Mitochondria ROS and Mitophagy in Acute Kidney Injury
Mitochondria ROS and Mitophagy in Acute Kidney Injury
Mitochondria ROS and Mitophagy in Acute Kidney Injury
Lianjiu Su, Jiahao Zhang, Hernando Gomez, John A Kellum & Zhiyong Peng
To cite this article: Lianjiu Su, Jiahao Zhang, Hernando Gomez, John A Kellum & Zhiyong Peng
(2023) Mitochondria ROS and mitophagy in acute kidney injury, Autophagy, 19:2, 401-414, DOI:
10.1080/15548627.2022.2084862
REVIEW
a
Department of Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan 430071, China; bBranch, Center for Cancer Research,
National Cancer Institute, National Institutes of HealthNeuro-Oncology, Bethesda, Maryland, USA; cCenter of Critical Care Nephrology, Department
of Critical Care Medicine, University of Pittsburgh Medical Center, Pittsburgh, USA
CONTACT Lianjiu Su [email protected] Department of Critical Care Medicine, Zhongnan Hospital of Wuhan University, 169 Donghu Rd, Wuchang
District, Wuhan 430071, Hubei province, China; Zhiyong Peng [email protected]
#
Contributed equally to this study
© 2022 The Author(s). Published by Informa UK Limited, trading as Taylor & Francis Group.
This is an Open Access article distributed under the terms of the Creative Commons Attribution-NonCommercial-NoDerivatives License (http://creativecommons.org/licenses/by-nc-nd/4.0/),
which permits non-commercial re-use, distribution, and reproduction in any medium, provided the original work is properly cited, and is not altered, transformed, or built upon in any way.
402 L. SU ET AL.
injury, kidney fibrosis and neurodegenerative disease [6–8]. by NOXs can also cause mitochondrial ROS production which is
However, the role mitophagy induction in AKI is not completely an important mechanistic pathways of ROS amplification or
understood. Here, we summarized the role of activation of propagation (summarized in Figure 1) [27].
mitophagy during different causes of kidney injury and the To protect cells from the oxidative stress, enzymatic and
mechanism of mitophagy activation in the kidney, discuss the non-enzymatic defense systems in mitochondria eliminate
excess ROS [28]. The non-enzymatic defense system includes
pharmacologic induction of mitophagy as a potential therapeutic
flavonoids, vitamins, and glutathione [29]. SOD (superoxide
strategy, and provide suggestions for future perspectives in this
dismutase), SOR (superoxide reductase), CAT (catalase), GPX
field. (glutathione peroxidase), GSR (glutathione-disulfide reduc
tase), PRDX (peroxiredoxin), and TXN (thioredoxin) consti
tute scavenging enzymes systems that are involved in the
The generation and elimination of mitochondria ROS
regulation of mitochondrial ROS [29]. SOD is in charge of
The main sources of cellular ROS are mitochondria and NOX converting the superoxide anion, the most dangerous ROS
(NADPH oxidase) [9]. Mitochondria utilize oxygen to gener produced [30], into hydrogen peroxide, which can be then
ate adenosine triphosphate (ATP) through oxidative phos converted to H2O by CAT [30].
phorylation (OXPHOS) which is one of sources of
mitochondrial ROS (mtROS) [10]. The electron transport
The relationship between ROS and mitophagy
chain (ETC) is a series of electron transporters embedded in
the inner mitochondrial membrane (IMM) that shuttle hydro Mitophagy is considered a bona fide strategy to limit mtROS
gen ions (H+) across the mitochondrial membrane from the production by removing the aged and damaged mitochondria
mitochondrial matrix into the intermembrane space to gen via the specific sequestration and engulfment of mitochondria
erate a potential ionic gradient that contains the energy neces in lysosome [31]. Mitophagy may function more broadly to
sary to synthesize ATP. Complex V allows H+ to enter the limit the deleterious effects of ROS on cellular function [32].
mitochondrial matrix in favor of a concentration gradient, ROS induces mtDNA damage, decreases the mitochondrial
releasing the necessary energy to couple phosphate into ade membrane potential, and induces oxidation of proteins and
nosine and synthesize ATP [11]. In this process, electrons are lipids [30]. Mitophagy after DNA damage is a vital cellular
transported through the ETC and are finally shuttled to response to maintain mitochondrial functions and DNA
molecular oxygen by complex IV [12]. Leak of electrons at repair. A previous study reported that suppression of mito
complexes I and III interact with oxygen to produce the phagy disturbs mitochondrial Ca2+ homeostasis, affects ATP
superoxide anion, the most important and dangerous production, and attenuates DNA repair [33]. Mitochondrial
mtROS [13,14]. It is now known that cells produce mtROS proteins and lipids also play important roles in maintaining
as important signaling molecules that participate in physiolo mitochondrial function. Specific mitochondrial lipids are cri
gic functions. However, abnormal increments in the produc tical for proper assembly of the electron transport chain
tion of mtROS are known to induce cell injury through complexes and for effective responses to mitophagy [34].
oxidative stress. Besides ATP production, mitochondria are Under stress conditions, cardiolipin (CL), which constitutes
also involved in heme and iron sulfur center biosynthesis almost 20% of the lipid content in the IMM, translocates to
which induce ROS production and play important roles in the outer mitochondrial membrane (OMM) where, together
oxidative damage [15,16]. with ceramide, it binds LC3 and LC3B-II to recruit phago
NOXs are central components for regulating the cellular phores to damaged organelles (discussed further in the next
redox balance [17]. There are seven isoforms of the NADPH section) [35]. Damage-induced ROS disrupt mitochondrial
oxidases have been identified: NOX1, CYBB/NOX2, NOX3, proteins and damage existing macromolecules. Furthermore,
NOX4, NOX5, DUOX1, and DUOX2 [18]. NADPH is derived these ROS oxidize ergosterol to ergosterol peroxide in the
from the metabolism of glucose through glycolysis wherein OMM [36], which acts as a signal to recruit VCP (valosin
G6PD (glucose-6-phosphate dehydrogenase) is the rate- containing protein) associated mitochondrial stress responsive
limiting enzyme [19]. Pyruvate, the end-product of glycolysis, 1, a component of a mitochondrial surveillance system [37],
is ultimately transported into mitochondria to undergo further to damaged mitochondria for the subsequent signal transduc
metabolism through the tricarboxylic acid (TCA) cycle generat tion to protein degradation by the proteasome which is
ing reduced equivalents in the form of NADH and FADH2 involved in maintaining mitochondrial protein homeostasis
which will then enter the electron transport chain in the last [35]. These data suggest that, oxidation of proteins and lipids
step of oxidative phosphorylation [20]. Complex I catalyzes the should have the presence of adaptive mitophagy for cell to
oxidation of NADPH/NADH generated in the TCA cycle survivor from oxidative damage.
