152]  [Article Influence: 152.0]  [Reference Citation Analysis (0)]
8.  Matsuzawa-Ishimoto Y, Hwang S, Cadwell K. Autophagy and Inflammation. Annu Rev Immunol. 2018;36:73-101.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 169]  [Cited by in F6Publishing: 225]  [Article Influence: 32.1]  [Reference Citation Analysis (0)]
9.  Billings LM, Oddo S, Green KN, McGaugh JL, LaFerla FM. Intraneuronal Abeta causes the onset of early Alzheimer's disease-related cognitive deficits in transgenic mice. Neuron. 2005;45:675-688.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 861]  [Cited by in F6Publishing: 930]  [Article Influence: 48.9]  [Reference Citation Analysis (0)]
10.  Lemasters JJ. Selective mitochondrial autophagy, or mitophagy, as a targeted defense against oxidative stress, mitochondrial dysfunction, and aging. Rejuvenation Res. 2005;8:3-5.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 839]  [Cited by in F6Publishing: 916]  [Article Influence: 48.2]  [Reference Citation Analysis (0)]
11.  Pickles S, Vigié P, Youle RJ. Mitophagy and Quality Control Mechanisms in Mitochondrial Maintenance. Curr Biol. 2018;28:R170-R185.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 813]  [Cited by in F6Publishing: 1170]  [Article Influence: 234.0]  [Reference Citation Analysis (0)]
12.  Wang S, Long H, Hou L, Feng B, Ma Z, Wu Y, Zeng Y, Cai J, Zhang DW, Zhao G. The mitophagy pathway and its implications in human diseases. Signal Transduct Target Ther. 2023;8:304.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 21]  [Article Influence: 21.0]  [Reference Citation Analysis (0)]
13.  Wang T, Liu K, Wen L, Yang Y, Yin X, Chen Y, He Y, Yang M, Wei Y, Wang B, Chen D. Autophagy and Gastrointestinal Diseases. Adv Exp Med Biol. 2020;1207:529-556.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 12]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
14.  Chang YF, Li JJ, Liu T, Wei CQ, Ma LW, Nikolenko VN, Chang WL. Morphological and biochemical characteristics associated with autophagy in gastrointestinal diseases. World J Gastroenterol. 2024;30:1524-1532.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
15.  De R, Mazumder S, Sarkar S, Debsharma S, Siddiqui AA, Saha SJ, Banerjee C, Nag S, Saha D, Bandyopadhyay U. Acute mental stress induces mitochondrial bioenergetic crisis and hyper-fission along with aberrant mitophagy in the gut mucosa in rodent model of stress-related mucosal disease. Free Radic Biol Med. 2017;113:424-438.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 26]  [Cited by in F6Publishing: 19]  [Article Influence: 2.7]  [Reference Citation Analysis (0)]
16.  Piao JY, Kim SJ, Kim DH, Park JH, Park SA, Han HJ, Na HK, Yoon K, Lee HN, Kim N, Hahm KB, Surh YJ. Helicobacter pylori infection induces STAT3 phosphorylation on Ser727 and autophagy in human gastric epithelial cells and mouse stomach. Sci Rep. 2020;10:15711.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in F6Publishing: 19]  [Article Influence: 4.8]  [Reference Citation Analysis (0)]
17.  Jia Q, Li L, Wang X, Wang Y, Jiang K, Yang K, Cong J, Cai G, Ling J. Hesperidin promotes gastric motility in rats with functional dyspepsia by regulating Drp1-mediated ICC mitophagy. Front Pharmacol. 2022;13:945624.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 8]  [Reference Citation Analysis (0)]
18.  Zhang J, Wang X, Wang F, Tang X. Xiangsha Liujunzi Decoction improves gastrointestinal motility in functional dyspepsia with spleen deficiency syndrome by restoring mitochondrial quality control homeostasis. Phytomedicine. 2022;105:154374.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 7]  [Article Influence: 3.5]  [Reference Citation Analysis (0)]
19.  Wang X, Liu X, Wang Y, Yang K, Yeertai Y, Jia Q, Li L, Jiang K, Du G, Ling J. Chaihu Shugan Powder inhibits interstitial cells of cajal mitophagy through USP30 in the treatment of functional dyspepsia. J Ethnopharmacol. 2024;323:117695.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
20.  Chu C, Wang X, Yang C, Chen F, Shi L, Xu W, Wang K, Liu B, Wang C, Sun D, Ding W. Neutrophil extracellular traps drive intestinal microvascular endothelial ferroptosis by impairing Fundc1-dependent mitophagy. Redox Biol. 2023;67:102906.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 9]  [Reference Citation Analysis (0)]