[21,22]. Cytosolic and mitochondrial NADH are exchanged Enhanced mitophagy is usually an early response to pro
through the malate-aspartate (MA) shuttle and the G3P (gly mote survival while overwhelming or prolonged mitochon
cerol-3-phosphate) shuttle which has been reviewed in previous drial damage can induce excessive, pathological activation of
studies [23–25]. All NOX family members share six highly con mitophagy, thereby inducing cell death and tissue injury
served transmembrane domains. The cytoplasmic COOH ter [38]. While complex I inhibition stimulates the activation
minus contains conserved NADPH binding domains [26]. The of mitophagy through mtROS generation, subsequent cell
active enzyme complex transports electrons to oxygen from death is ultimately a consequence of mtROS that are mito
NADPH to produce superoxide free radical [26]. ROS produced phagy- dependent [39]. These data suggest that the process
AUTOPHAGY 403
of mitophagy may, in some cases, increase mtROS levels conditions focus on the PINK1-PRKN pathway [41].
which could trigger a cell to further induce mitophagy and However, there are PRKN-independent mechanisms that can
therefore propagate the elevation in mtROS levels through trigger the activation of mitophagy including those driving
a positive feedback loop [40]. ubiquitin ligases, receptor and CL.
Figure 1. The generation and elimination of mitochondria ROS. In the cytosol, NADPH is primarily produced by G6PD in the glycolysis pathway. The cytosolic and
mitochondrial NADPH is exchanged through two shuttles. NOXs transports electrons to oxygen from NADPH to produce superoxide free radical which was converted
by SOD (SOD2 in mitochondria) to hydrogen peroxide, and finally converted by catalase to harmless H2O. Mitochondrial ROS are produced from the leakage of
electrons to form superoxide at complex I and complex III in the electron transport chain. Mitochondria utilize oxygen to generate ATP through OXPHOS.
Abbreviations: ATP: adenosine triphosphate; CAT: catalase; ETC: electron transport chain; G6PD: glucose-6-phosphate dehydrogenase; MA/G3P: the malate-aspartate
(MA) and the glycerol-3-phosphate(G3P) shuttle; NOX: NADPH oxidase; OXPHOS: oxidative phosphorylation; SOD: superoxide dismutase; TCA: tricarboxylic acid.
404 L. SU ET AL.
targeting signal at the N terminus and also plays an important Cardiolipin-mediated mitophagy
role in this process [42]. Normally, PINK1 is constitutively CL is a hallmark mitochondrial lipid which is almost exclu
imported to the inner membrane via the TIMM (translocase sively found at the IMM [56]. CL can have different roles in
of inner mitochondrial membrane)-TOMM (translocase of mitochondrial quality control and morphology depending on
outer mitochondrial membrane) complex. PINK1 is then its location. When located in the IMM, CL cooperates with
cleaved by several proteases including PMP (peptidase, mito OPA1 (OPA1 mitochondrial dynamin like GTPase) to induce
chondrial processing) which removes PINK1’s N-terminal IMM fusion. In response to stress, the mitochondrial PLSCR3
mitochondrial targeting signal and the inner membrane PARL (phospholipid scramblase 3) allows the translocation of CL
(presenilin associated rhomboid like), cleaves PINK1 between from the inner to the outer leaflet of the membrane to bind
amino acids A103 and F104 in its hydrophobic domain span directly to LC3 to induce mitophagy [57]. During mitophagy,
ning the IMM, which ultimately results in proteasomal degra CL can also interact with BECN1 (beclin 1), a central regu
dation [43–46]. Mitochondrial membrane potential (ΔΨm) is lator of autophagy, and recruit the autophagic machinery by
a key indicator of mitochondrial health and injury. PINK1 is its interaction with LC3 [58].
imported as normal to the OMM of depolarized mitochondria,
however the loss of ΔΨm prevents its import and subsequent Ubiquitin-mediated mitophagy
cleavage in the IMM [47]. When mitochondrial import is dis Mitochondria ubiquitination plays a central role in mitophagy
rupted by depolarization, unprocessed PINK1 accumulates spe due to the E3 ligase activity of protein PRKN. Several ubiqui
cifically at the OMM of dysfunctional mitochondria. In tin ligases other than PRKN have been proved to have com
response to various stressors, PINK1 accumulates at OMM mon effect with PRKN. MUL1 (mitochondrial E3 ubiquitin
bound to the TOMM complex where it is activated through protein ligase 1) is a resident mitochondrial ubiquitin E3
dimerization and autophosphorylation [48]. PINK1 can stabi ligase inserted in the OMM and has a multifunction including
lize on the outer membrane to recruit PRKN from the cytosol to mitochondrial fusion, interplay between the endoplasmic reti
damaged mitochondria in response to decreased ΔΨm. PINK1 culum and mitochondria and mitophagy [59,60]. MUL1 acts
will phosphorylate PRKN on the Ub-like domain on the Ser in parallel with the PINK1-PRKN pathway in compensate for
resulting in an increase of its E3 activity (Ub ligase activity) and PINK1-PRKN loss in their mutant phenotypes [60]. MUL1
the formation of polyubiquitin chains on the surface of depo can act not only as a ubiquitin ligase but also as a mitophagy
larized mitochondrial membranes [49,50]. Activated PRKN receptor. It can directly recruit the autophagy machinery by
polyubiquitinates numerous substrates of OMM proteins, lead interaction of GABARAP (GABA type A receptor-associated
ing to the recruitment of the autophagy machinery including protein (Atg8-family protein) [61]. HUWE1 (HECT, UBA
the Ub- and LC3-binding receptor SQSTM1/p62 (sequesto and WWE domain containing E3 ubiquitin protein ligase 1)
some 1) (summarized in Figure 2) [51]. has been also identified play a role in AMBRA1-mediated
mitophagy in a PRKN-independent pathway (summarized in
Figure 3) [62].
PRKN-independent mitophagy
Mitophagy in acute kidney injury
Receptor-mediated mitophagy
The OMM exists of several LC3-interacting regions (LIR) con The kidney is only second to the heart in mitochondrial
taining autophagic receptors anchored in the membrane of the density and oxygen consumption. This is due to the high-
phagophore [52]. Unlike PINK1-PRKN mediated mitophagy energy demand necessary to reabsorb ~70% of the solute load
requiring the translocation of PRKN to the damaged mitochon filtered through the glomerulus and for excretion [63].
dria, some receptors, like BNIP3 (BCL2 interacting protein 3) Increased oxidative stress, inflammation, and uncoupling of
and FUNDC1 (FUN14 domain containing 1), can bind to LC3 oxygen consumption from ATP, all of which are associated
proteins, thereby linking the phagophore to the targeted mito with AKI, promote mitochondrial damage which can trigger
chondria, directly inducing mitophagy [52]. This process is mitophagy [63]. While the role of mitophagy in AKI has been
known as receptor-mediated mitophagy. A growing number controversial, many studies have now demonstrated the pro
of OMM proteins containing LIR domains have also been tective effect of mitophagy in AKI, including PRKN-
identified including BNIP3, its homolog BNIP3L/NIX (BCL2 independent pathways, such as BNIP3-mediated mitophagy
interacting protein 3 like), and FUNDC1, which have been [64] and PRKN-dependent pathways [65]. A few studies have
reviewed previously [41,53]. Proteins in the IMM can also act also reported a damage function of mitophagy in kidney
as crucial mitophagy receptors involved in targeting mitochon injury [66]. Some recent reviews have summarized mitochon
dria for autophagic degradation. PHB2 (prohibitin 2), an IMM drial biogenesis in kidney injury and repair including mito
protein, binds the phagophore membrane-associated protein phagy [67,68]. The role of mitophagy in various pathological
LC3 through a LIR domain which is required for PRKN- conditions of acute kidney injury is summarized in Table 1.
induced mitophagy in mammalian cells [54]. Some receptors, Abbreviations: AKI: acute kidney injury; BNIP3: BCL2 inter
like activating molecule in AMBRA1 (autophagy and beclin 1 acting protein 3; BUMPT cells: Boston University mouse proximal
regulator 1) which is a BCL2 homology domain 3 (BH3)-like tubule cells; CCCP: carbonyl cyanide m-chlorophenylhydrazone;
protein, can play a role in the selective degradation of ubiqui CKO: conditional knockout; CLP: cecal ligation and puncture;
tylated mitochondria, transducing both canonical PINK1- CNP: 2’,3’-cyclic nucleotide 3’phosphodiesterase; DNM1L: dyna
PRKN-dependent and -independent mitophagy [55]. min 1 like; FUNDC1: FUN14 domain containing 1; H/R: hypoxia/
AUTOPHAGY 405
Figure 2. PRKN-dependent mitophagy. PINK1 is constitutively processed by mitochondrial proteases, PMP and PARL, resulting in its proteasomal degradation in
normal condition (a); In damaged mitochondria, PINK1 accumulates at OMM bound to the TOMM complex where it is activated through auto-phosphorylation.
Activated PINK1 subsequently phosphorylates ubiquitin, which triggers recruitment of PRKN recruitment to mitochondria and activation of its E3 ligase activity. It
further ubiquitinates mitochondrial substrates and initiate autophagosome formation. PRKN acts as an enhancer of this signaling through further ubiquitination of
mitochondrial proteins (b). Abbreviations: IMM: inner mitochondrial membrane; OMM: outer mitochondrial membrane; PARL: presenilin associated rhomboid like;
PINK1: PTEN induced kinase 1; PMP: peptidase, mitochondrial processing; PRKN: parkin RBR E3 ubiquitin protein ligase; SQSTM1/p62: sequestosome 1; TIMM:
translocase of inner mitochondrial membrane; TOMM: translocase of outer mitochondrial membrane.
406 L. SU ET AL.
Figure 3. PRKN-independent mitophagy. Receptor mediated mitophagy: BNIP3, BNIP3L and FUNDC1, are OMM receptors containing an LIR-domain that directly
binds to LC3 proteins to recruit the phagophore to the damaged mitochondria and leading to its degradation; cardiolipin-mediated mitophagy: IMM CL can be
translocated to the OMM through the action of PLSCR3. Once at the OMM, CL binds to LC3A to recruit the phagophore and to remove the damaged mitochondria;
Ubiquitin mediated mitophagy: Some proteins have E3 ubiquitin ligases activities which can be located at damaged mitochondria to ubiquitinate OMM proteins, and
subsequently recruit the phagophore to damaged mitochondria. Abbreviations: BNIP3: BCL2 interacting protein 3; BNIP3L/NIX: BCL2 interacting protein 3 like; CL:
cardiolipin; FUNDC1: FUN14 domain containing 1; E3: enzyme 3; IMM: inner mitochondrial membranes; OMM: outer mitochondrial membranes; LIR: LC3-interacting
region; PLSCR3: phospholipid scramblase 3.
mitophagy resulting in the accumulation of damaged mito proximal tubular cells (RPTC) cell and mice induced IRI
chondria, increasing production of reactive oxygen species, model, which suggesting that mitophagy plays an important
cell death and inflammation in oxygen-glucose deprivation- role in the protective effect of IPC [72]. DNM1L/DRP1 (dyna
reperfusion (OGD-R) and IRI model [64]. Tang and his co- min 1 like) translocated to mitochondria and was phosphory
authors proved that PINK1-PRKN-mediated mitophagy plays lated at S616 in response to IRI [73]. Inhibiting DNM1L
an important role in mitochondrial quality control, tubular phosphorylation significantly suppressed without affecting
cell survival, and renal function in both in vitro and in vivo general autophagy suggesting that DNM1L was involved the
models of ischemic AKI [65]. Knockdown of pink1 sup process of mitochondrial fragmentation and downregulation
pressed mitophagy and reduced the cytoprotective effect of of mitophagy significantly aggravated kidney dysfunction
ischemic preconditioning (IPC) when treated carbonyl indicating that mitophagy was activated via DNM1L-
cyanide m-chlorophenylhydrazone (CCCP) in the rat dependent pathway to protect cells from IRI-induced
Table 1. The role of mitophagy in various pathological conditions of acute kidney injury.