21.  Ling C, Versloot CJ, Arvidsson Kvissberg ME, Hu G, Swain N, Horcas-Nieto JM, Miraglia E, Thind MK, Farooqui A, Gerding A, van Eunen K, Koster MH, Kloosterhuis NJ, Chi L, ChenMi Y, Langelaar-Makkinje M, Bourdon C, Swann J, Smit M, de Bruin A, Youssef SA, Feenstra M, van Dijk TH, Thedieck K, Jonker JW, Kim PK, Bakker BM, Bandsma RHJ. Rebalancing of mitochondrial homeostasis through an NAD(+)-SIRT1 pathway preserves intestinal barrier function in severe malnutrition. EBioMedicine. 2023;96:104809.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 4]  [Article Influence: 4.0]  [Reference Citation Analysis (0)]
22.  Dou X, Qiao L, Song X, Chang J, Pi S, Zhang X, Zeng X, Zhu L, Xu C. Biogenic selenium nanoparticles alleviate intestinal epithelial barrier injury by regulating mitochondria-lysosome crosstalk. Food Funct. 2023;14:4891-4904.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
23.  Yan S, Qiao L, Dou X, Song X, Chen Y, Zhang B, Xu C. Biogenic selenium nanoparticles by Lactobacillus casei ATCC 393 alleviate the intestinal permeability, mitochondrial dysfunction and mitophagy induced by oxidative stress. Food Funct. 2021;12:7068-7080.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 11]  [Article Influence: 3.7]  [Reference Citation Analysis (0)]
24.  Xu D, Ma Y, Peng C, Gan Y, Wang Y, Chen Z, Han X, Chen Y. Differently surface-labeled polystyrene nanoplastics at an environmentally relevant concentration induced Crohn's ileitis-like features via triggering intestinal epithelial cell necroptosis. Environ Int. 2023;176:107968.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 15]  [Reference Citation Analysis (0)]
25.  Bordi M, Darji S, Sato Y, Mellén M, Berg MJ, Kumar A, Jiang Y, Nixon RA. mTOR hyperactivation in Down Syndrome underlies deficits in autophagy induction, autophagosome formation, and mitophagy. Cell Death Dis. 2019;10:563.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 50]  [Cited by in F6Publishing: 51]  [Article Influence: 10.2]  [Reference Citation Analysis (0)]
26.  Villa E, Proïcs E, Rubio-Patiño C, Obba S, Zunino B, Bossowski JP, Rozier RM, Chiche J, Mondragón L, Riley JS, Marchetti S, Verhoeyen E, Tait SWG, Ricci JE. Parkin-Independent Mitophagy Controls Chemotherapeutic Response in Cancer Cells. Cell Rep. 2017;20:2846-2859.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 159]  [Cited by in F6Publishing: 168]  [Article Influence: 24.0]  [Reference Citation Analysis (0)]
27.  Yun J, Puri R, Yang H, Lizzio MA, Wu C, Sheng ZH, Guo M. MUL1 acts in parallel to the PINK1/parkin pathway in regulating mitofusin and compensates for loss of PINK1/parkin. Elife. 2014;3:e01958.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 181]  [Cited by in F6Publishing: 220]  [Article Influence: 22.0]  [Reference Citation Analysis (0)]
28.  Fu M, St-Pierre P, Shankar J, Wang PT, Joshi B, Nabi IR. Regulation of mitophagy by the Gp78 E3 ubiquitin ligase. Mol Biol Cell. 2013;24:1153-1162.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 138]  [Cited by in F6Publishing: 144]  [Article Influence: 13.1]  [Reference Citation Analysis (0)]
29.  Gan ZY, Callegari S, Cobbold SA, Cotton TR, Mlodzianoski MJ, Schubert AF, Geoghegan ND, Rogers KL, Leis A, Dewson G, Glukhova A, Komander D. Activation mechanism of PINK1. Nature. 2022;602:328-335.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 20]  [Cited by in F6Publishing: 54]  [Article Influence: 27.0]  [Reference Citation Analysis (0)]
30.  Yamano K, Youle RJ. PINK1 is degraded through the N-end rule pathway. Autophagy. 2013;9:1758-1769.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 396]  [Cited by in F6Publishing: 455]  [Article Influence: 41.4]  [Reference Citation Analysis (0)]
31.  Sandoval-Acuña C, Torrealba N, Tomkova V, Jadhav SB, Blazkova K, Merta L, Lettlova S, Adamcová MK, Rosel D, Brábek J, Neuzil J, Stursa J, Werner L, Truksa J. Targeting Mitochondrial Iron Metabolism Suppresses Tumor Growth and Metastasis by Inducing Mitochondrial Dysfunction and Mitophagy. Cancer Res. 2021;81:2289-2303.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 16]  [Cited by in F6Publishing: 44]  [Article Influence: 14.7]  [Reference Citation Analysis (0)]