Pathways Factors Model Cells and Animals Mechanisms Effect Reference
PRKN-dependent IRI CCCP for HK-2 cells and IRI for mice PINK1 siRNA, PRKN siRNA, or double siRNA in cells; pink1-KO, prkn-KO 1. Reduce mitochondrial damage Activation of mitophagy [65]
or double-KO in mice. 2. Modulate the removal of ROS protects against AKI
3. Relieve inflammatory response
4. Modulate cell death
IRI H/R for HK-2 cells and IRI for mice PmirGLO-Dual-luciferase reporter vector of MEG3 and RTKN in cells; Promoting apoptosis Activation of mitophagy [66]
AAV-sh-MEG3 vector in mice via tail vein. aggravates AKI
IRI CCCP for RPTC cells and IRI for mice Pink1 shRNA in cells 1.Suppressed mitochondrial Activation of mitophagy [72]
PT-atg7-KO in mice depolarization protects against AKI
2.Improved ATP production
3.Inhibited the generation of ROS
Sepsis LPS for RPTC cells and LPS or CLP for Pink1 siRNA, Prkn siRNA, or Optn siRNA in cells; pink1-KO or prkn KO 1. Mitochondrial quality control Activation of mitophagy [81]
mice in mice 2. Reduce cells apoptosis protects against AKI
Sepsis LPS for RPTC cells Pink1 siRNA and Prkn siRNA in cells 1. Inhibited the apoptosis Activation of mitophagy [82]
2. Remove damaged mitochondria protects against AKI
Sepsis LPS for HK-2 cells and CLP for rats PRKN siRNA or SIRT1 inhibitor in HK-2 cells; Prkn silencing lentivirus 1. Inhibited the apoptosis Activation of mitophagy [83]
and SIRT1 inhibitor EX527 in rats via tail vein 2. Inhibited the pyroptosis protects against AKI
3. Remove damaged mitochondria
Sepsis CLP for mice prkn-KO in mice 1. Inhibition of mitochondrial Activation of mitophagy [84]
dysfunction protects against AKI
2. Inhibition of NLRP3 inflammasome
activation
Cisplatin Cisplatin injected intraperitoneally pink1-KO or prkn-KO in mice Reduce cells apoptosis Activation of mitophagy [87]
for mice protects against AKI
Cisplatin Cisplatin for RTECs and cisplatin - 1. Reversed cellular ROS induced by Activation of mitophagy [88]
injected intraperitoneally for mice cisplatin protects against AKI
2. Reversed mitochondrial membrane
potential level induced by cisplatin
Cisplatin Cisplatin for HK-2 PINK1 siRNA, PRKN siRNA and PINK1 or PRKN overexpression plasmids 1. Protected against mitochondrial Activation of mitophagy [89]
in HK-2 dysfunction protects against AKI
2. Protected against cell injury
Cisplatin Intraperitoneal injection of cisplatin pink1-KO in rats 1. PINK1 deficiency inhibited DNM1L- Excessive mitophagy [90]
for rats mediated mitochondrial fission aggravates AKI
2. PINK1 deficiency inhibited excessive
mitophagy
Contrast Iohexol for HK-2 and iohexol PINK1 siRNA or PRKN siRNA in cells; pink1-KO or prkn-KO in mice 1. Reduce mitochondrial ROS Activation of mitophagy [91]
administration for mice 2. Reduce NLRP3 inflammasome protects against AKI
activation
Contrast Ioversol for HK-2 and ioversol for - 1. Reduce oxidative stress Activation of mitophagy [94]
rats 2. Reduce mitochondrial damage protects against AKI
PRKN-independent IRI OGD-R for BUMPT cells and IRI for Bnip3 silence in cells; bnip3-KO in mice 1. Eliminate damaged mitochondria Activation of mitophagy [64]
mice 2. Modulate the removal of ROS protects against AKI
3. Modulate cell death
4. Relieve inflammatory response
IRI IRI for rats Mdivi-1, an inhibitor of DNM1L, in rats 1. Mitophagic clearance of damaged Activation of mitophagy [73]
mitochondria protects against AKI
2. Protects cells apoptosis.
IRI IRI for mice cnp-KO in mice. Aggressive removal of injured Activation of mitophagy [74]
mitochondria protects against AKI
IRI H/R for HK-2 cells and IRI for mice HIF1A siRNA, BNIP3 siRNA, and BNIP3-overexpression plasmid for cell; 1. Inhibited apoptosis Activation of mitophagy [75]
hif1a-CKO and bnip3-overexpression adenovirus in mice 2. Inhibited ROS production protects against AKI
IRI IRI for mice and rotenone for fundc1-PTKO, dnm1l-PTKO, ulk1-PTKO and fundc1-dnm1l-PTKO in 1. Mitochondrial quality control Activation of mitophagy [76]
AUTOPHAGY
primary tubule cells mice 2. Reduce ROS oxidative stress protects against AKI
3. Reduce mitochondrial apoptosis
Contrast Iohexol for HK-2 and iohexol BNIP3 siRNA in cells; bnip3-KO in mice Reduce cells apoptosis Activation of mitophagy [93]
administration for mice protects against AKI
407
408 L. SU ET AL.
apoptosis [73]. Similar results were obtained with cnp 2’,3’- PRKN shown the contrary phenomenon suggesting mito
cyclic nucleotide 3’-phosphodiesterase) deletion mice which phagy plays a cytoprotective role against cisplatin injury
attenuates IRI-induced AKI, in part by accelerating mito [89]. However, Liu et al reported that pink1 deficiency
phagy with targeted removal of damaged mitochondria [74]. ameliorated cisplatin-induced AKI in rats, possibly via
Fu et al reported that BNIP3 overexpression reversed the inhibiting DNM1L-mediated mitochondrial fission and
inhibitory effect of HIF1A (hypoxia inducible factor 1 subunit excessive mitophagy [90].
alpha) knockout on mitophagy and prevented enhanced kid
ney damage in vivo and vitro [75]. Wang and his co-authors
demonstrated that FUNDC1-dependent mitophagy, primarily Mitophagy in response to contrast
driven by IPC, confers resistance to AKI through reconcilia
Iodinated intravascular contrast is widely used for vessel
tion of mitochondrial fission in a rotenone treated model
and chamber imaging in coronary angiography and per
in vitro and IRI model in vivo [76]. However, Zhou et al
cutaneous intervention. Despite advancements in imaging
reported that activation of mitophagy also aggravates kidney
and interventional techniques, contrast-induced acute kid
ischemia-reperfusion injury by trigger the WNT-CTNNB1
ney injury (CI-AKI) occurs in more than 30% of patients
/beta-catenin pathway in vitro and vivo [66].
after intravenous iodinated [91,92]. Mitophagy has been
shown to play an important role in CI-AKI. Lin et al
Mitophagy in response to sepsis reported that PINK1-PRKN-mediated mitophagy pre
vented apoptosis and tissue damage in CI-AKI in vitro
Sepsis is defined as organ dysfunction resulting from the
and vivo through reducing mitochondrial ROS and sub
host’s deleterious response to infection and one of the
sequent NLRP3 (NLR family, pyrin domain containing 3)
most common organs affected is the kidney [77]. Sepsis
inflammasome activation [91]. BNIP3-mediated mito
and inflammation at the tissue and cellular level are asso
phagy also has the protective function in CI-AKI [93].