32.  Basit F, van Oppen LM, Schöckel L, Bossenbroek HM, van Emst-de Vries SE, Hermeling JC, Grefte S, Kopitz C, Heroult M, Hgm Willems P, Koopman WJ. Mitochondrial complex I inhibition triggers a mitophagy-dependent ROS increase leading to necroptosis and ferroptosis in melanoma cells. Cell Death Dis. 2017;8:e2716.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 207]  [Cited by in F6Publishing: 346]  [Article Influence: 49.4]  [Reference Citation Analysis (0)]
33.  Malpartida AB, Williamson M, Narendra DP, Wade-Martins R, Ryan BJ. Mitochondrial Dysfunction and Mitophagy in Parkinson's Disease: From Mechanism to Therapy. Trends Biochem Sci. 2021;46:329-343.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 91]  [Cited by in F6Publishing: 226]  [Article Influence: 56.5]  [Reference Citation Analysis (0)]
34.  Tan HWS, Lu G, Dong H, Cho YL, Natalia A, Wang L, Chan C, Kappei D, Taneja R, Ling SC, Shao H, Tsai SY, Ding WX, Shen HM. A degradative to secretory autophagy switch mediates mitochondria clearance in the absence of the mATG8-conjugation machinery. Nat Commun. 2022;13:3720.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 39]  [Article Influence: 19.5]  [Reference Citation Analysis (0)]
35.  Qiu Y, Wang J, Li H, Yang B, He Q, Weng Q. Emerging views of OPTN (optineurin) function in the autophagic process associated with disease. Autophagy. 2022;18:73-85.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in F6Publishing: 40]  [Article Influence: 13.3]  [Reference Citation Analysis (0)]
36.  Fang EF, Hou Y, Palikaras K, Adriaanse BA, Kerr JS, Yang B, Lautrup S, Hasan-Olive MM, Caponio D, Dan X, Rocktäschel P, Croteau DL, Akbari M, Greig NH, Fladby T, Nilsen H, Cader MZ, Mattson MP, Tavernarakis N, Bohr VA. Mitophagy inhibits amyloid-β and tau pathology and reverses cognitive deficits in models of Alzheimer's disease. Nat Neurosci. 2019;22:401-412.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 627]  [Cited by in F6Publishing: 939]  [Article Influence: 187.8]  [Reference Citation Analysis (0)]
37.  Yamada T, Murata D, Adachi Y, Itoh K, Kameoka S, Igarashi A, Kato T, Araki Y, Huganir RL, Dawson TM, Yanagawa T, Okamoto K, Iijima M, Sesaki H. Mitochondrial Stasis Reveals p62-Mediated Ubiquitination in Parkin-Independent Mitophagy and Mitigates Nonalcoholic Fatty Liver Disease. Cell Metab. 2018;28:588-604.e5.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 134]  [Cited by in F6Publishing: 172]  [Article Influence: 28.7]  [Reference Citation Analysis (0)]
38.  Heo JM, Ordureau A, Swarup S, Paulo JA, Shen K, Sabatini DM, Harper JW. RAB7A phosphorylation by TBK1 promotes mitophagy via the PINK-PARKIN pathway. Sci Adv. 2018;4:eaav0443.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 114]  [Cited by in F6Publishing: 125]  [Article Influence: 20.8]  [Reference Citation Analysis (0)]
39.  Chen M, Chen Z, Wang Y, Tan Z, Zhu C, Li Y, Han Z, Chen L, Gao R, Liu L, Chen Q. Mitophagy receptor FUNDC1 regulates mitochondrial dynamics and mitophagy. Autophagy. 2016;12:689-702.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 233]  [Cited by in F6Publishing: 341]  [Article Influence: 42.6]  [Reference Citation Analysis (0)]
40.  Yan C, Gong L, Chen L, Xu M, Abou-Hamdan H, Tang M, Désaubry L, Song Z. PHB2 (prohibitin 2) promotes PINK1-PRKN/Parkin-dependent mitophagy by the PARL-PGAM5-PINK1 axis. Autophagy. 2020;16:419-434.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 105]  [Cited by in F6Publishing: 213]  [Article Influence: 42.6]  [Reference Citation Analysis (0)]
41.  Sun S, Hou H, Ma G, Ma Q, Li N, Zhang L, Dong C, Cao M, Tam KY, Ying Z, Wang H. The interaction between E3 ubiquitin ligase Parkin and mitophagy receptor PHB2 links inner mitochondrial membrane ubiquitination to efficient mitophagy. J Biol Chem. 2022;298:102704.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 10]  [Reference Citation Analysis (0)]
42.  Wang L, Cho YL, Tang Y, Wang J, Park JE, Wu Y, Wang C, Tong Y, Chawla R, Zhang J, Shi Y, Deng S, Lu G, Tan HW, Pawijit P, Lim GG, Chan HY, Fang L, Yu H, Liou YC, Karthik M, Bay BH, Lim KL, Sze SK, Yap CT, Shen HM. PTEN-L is a novel protein phosphatase for ubiquitin dephosphorylation to inhibit PINK1-Parkin-mediated mitophagy. Cell Res. 2018;28:787-802.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 82]  [Cited by in F6Publishing: 111]  [Article Influence: 18.5]  [Reference Citation Analysis (0)]
43.  Imberechts D, Kinnart I, Wauters F, Terbeek J, Manders L, Wierda K, Eggermont K, Madeiro RF, Sue C, Verfaillie C, Vandenberghe W. DJ-1 is an essential downstream mediator in PINK1/parkin-dependent mitophagy. Brain. 2022;145:4368-4384.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 31]  [Article Influence: 15.5]  [Reference Citation Analysis (0)]
44.  Shen JL, Fortier TM, Wang R, Baehrecke EH. Vps13D functions in a Pink1-dependent and Parkin-independent mitophagy pathway. J Cell Biol. 2021;220.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in F6Publishing: 22]  [Article Influence: 7.3]  [Reference Citation Analysis (0)]
45.  Lazarou M, Sliter DA, Kane LA, Sarraf SA, Wang C, Burman JL, Sideris DP, Fogel AI, Youle RJ. The ubiquitin kinase PINK1 recruits autophagy receptors to induce mitophagy. Nature. 2015;524:309-314.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1506]  [Cited by in F6Publishing: 1802]  [Article Influence: 200.2]  [Reference Citation Analysis (0)]
46.  Bhujabal Z, Birgisdottir ÅB, Sjøttem E, Brenne HB, Øvervatn A, Habisov S, Kirkin V, Lamark T, Johansen T. FKBP8 recruits LC3A to mediate Parkin-independent mitophagy. EMBO Rep. 2017;18:947-961.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 224]  [Cited by in F6Publishing: 278]  [Article Influence: 39.7]  [Reference Citation Analysis (0)]
47.  Mazure NM, Pouysségur J. Hypoxia-induced autophagy: cell death or cell survival? Curr Opin Cell Biol. 2010;22:177-180.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 421]  [Cited by in F6Publishing: 474]  [Article Influence: 31.6]  [Reference Citation Analysis (0)]
48.  Nguyen-Dien GT, Kozul KL, Cui Y, Townsend B, Kulkarni PG, Ooi SS, Marzio A, Carrodus N, Zuryn S, Pagano M, Parton RG, Lazarou M, Millard SS, Taylor RW, Collins BM, Jones MJ, Pagan JK. FBXL4 suppresses mitophagy by restricting the accumulation of NIX and BNIP3 mitophagy receptors. EMBO J. 2023;42:e112767.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in F6Publishing: 7]  [Article Influence: 7.0]  [Reference Citation Analysis (0)]
49.  Zhu Y, Massen S, Terenzio M, Lang V, Chen-Lindner S, Eils R, Novak I, Dikic I, Hamacher-Brady A, Brady NR. Modulation of serines 17 and 24 in the LC3-interacting region of Bnip3 determines pro-survival mitophagy versus apoptosis. J Biol Chem. 2013;288:1099-1113.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 291]  [Cited by in F6Publishing: 326]  [Article Influence: 27.2]  [Reference Citation Analysis (0)]
50.  Rogov VV, Suzuki H, Marinković M, Lang V, Kato R, Kawasaki M, Buljubašić M, Šprung M, Rogova N, Wakatsuki S, Hamacher-Brady A, Dötsch V, Dikic I, Brady NR, Novak I. Phosphorylation of the mitochondrial autophagy receptor Nix enhances its interaction with LC3 proteins. Sci Rep. 2017;7:1131.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 137]  [Cited by in F6Publishing: 190]  [Article Influence: 27.1]  [Reference Citation Analysis (0)]
51.  Marinković M, Šprung M, Novak I. Dimerization of mitophagy receptor BNIP3L/NIX is essential for recruitment of autophagic machinery. Autophagy. 2021;17:1232-1243.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 53]  [Cited by in F6Publishing: 115]  [Article Influence: 28.8]  [Reference Citation Analysis (0)]
52.  Liu L, Feng D, Chen G, Chen M, Zheng Q, Song P, Ma Q, Zhu C, Wang R, Qi W, Huang L, Xue P, Li B, Wang X, Jin H, Wang J, Yang F, Liu P, Zhu Y, Sui S, Chen Q. Mitochondrial outer-membrane protein FUNDC1 mediates hypoxia-induced mitophagy in mammalian cells. Nat Cell Biol. 2012;14:177-185.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 960]  [Cited by in F6Publishing: 1128]  [Article Influence: 94.0]  [Reference Citation Analysis (0)]
53.  Chen G, Han Z, Feng D, Chen Y, Chen L, Wu H, Huang L, Zhou C, Cai X, Fu C, Duan L, Wang X, Liu L, Liu X, Shen Y, Zhu Y, Chen Q. A regulatory signaling loop comprising the PGAM5 phosphatase and CK2 controls receptor-mediated mitophagy. Mol Cell. 2014;54:362-377.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 327]  [Cited by in F6Publishing: 398]  [Article Influence: 39.8]  [Reference Citation Analysis (0)]