ciated with decreased levels of intracellular ATP and with
Some drugs have the protective effect in CI-AKI by reg
mitochondrial injury in various organs, including the kid
ulating mitophagy. Bae et al reported that paricalcitol, an
ney [78]. Ultrastructural changes are observed in mito
active vitamin D analogue, could play a renoprotective
chondria during sepsis-induced AKI including decreased
role against CI-AKI through the PRKN-dependent path
mitochondrial mass, disruption of cristae, and extensive
way [94]. Tetramethylpyrazine can be protective against
mitochondrial swelling [79]. Sepsis also induces profound
CI-AKI because it can ameliorate renal oxidative stress
alterations in kidney tubular epithelial cell metabolism and
and aberrant mitochondrial dynamics, and modulate
in peritubular capillary flow distribution, all of which can
mitophagy in tubular cells in vitro and vivo [95]. Ward
result in significant energy imbalance and further mito
et al showed that the radiocontrast agent diatrizoic acid
chondrial injury [80]. Importantly, mitophagy is activated
could induce mitophagy and oxidative stress via calcium
early in the course of sepsis, providing the cell with
dysregulation, however, the role of the mitophagy in this
a mechanism to remove damaged mitochondria which is
process has not been further investigated [96].
important to minimize cell injury and accelerate recovery
[78]. Several studies demonstrated that PRKN-dependent
pathway of mitophagy was induced in in mouse models of
Mitophagy in the other drug-induced AKI
septic AKI induced by lipopolysaccharide (LPS) treatment
or by cecal ligation and puncture or in cultured proximal Many drugs can induce AKI, including some that are
tubular epithelial cells exposed to LPS, which plays an classically used as models of AKI such as folic acid (FA)
important role in mitochondrial quality control, tubular and oxalate. The role of mitophagy in the pathobiology of
cell survival, and renal function in septic AKI [81–84]. AKI from these drugs is poorly understood. However,
These protective effect of mitophagy in sepsis induced previous studies indicated that mitochondria play
AKI could be attributed to a decreasing inflamma a critical role in their cytotoxicity to kidney. Zhang et al
tion [85]. reported that inhibition of mitochondrial complex
I activity aggravated renal tubular injury, mitochondrial
damage and oxidative stress in FA-induced AKI [97]. This
Mitophagy in response to cisplatin
cytotoxicity effect of FA may due to trigger a mitophagy
Cisplatin is a widely used chemotherapeutic drug with dependent ROS increase leading to cell death [39].
notorious toxicity to the kidneys, which involves mito Mitophagy interfaces with mitochondrial biogenesis to
chondrial dysfunction and damage in renal tubular cells. regulate mitochondrial content and longevity [98].
Mitophagy induction and its cytoprotective role have been During the FA-induced kidney injury, the mitochondrial
demonstrated both in vitro and in vivo in models of biogenesis was suppressed and mitochondrial dysfunction
cisplatin-induced AKI [86–88]. Both pink1 and prkn is persistent to promote the progression of kidney injury
knockout mice showed more severe renal functional loss, [99]. Acute oxalosis displayed calcium oxalate crystals
tissue damage, and apoptosis during cisplatin treatment inside distal tubular epithelial cells caused AKI associated
[87]. In vitro, knockdown of PINK1-PRKN induced the with mitochondrial changes characteristic of mitochon
aggravation of mitochondrial function, leading to the drial permeability transition [100], which can finally lead
increase of cell injury, while the overexpression of PINK1- to mitophagy [101].
AUTOPHAGY 409
Figure 4. Crosstalk between mitophagy and other type of cell death. VDAC regulates mitochondria iron homeostasis through CISD1 which may affect the iron-
dependent cell death of ferroptosis. Translocator protein interacts with VDAC in PINK-PRKN-dependent mitophagy. VDAC gate the Ca2+ transport to control energy
production and metabolism by modulating enzyme activity of TCA which impacts the production of mitochondria ROS in the electron transport chain. GSK3B
increased VDAC phosphorylation to control MPT. SLC7A11/xCT regulates extracellular cystine and intracellular glutamate exchanging which influence the TCA and
GSH metabolism through glutamate and cystine, respectively. BCL2 family proteins regulate apoptosis through controlling the assembly of multimeric BAX-BAK1
channels. The interaction of BECN1-BCL2 inhibits autophagy and mitophagy but increase apoptosis. BCL2 family proteins, like BNIP3, can disrupt interaction between
the BECN1 and antiapoptotic BCL2 family. NLRP3 process pro-CASP1 to active CASP1, which cleaves pro-inflammatory IL1B to mature IL1B causing pyroptosis. NLRP3
binding of its LIR motif to LC3 induce mitophagy while CASP1 inhibits mitophagy to amplify mitochondrial damage. VDAC activates NLRP3 to induce pyroptosis.
Abbreviations: BAK1: BCL2 antagonist/killer 1; BAX: BCL2 associated X, apoptosis regulator; BCL2: BCL2 apoptosis regulator; BECN1: beclin 1; CASP1: caspase 1;
CISD1: CDGSH iron sulfur domain 1; GPX: glutathione peroxidase; GSH: glutathione; GSK3B: glycogen synthase kinase 3 beta; IL1B: interleukin 1 beta; LIR: LC3-
interacting region; MPT: mitochondrial permeability transition; NLRP3: NLR family pyrin domain containing 3; PINK1: PTEN induced kinase 1; PRKN: parkin RBR E3
ubiquitin protein ligase; SLC7A11/xCT: solute carrier family 7 member 11; TCA: tricarboxylic acid; VDAC: voltage dependent anion channel.
Interplay of mitophagy, apoptosis, pyroptosis and mechanism of interaction between mitophagy and various cell
ferroptosis in AKI death forms.