54.  Wu H, Xue D, Chen G, Han Z, Huang L, Zhu C, Wang X, Jin H, Wang J, Zhu Y, Liu L, Chen Q. The BCL2L1 and PGAM5 axis defines hypoxia-induced receptor-mediated mitophagy. Autophagy. 2014;10:1712-1725.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 114]  [Cited by in F6Publishing: 132]  [Article Influence: 13.2]  [Reference Citation Analysis (0)]
55.  Wu H, Wei H, Zhang D, Sehgal SA, Wang X, Qin Y, Liu L, Chen Q. Defective mitochondrial ISCs biogenesis switches on IRP1 to fine tune selective mitophagy. Redox Biol. 2020;36:101661.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 12]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
56.  Murakawa T, Yamaguchi O, Hashimoto A, Hikoso S, Takeda T, Oka T, Yasui H, Ueda H, Akazawa Y, Nakayama H, Taneike M, Misaka T, Omiya S, Shah AM, Yamamoto A, Nishida K, Ohsumi Y, Okamoto K, Sakata Y, Otsu K. Bcl-2-like protein 13 is a mammalian Atg32 homologue that mediates mitophagy and mitochondrial fragmentation. Nat Commun. 2015;6:7527.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 284]  [Cited by in F6Publishing: 337]  [Article Influence: 37.4]  [Reference Citation Analysis (0)]
57.  Murakawa T, Okamoto K, Omiya S, Taneike M, Yamaguchi O, Otsu K. A Mammalian Mitophagy Receptor, Bcl2-L-13, Recruits the ULK1 Complex to Induce Mitophagy. Cell Rep. 2019;26:338-345.e6.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 47]  [Cited by in F6Publishing: 70]  [Article Influence: 17.5]  [Reference Citation Analysis (0)]
58.  Yoo SM, Yamashita SI, Kim H, Na D, Lee H, Kim SJ, Cho DH, Kanki T, Jung YK. FKBP8 LIRL-dependent mitochondrial fragmentation facilitates mitophagy under stress conditions. FASEB J. 2020;34:2944-2957.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 20]  [Cited by in F6Publishing: 34]  [Article Influence: 6.8]  [Reference Citation Analysis (0)]
59.  Cao M, Tang Y, Luo Y, Gu F, Zhu Y, Liu X, Yan C, Hu W, Wang S, Chao X, Xu H, Chen HB, Wang L. Natural compounds modulating mitophagy: Implications for cancer therapy. Cancer Lett. 2024;582:216590.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
60.  Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, Bray F. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J Clin. 2021;71:209-249.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 50630]  [Cited by in F6Publishing: 46677]  [Article Influence: 15559.0]  [Reference Citation Analysis (47)]
61.  Chen H, Wang X, Shao S, Zhang J, Tan X, Chen W. Value of EUS in determining infiltration depth of early carcinoma and associated precancerous lesions in the upper gastrointestinal tract. Endosc Ultrasound. 2022;11:503-510.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 2]  [Reference Citation Analysis (0)]
62.  Vanella G, Bronswijk M, Arcidiacono PG, Larghi A, Wanrooij RLJV, de Boer YS, Rimbas M, Khashab M, van der Merwe SW. Current landscape of therapeutic EUS: Changing paradigms in gastroenterology practice. Endosc Ultrasound. 2023;12:16-28.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 1]  [Reference Citation Analysis (0)]
63.  Yan H, Qiu C, Sun W, Gu M, Xiao F, Zou J, Zhang L. Yap regulates gastric cancer survival and migration via SIRT1/Mfn2/mitophagy. Oncol Rep. 2018;39:1671-1681.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 19]  [Cited by in F6Publishing: 50]  [Article Influence: 8.3]  [Reference Citation Analysis (0)]
64.  Wang X, Zhang L, Chan FKL, Ji J, Yu J, Liang JQ. Gamma-glutamyltransferase 7 suppresses gastric cancer by cooperating with RAB7 to induce mitophagy. Oncogene. 2022;41:3485-3497.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 1]  [Article Influence: 0.5]  [Reference Citation Analysis (0)]
65.  Yan H, Xiao F, Zou J, Qiu C, Sun W, Gu M, Zhang L. NR4A1-induced increase in the sensitivity of a human gastric cancer line to TNFα-mediated apoptosis is associated with the inhibition of JNK/Parkin-dependent mitophagy. Int J Oncol. 2018;52:367-378.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 17]  [Article Influence: 2.4]  [Reference Citation Analysis (0)]
66.  Xiao YY, Xiao JX, Wang XY, Wang T, Qu XH, Jiang LP, Tou FF, Chen ZP, Han XJ. Metformin-induced AMPK activation promotes cisplatin resistance through PINK1/Parkin dependent mitophagy in gastric cancer. Front Oncol. 2022;12:956190.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 6]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
67.  Wang R, Lee YG, Dhandapani S, Baek NI, Kim KP, Cho YE, Xu X, Kim YJ. 8-paradol from ginger exacerbates PINK1/Parkin mediated mitophagy to induce apoptosis in human gastric adenocarcinoma. Pharmacol Res. 2023;187:106610.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in F6Publishing: 3]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
68.  Gong Y, Tang N, Liu P, Sun Y, Lu S, Liu W, Tan L, Song C, Qiu X, Liao Y, Yu S, Liu X, Lin SH, Ding C. Newcastle disease virus degrades SIRT3 via PINK1-PRKN-dependent mitophagy to reprogram energy metabolism in infected cells. Autophagy. 2022;18:1503-1521.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 57]  [Article Influence: 19.0]  [Reference Citation Analysis (0)]
69.  Shin AE, Giancotti FG, Rustgi AK. Metastatic colorectal cancer: mechanisms and emerging therapeutics. Trends Pharmacol Sci. 2023;44:222-236.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 29]  [Reference Citation Analysis (0)]
70.  Yin K, Lee J, Liu Z, Kim H, Martin DR, Wu D, Liu M, Xue X. Mitophagy protein PINK1 suppresses colon tumor growth by metabolic reprogramming via p53 activation and reducing acetyl-CoA production. Cell Death Differ. 2021;28:2421-2435.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 21]  [Cited by in F6Publishing: 50]  [Article Influence: 16.7]  [Reference Citation Analysis (0)]
71.  Wang S, Jiang X, Xie X, Yin J, Zhang J, Liu T, Chen S, Wang Y, Zhou X, Cui R, Jiang H. piR-823 inhibits cell apoptosis via modulating mitophagy by binding to PINK1 in colorectal cancer. Cell Death Dis. 2022;13:465.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 12]  [Article Influence: 6.0]  [Reference Citation Analysis (0)]
72.  Chen B, Das NK, Talukder I, Singhal R, Castillo C, Andren A, Mancias JD, Lyssiotis CA, Shah YM. PTEN-induced kinase PINK1 supports colorectal cancer growth by regulating the labile iron pool. J Biol Chem. 2023;299:104691.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 4]  [Article Influence: 4.0]  [Reference Citation Analysis (0)]
73.  Li Q, Qi F, Meng X, Zhu C, Gao Y. Mst1 regulates colorectal cancer stress response via inhibiting Bnip3-related mitophagy by activation of JNK/p53 pathway. Cell Biol Toxicol. 2018;34:263-277.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 33]  [Cited by in F6Publishing: 42]  [Article Influence: 6.0]  [Reference Citation Analysis (0)]
74.  Tang J, Peng W, Ji J, Peng C, Wang T, Yang P, Gu J, Feng Y, Jin K, Wang X, Sun Y. GPR176 Promotes Cancer Progression by Interacting with G Protein GNAS to Restrain Cell Mitophagy in Colorectal Cancer. Adv Sci (Weinh). 2023;10:e2205627.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 3]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
75.  Ziegler PK, Bollrath J, Pallangyo CK, Matsutani T, Canli Ö, De Oliveira T, Diamanti MA, Müller N, Gamrekelashvili J, Putoczki T, Horst D, Mankan AK, Öner MG, Müller S, Müller-Höcker J, Kirchner T, Slotta-Huspenina J, Taketo MM, Reinheckel T, Dröse S, Larner AC, Wels WS, Ernst M, Greten TF, Arkan MC, Korn T, Wirth D, Greten FR. Mitophagy in Intestinal Epithelial Cells Triggers Adaptive Immunity during Tumorigenesis. Cell. 2018;174:88-101.e16.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 56]  [Cited by in F6Publishing: 84]  [Article Influence: 14.0]  [Reference Citation Analysis (0)]
76.  Yan C, Luo L, Guo CY, Goto S, Urata Y, Shao JH, Li TS. Doxorubicin-induced mitophagy contributes to drug resistance in cancer stem cells from HCT8 human colorectal cancer cells. Cancer Lett. 2017;388:34-42.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 79]  [Cited by in F6Publishing: 99]  [Article Influence: 12.4]  [Reference Citation Analysis (0)]
77.  Wei Y, Xiao G, Xu H, Sun X, Shi Y, Wang F, Kang J, Peng J, Zhou F. Radiation resistance of cancer cells caused by mitochondrial dysfunction depends on SIRT3-mediated mitophagy. FEBS J. 2023;290:3629-3645.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 7]  [Reference Citation Analysis (0)]