Mitophagy is mainly thought of as a survival mechanism by Both mitophagy and apoptosis are generally induced in
response to a common stimulus. The crosstalk between mito
removing damaged mitochondria and related ROS from these
phagy and apoptosis is mediated by several key molecules
damaged mitochondria. In addition to elimination of damaged
including members of the BCL2 (BCL2 apoptosis regulator)
mitochondria, mitophagy prevents cell death by regulating genes family and its interacting protein like BNIP3 and FUNDC1,
associated with programmed cell death [32], which is a general and other mitophagy-related proteins. BCL2 family proteins
410 L. SU ET AL.
regulate apoptosis by controlling the assembly of multimeric [120]. Cystine is a source of the synthesis of the glutathione
BAX (BCL2 associated X, apoptosis regulator)-BAK1 (BCL2 (GSH) which is a cofactor of the GPX4 [119]. Thus, on the
antagonist/killer 1) channels and thereby, modulating the one hand, erastin targets the SLC7A11 receptor on membrane
permeability of the OMM [102]. BCL2 regulates macroauto to influence the activity of GPX4 to induce ferroptosis. On the
phagy through binding to the autophagy regulator BECN1 by other hand, it can also activate VDAC receptor located at
interacting with its BH3-only domain and blocking its parti mitochondrial outer membrane to increase Ca2+ transport
cipation in the triggering of autophagosome formation. BCL2 which controls energy production and metabolism by mod
family proteins have also been proved to inhibit PINK1- ulating critical enzymes in the TCA cycle [121]. Besides,
PRKN-dependent mitophagy [103]. Some BCL2-interacting glutamate can transfer to α-Ketoglutarate through alanine
proteins like BNIP3 and FUNDC1 that disrupt interaction aminotransferase which is involved in TCA cycle [122].
between the BECN1 and antiapoptotic BCL2 family to pro Thus, mitochondria may play an important role in the cross
mote positive regulation of mitophagy [104,105]. In a renal talk between mitophagy and ferroptosis.
IRI model, BNIP3 in kidney cell induced light chain 3 expres Increasing evidence shows that mitochondrial dysfunction
sion and formation of autophagosomes which were mainly has an important role in ferroptosis [123–125]. GSK3B (gly
localized to the mitochondria, suggesting that mitophagy is cogen synthase kinase 3 beta) resides at the nexus of multiple
induced in renal tubules by BNIP3, which may be activated to signaling pathways implicated in the regulation of mitochon
protect the renal tubules during IRI-AKI [106]. BCL2 binding drial permeability transition (MPT) which finally caused
to BECN1 is known to inhibit autophagy [107]. Disruption of mitochondrial clearance though mitophagy [39,126]. GSK3B
BECN1-BCL2 interactions can enhance of autophagy activity increased VDAC phosphorylation, key MPT regulators
to protect kidney from ischemia-reperfusion injury [108]. located at OMM, and sensitized cells to MPT during drug-
BCL2 family proteins inhibits apoptosis and autophagy in induced oxidant stress kidney injury [127]. VDAC is an
kidney injury has been largely investigated [109–111]. important protein in the crosstalk between mitophagy and
BNIP3, inhibit mitophagy, and enhancing apoptosis and ferroptosis, and serves as a mitochondrial docking site to
ROS production during IRI [75]. recruit PRKN from the cytosol to defective mitochondria to
Pyroptosis is a pro-inflammatory form of regulated cell induce mitophagy [128]. The translocator protein interacts
death and is dependent on the enzymatic activity of inflam with VDAC1 which plays an important role in PINK1-
matory proteases that belong to the family of cysteine- PRKN-dependent mitophagy [129]. This procedure may be
dependent aspartate-specific proteases (caspases) [112]. enrolled during the kidney injury. VDAC was reported as the
Mitochondrial components are recognized as danger- only channel-forming protein and allowed the metabolites
associated molecular patterns by cytosolic pattern recognition and irons across the outer membrane, erastin-induced
receptors such as NOD-like receptors family member of VDAC opening mediated mitochondrial iron uptake may
NLRP3 inflammasomes [113]. They process pro-CASP1 (cas accelerate ferroptosis [130]. MitoNEET, also referred to as
pase 1) to active CASP1, which cleaves pro-inflammatory CISD1 (CDGSH iron sulfur domain 1), a redox-sensitive
IL1B (interleukin 1 beta) to mature IL1B causing inflamma (2Fe-2S) cluster protein, is an OMM protein essential for
tion and cell death by pyroptosis. NOD-like receptor (NLR) sensing and regulation of iron and ROS homeostasis [131].
family member with a mitochondrial targeting sequence, con CISD gates VDAC when oxidized in a redox-dependent man
tains a LIR and binding of its LIR motif to LC3 induce ner in cells, closing the pore and likely disrupting flow of iron
mitophagy [114,115]. Another study demonstrated that [132]. Mitochondrial TCA plays an important role in mito
NLRP3 activators induced mitochondrial damage, leading to phagy and ferroptosis. The flux of respiratory substrates,
their PRKN-dependent mitophagy [116]. Inhibition of NLRP3 ADP, and Pi into mitochondria and the release of mitochon
inflammasome activation was dependent on PRKN-mediated drial ATP to the cytosol occur through voltage-dependent
mitophagy in sepsis-induced acute injury [84]. CASP1 inhibits anion channels [39]. Thus, inhibition VDAC caused mito
mitophagy to amplify mitochondrial damage, mediated in chondrial TCA cycle inhibition which mitigate mitochondrial
part by cleavage of the key mitophagy regulator PRKN membrane potential hyperpolarization, lipid peroxide accu
[117]. Mitochondria dysfunction induced NLRP3 activation. mulation and triggers a mitophagy-dependent ROS increase
Both ROS generation and inflammasome activation are sup leading to ferroptosis (summarized in Figure 4) [39,133,134].
pressed when mitochondrial activity is dysregulated by inhi
bition of the VDAC (voltage dependent anion channel) [118].
Conclusion
These studies indicated that mitophagy removed damage
mitochondria caused by pyroptosis is a compensatory Mitophagy is a key cellular homeostatic mechanism that is
mechanism to the pyroptotic cell death through the interplay activated early during AKI. The effect of mitophagy and the
protein of NLRP3 and CASP1. relationship between mtROS levels and mitophagy are per
Ferroptosis is a form of cell death that results from the haps more complicated. During AKI, mitophagy is activated
catastrophic accumulation of lipid ROS caused by inactive early through PRKN-dependent and independent signaling
GPX4 and the accumulation of iron [119]. Erastin, a special pathways. The activation of mitophagy is protective in this
ferroptosis inducer, targets cystine/glutamate antiporter context, removing dysfunctional mitochondria from TEC and
SLC7A11/xCT (solute carrier family 7 member 11), to inhibit decreasing thereby local inflammation and oxidative damage.
extracellular cystine and intracellular glutamate exchange The crosstalk between mitophagy and forms of cell death such
AUTOPHAGY 411
as apoptosis and pyroptosis is an important contributor to [10] Shadel GS, Horvath TL. Mitochondrial ROS signaling in organis
some forms of AKI, and emerging evidence suggests an mal homeostasis. Cell. 2015;163(3):560–569.