78.  Boyle KA, Van Wickle J, Hill RB, Marchese A, Kalyanaraman B, Dwinell MB. Mitochondria-targeted drugs stimulate mitophagy and abrogate colon cancer cell proliferation. J Biol Chem. 2018;293:14891-14904.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 69]  [Cited by in F6Publishing: 87]  [Article Influence: 14.5]  [Reference Citation Analysis (0)]
79.  Zhang K, Zhang D, Wang J, Wang Y, Hu J, Zhou Y, Zhou X, Nie S, Xie M. Aloe gel glucomannan induced colon cancer cell death via mitochondrial damage-driven PINK1/Parkin mitophagy pathway. Carbohydr Polym. 2022;295:119841.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
80.  D'Onofrio N, Martino E, Mele L, Colloca A, Maione M, Cautela D, Castaldo D, Balestrieri ML. Colorectal Cancer Apoptosis Induced by Dietary δ-Valerobetaine Involves PINK1/Parkin Dependent-Mitophagy and SIRT3. Int J Mol Sci. 2021;22.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 9]  [Cited by in F6Publishing: 16]  [Article Influence: 5.3]  [Reference Citation Analysis (0)]
81.  Liang L, Sun W, Wei X, Wang L, Ruan H, Zhang J, Li S, Zhao B, Li M, Cai Z, Huang J. Oxymatrine suppresses colorectal cancer progression by inhibiting NLRP3 inflammasome activation through mitophagy induction in vitro and in vivo. Phytother Res. 2023;37:3342-3362.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
82.  Guo W, Sun Y, Liu W, Wu X, Guo L, Cai P, Shen Y, Shu Y, Gu Y, Xu Q. Small molecule-driven mitophagy-mediated NLRP3 inflammasome inhibition is responsible for the prevention of colitis-associated cancer. Autophagy. 2014;10:972-985.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 178]  [Cited by in F6Publishing: 184]  [Article Influence: 20.4]  [Reference Citation Analysis (0)]
83.  Park J, Cheon JH. Incidence and Prevalence of Inflammatory Bowel Disease across Asia. Yonsei Med J. 2021;62:99-108.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 32]  [Cited by in F6Publishing: 64]  [Article Influence: 21.3]  [Reference Citation Analysis (0)]
84.  Nakahira K, Haspel JA, Rathinam VA, Lee SJ, Dolinay T, Lam HC, Englert JA, Rabinovitch M, Cernadas M, Kim HP, Fitzgerald KA, Ryter SW, Choi AM. Autophagy proteins regulate innate immune responses by inhibiting the release of mitochondrial DNA mediated by the NALP3 inflammasome. Nat Immunol. 2011;12:222-230.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1951]  [Cited by in F6Publishing: 2207]  [Article Influence: 157.6]  [Reference Citation Analysis (0)]
85.  Zhang Q, Raoof M, Chen Y, Sumi Y, Sursal T, Junger W, Brohi K, Itagaki K, Hauser CJ. Circulating mitochondrial DAMPs cause inflammatory responses to injury. Nature. 2010;464:104-107.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2365]  [Cited by in F6Publishing: 2559]  [Article Influence: 182.8]  [Reference Citation Analysis (0)]
86.  Sena LA, Chandel NS. Physiological roles of mitochondrial reactive oxygen species. Mol Cell. 2012;48:158-167.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1626]  [Cited by in F6Publishing: 1801]  [Article Influence: 163.7]  [Reference Citation Analysis (0)]
87.  Vincent G, Novak EA, Siow VS, Cunningham KE, Griffith BD, Comerford TE, Mentrup HL, Stolz DB, Loughran P, Ranganathan S, Mollen KP. Nix-Mediated Mitophagy Modulates Mitochondrial Damage During Intestinal Inflammation. Antioxid Redox Signal. 2020;33:1-19.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 19]  [Cited by in F6Publishing: 22]  [Article Influence: 5.5]  [Reference Citation Analysis (0)]
88.  Chen Y, Li J, Li S, Cheng Y, Fu X, Zhu L. Uncovering the Novel Role of NR1D1 in Regulating BNIP3-Mediated Mitophagy in Ulcerative Colitis. Int J Mol Sci. 2023;24.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
89.  Martín-Reyes F, Bernal M, Rodríguez-Díaz C, Rodríguez-de Los Reyes D, Ho-Plagaro A, Rodríguez-Pacheco F, Camacho-Martel L, Camargo-Camero R, Rodríguez-González FJ, Alcain-Martínez G, Martín-Masot R, Navas-López VM, Villanueva-Paz M, Lucena MI, García-Fuentes E, López-Gómez C. Mitochondrial Stress Links Environmental Triggers with Pro-Inflammatory Signaling in Crohn's Disease. Antioxidants (Basel). 2023;12.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
90.  Mishra SR, Mahapatra KK, Behera BP, Patra S, Bhol CS, Panigrahi DP, Praharaj PP, Singh A, Patil S, Dhiman R, Bhutia SK. Mitochondrial dysfunction as a driver of NLRP3 inflammasome activation and its modulation through mitophagy for potential therapeutics. Int J Biochem Cell Biol. 2021;136:106013.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 19]  [Cited by in F6Publishing: 57]  [Article Influence: 19.0]  [Reference Citation Analysis (0)]
91.  Zhen Y, Zhang H. NLRP3 Inflammasome and Inflammatory Bowel Disease. Front Immunol. 2019;10:276.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 290]  [Cited by in F6Publishing: 372]  [Article Influence: 74.4]  [Reference Citation Analysis (0)]
92.  Bian Z, Zhang Q, Qin Y, Sun X, Liu L, Liu H, Mao L, Yan Y, Liao W, Zha L, Sun S. Sodium Butyrate Inhibits Oxidative Stress and NF-κB/NLRP3 Activation in Dextran Sulfate Sodium Salt-Induced Colitis in Mice with Involvement of the Nrf2 Signaling Pathway and Mitophagy. Dig Dis Sci. 2023;68:2981-2996.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
93.  Luo T, Jia X, Feng WD, Wang JY, Xie F, Kong LD, Wang XJ, Lian R, Liu X, Chu YJ, Wang Y, Xu AL. Bergapten inhibits NLRP3 inflammasome activation and pyroptosis via promoting mitophagy. Acta Pharmacol Sin. 2023;44:1867-1878.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 3]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
94.  Liu C, Wang J, Yang Y, Liu X, Zhu Y, Zou J, Peng S, Le TH, Chen Y, Zhao S, He B, Mi Q, Zhang X, Du Q. Ginsenoside Rd ameliorates colitis by inducing p62-driven mitophagy-mediated NLRP3 inflammasome inactivation in mice. Biochem Pharmacol. 2018;155:366-379.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 48]  [Cited by in F6Publishing: 65]  [Article Influence: 10.8]  [Reference Citation Analysis (0)]
95.  Li P, Chen G, Zhang J, Pei C, Chen Y, Gong J, Deng S, Cai K, Li H, Wang D, Shen B, Xie Z, Liao Q. Live Lactobacillus acidophilus alleviates ulcerative colitis via the SCFAs/mitophagy/NLRP3 inflammasome axis. Food Funct. 2022;13:2985-2997.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 21]  [Article Influence: 10.5]  [Reference Citation Analysis (0)]
96.  Ma Z, Wu J, Wu Y, Sun X, Rao Z, Sun N, Fu Y, Zhang Z, Li J, Xiao M, Zeng Q, Han C, Ding D, Zhang H, Yuan H, Zhang J, Yang S, Chen Y. Parkin increases the risk of colitis by downregulation of VDR via autophagy-lysosome degradation. Int J Biol Sci. 2023;19:1633-1644.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
97.  Vakifahmetoglu-Norberg H, Ouchida AT, Norberg E. The role of mitochondria in metabolism and cell death. Biochem Biophys Res Commun. 2017;482:426-431.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 297]  [Cited by in F6Publishing: 270]  [Article Influence: 38.6]  [Reference Citation Analysis (0)]
98.  Putney JW Jr, Thomas AP. Calcium signaling: double duty for calcium at the mitochondrial uniporter. Curr Biol. 2006;16:R812-R815.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 27]  [Cited by in F6Publishing: 23]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
99.  Mohanty A, Tiwari-Pandey R, Pandey NR. Mitochondria: the indispensable players in innate immunity and guardians of the inflammatory response. J Cell Commun Signal. 2019;13:303-318.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 64]  [Cited by in F6Publishing: 89]  [Article Influence: 17.8]  [Reference Citation Analysis (0)]
100.  Picard M, Shirihai OS. Mitochondrial signal transduction. Cell Metab. 2022;34:1620-1653.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 130]  [Article Influence: 65.0]  [Reference Citation Analysis (0)]
101.  Liu L, Li Y, Chen G, Chen Q. Crosstalk between mitochondrial biogenesis and mitophagy to maintain mitochondrial homeostasis. J Biomed Sci. 2023;30:86.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 6]  [Reference Citation Analysis (0)]
102.  Panigrahi DP, Praharaj PP, Bhol CS, Mahapatra KK, Patra S, Behera BP, Mishra SR, Bhutia SK. The emerging, multifaceted role of mitophagy in cancer and cancer therapeutics. Semin Cancer Biol. 2020;66:45-58.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 72]  [Cited by in F6Publishing: 144]  [Article Influence: 28.8]  [Reference Citation Analysis (0)]
103.  Dong Y, Zhuang XX, Wang YT, Tan J, Feng D, Li M, Zhong Q, Song Z, Shen HM, Fang EF, Lu JH. Chemical mitophagy modulators: Drug development strategies and novel regulatory mechanisms. Pharmacol Res. 2023;194:106835.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]