[11] Cadonic C, Sabbir MG, Albensi BC. Mechanisms of
important link between iron metabolism, mitophagy and
Mitochondrial dysfunction in Alzheimer’s disease. Mol
a novel form of cell death known as ferroptosis. Further Neurobiol. 2016;53(9):6078–6090.
study could explore and validate the interplay of mitophagy [12] Nolfi-Donegan D, Braganza A, Shiva S. Mitochondrial electron
and ferroptosis. transport chain: oxidative phosphorylation, oxidant production,
and methods of measurement. Redox Biol. 2020;37:101674.
[13] Sumegi K, Fekete K, Antus C, et al. BGP-15 Protects against
Acknowledgments oxidative stress- or lipopolysaccharide-induced Mitochondrial
destabilization and reduces mitochondrial production of reactive
Not applicable. oxygen species. PLoS One. 2017;12(1):e0169372. :https://doi.org/
10.1371/journal.pone.0169372
[14] Iwasaki Y, Takeshima Y, Fujio K. Basic mechanism of immune system
Disclosure statement activation by mitochondria. Immunol Med. 2020;43(4):142–147.
[15] Nogueira NP, Saraiva FMS, Oliveira MP, et al. Heme modulates
None of the authors declared any conflict of interest in this work. trypanosoma cruzi bioenergetics inducing mitochondrial ROS
production. Free Radic Biol Med. 2017;108:183–191.
[16] Dong D, Hao Q, Zhang P, et al. Endoplasmic reticulum Ca2+
Data availability statement release causes rieske iron-sulfur protein-mediated mitochondrial
ROS generation in pulmonary artery smooth muscle cells. Biosci
The data supporting the findings of this study are available within the Rep. 2019;39(12). https://doi.org/10.1042/BSR20192414
article [and/or] its supplementary materials. [17] Rudolf J, Raad H, Taieb A, et al. NADPH oxidases and their roles
in skin homeostasis and carcinogenesis. Antioxid Redox Signal.
2018;28(13):1238–1261. https://doi.org/10.1089/ars.2017.7282
Funding [18] Hahner F, Moll F, Schroder K. NADPH oxidases in the differentia
tion of endothelial cells. Cardiovasc Res. 2020;116(2):262–268.
This work was supported by the National Natural Science Foundation of [19] Stanton RC. Glucose-6-phosphate dehydrogenase, NADPH, and
China [82102298]; National Natural Science Foundation of China cell survival. IUBMB Life. 2012;64(5):362–369.
[81772046]; National Natural Science Foundation of China [81971816]; [20] Gray LR, Tompkins SC, Taylor EB. Regulation of pyruvate metabo
Innovation Cultivation Foundation of Wuhan University/Zhongnan lism and human disease. Cell Mol Life Sci. 2014;71(14):2577–2604.
Hospital [413000345/CXPY2020017]. [21] SP A, AJ M, J R. Mammalian NADH:ubiquinone oxidoreductase
(complex I) and nicotinamide nucleotide transhydrogenase (Nnt)
together regulate the mitochondrial production of H(2)O(2)–
ORCID implications for their role in disease,especially cancer. J Bioenerg
Biomembr. 2011;43(5):541–564.
Zhiyong Peng http://orcid.org/0000-0002-3873-9607 [22] Agledal L, Niere M, Ziegler M. The phosphate makes a difference:
cellular functions of NADP. Redox Rep. 2010;15(1):2–10.
[23] Glancy B, Kane DA, Kavazis AN, et al. Mitochondrial lactate
References metabolism: history and implications for exercise and disease.
J Physiol. 2021;599(3):863–888. https://doi.org/10.1113/JP278930
[1] Yang X, Zhang R, Nakahira K, et al. Mitochondrial DNA muta [24] McKenna MC, Waagepetersen HS, Schousboe A, et al. Neuronal
tion, diseases, and nutrient-regulated mitophagy. Annu Rev Nutr. and astrocytic shuttle mechanisms for cytosolic-mitochondrial
2019;39(1):201–226. https://doi.org/10.1146/annurev-nutr transfer of reducing equivalents: current evidence and pharmaco
-082018-124643 logical tools. Biochem Pharmacol. 2006;71(4):399–407. https://doi.
[2] VanderVeen BN, Fix DK, Carson JA. Disrupted skeletal muscle org/10.1016/j.bcp.2005.10.011
Mitochondrial dynamics, Mitophagy, and biogenesis during can [25] Bakker BM, Overkamp KM, van Maris AJ, et al. Stoichiometry
cer cachexia: a role for inflammation. Oxid Med Cell Longev. and compartmentation of NADH metabolism in Saccharomyces
2017;2017:3292087. 2017. https://doi.org/10.1155/2017/3292087
cerevisiae. FEMS Microbiol Rev. 2001;25(1):15–37. https://doi.
[3] Xiong H, Chen S, Lai L, et al. Modulation of miR-34a/SIRT1
org/10.1111/j.1574-6976.2001.tb00570.x
signaling protects cochlear hair cells against oxidative stress and
[26] Bedard K, Krause KH. The NOX family of ROS-generating
delays age-related hearing loss through coordinated regulation of
NADPH oxidases: physiology and pathophysiology. Physiol Rev.
mitophagy and mitochondrial biogenesis. Neurobiol Aging.
2007;87(1):245–313.
2019;79:30–42.
[27] Zhang Y, Murugesan P, Huang K, et al. NADPH oxidases and
[4] Kaushal GP, Shah SV. Autophagy in acute kidney injury. Kidney
oxidase crosstalk in cardiovascular diseases: novel therapeutic
Int. 2016;89(4):779–791.
targets. Nat Rev Cardiol. 2020;17(3):170–194. https://doi.org/10.
[5] Song SB, Jang SY, Kang HT, et al. Modulation of Mitochondrial
1038/s41569-019-0260-8
membrane potential and ROS generation by nicotinamide in
[28] Czarna M, Jarmuszkiewicz W. Role of mitochondria in reactive
a manner independent of SIRT1 and Mitophagy. Mol Cells.
oxygen species generation and removal; relevance to signaling and
2017;40(7):503–514. https://doi.org/10.14348/molcells.2017.0081
programmed cell death. Postepy Biochem. 2006;52(2):145–156.
[6] Choi ME. Autophagy in kidney disease. Annu Rev Physiol.
[29] Ismail T, Kim Y, Lee H, et al. Interplay between Mitochondrial
2019;82:297–322.
peroxiredoxins and ROS in cancer development and progression.
[7] Fivenson EM, Lautrup S, Sun N, et al. Mitophagy in neurodegen
eration and aging. Neurochem Int. 2017;109:202–209. Int J Mol Sci. 2019;20(18):4407. https://doi.org/10.3390/
[8] Gong G, Song M, Csordas G, et al. Parkin-mediated mitophagy ijms20184407
directs perinatal cardiac metabolic maturation in mice. Science. [30] Suski JM, Lebiedzinska M, Bonora M, et al. Relation between
2015;350(6265):aad2459. https://doi.org/10.1126/science.aad2459 mitochondrial membrane potential and ROS formation.
[9] Cristofano MD, F A, Giacomo MD, et al. Mechanisms underlying Methods Mol Biol. 2012;810:183–205.
the hormetic effect of conjugated linoleic acid: focus on Nrf2, [31] Evans CS, Holzbaur EL. Degradation of engulfed mitochondria is
mitochondria and NADPH oxidases. Free Radic Biol Med. rate-limiting in optineurin-mediated mitophagy in neurons. Elife.
2021;167:276–286. 2020;9. https://doi.org/10.7554/eLife.50260
412 L. SU ET AL.
[32] Ma K, Chen G, Li W, et al. Mitophagy, Mitochondrial home [52] Liu L, Sakakibara K, Chen Q, et al. Receptor-mediated mitophagy
ostasis, and cell fate. Front Cell Dev Biol. 2020;8:467. in yeast and mammalian systems. Cell Res. 2014;24(7):787–795.
[33] Dan X, Babbar M, Moore A, et al. DNA damage invokes mito https://doi.org/10.1038/cr.2014.75
phagy through a pathway involving Spata18. Nucleic Acids Res. [53] Chu CT. Multiple pathways for mitophagy: a neurodegenerative
2020;48(12):6611–6623. https://doi.org/10.1093/nar/gkaa393 conundrum for Parkinson’s disease. Neurosci Lett.
[34] Nielson JR, Rutter JP. Lipid-mediated signals that regulate mito 2019;697:66–71.
chondrial biology. J Biol Chem. 2018;293(20):7517–7521. [54] Wei Y, Chiang WC, Sumpter R Jr., et al. Prohibitin 2 is an inner
[35] Chu CT, Ji J, Dagda RK, et al. Cardiolipin externalization to the Mitochondrial membrane mitophagy receptor. Cell. 2017;168(1–
outer mitochondrial membrane acts as an elimination signal for 2):224–238 e210. https://doi.org/10.1016/j.cell.2016.11.042
mitophagy in neuronal cells. Nat Cell Biol. 2013;15 [55] Strappazzon F, Nazio F, Corrado M, et al. AMBRA1 is able to
(10):1197–1205. https://doi.org/10.1038/ncb2837 induce mitophagy via LC3 binding, regardless of PARKIN and
[36] Bocking T, Barrow KD, Netting AG, et al. Effects of singlet p62/SQSTM1. Cell Death Differ. 2015;22(3):419–432. https://doi.
oxygen on membrane sterols in the yeast Saccharomyces org/10.1038/cdd.2014.139
cerevisiae. Eur J Biochem. 2000;267(6):1607–1618. https://doi. [56] Pangborn MC. A simplified preparation of cardiolipin, with note
org/10.1046/j.1432-1327.2000.01179.x on purification of lecithin for serologic use. J Biol Chem. 1945;161
[37] Heo JM, Nielson JR, Dephoure N, et al. Intramolecular interactions (1):71–82.
control Vms1 translocation to damaged mitochondria. Mol Biol Cell. [57] Dudek J. Role of cardiolipin in mitochondrial signaling pathways.
2013;24(9):1263–1273. https://doi.org/10.1091/mbc.e13-02-0072 Front Cell Dev Biol. 2017;5:90.
[38] Aggarwal S, Mannam P, Zhang J. Differential regulation of auto [58] Huang W, Choi W, Hu W, et al. Crystal structure and bio
phagy and mitophagy in pulmonary diseases. Am J Physiol Lung chemical analyses reveal Beclin 1 as a novel membrane binding
Cell Mol Physiol. 2016;311(2):L433–452. protein. Cell Res. 2012;22(3):473–489. https://doi.org/10.1038/
[39] Basit F, van Oppen LM, Schockel L, et al. Mitochondrial complex cr.2012.24
I inhibition triggers a mitophagy-dependent ROS increase leading [59] Puri R, Cheng XT, Lin MY, et al. Mul1 restrains Parkin-mediated
to necroptosis and ferroptosis in melanoma cells. Cell Death Dis. mitophagy in mature neurons by maintaining ER-mitochondrial
2017;8(3):e2716. https://doi.org/10.1038/cddis.2017.133 contacts. Nat Commun. 2019;10(1):3645. https://doi.org/10.1038/
[40] Schofield JH, Schafer ZT. Mitochondrial reactive oxygen species s41467-019-11636-5
and mitophagy: a complex and nuanced relationship. Antioxid [60] Yun J, Puri R, Yang H, et al. MUL1 acts in parallel to the PINK1/
Redox Signal. 2021;34(7):517–530. parkin pathway in regulating mitofusin and compensates for loss
[41] Villa E, Marchetti S, Ricci JE. No parkin zone: mitophagy without of PINK1/parkin. Elife. 2014;3:e01958.
Parkin. Trends Cell Biol. 2018;28(11):882–895. [61] Ambivero CT, Cilenti L, Main S, et al. Mulan E3 ubiquitin ligase
[42] Valente EM, Abou-Sleiman PM, Caputo V, et al. Hereditary early- interacts with multiple E2 conjugating enzymes and participates
onset Parkinson’s disease caused by mutations in PINK1. Science. in mitophagy by recruiting GABARAP. Cell Signal. 2014;26
2004;304(5674):1158–1160. https://doi.org/10.1126/science. (12):2921–2929. https://doi.org/10.1016/j.cellsig.2014.09.004
1096284 [62] Di Rita A, Peschiaroli A, DA P, et al. HUWE1 E3 ligase promotes
[43] Takatori S, Ito G, Iwatsubo T. Cytoplasmic localization and pro PINK1/PARKIN-independent mitophagy by regulating AMBRA1
teasomal degradation of N-terminally cleaved form of PINK1. activation via IKKalpha. Nat Commun. 2018;9(1):3755. https://
Neurosci Lett. 2008;430(1):13–17. doi.org/10.1038/s41467-018-05722-3
[44] Zhang C, Wang R, Liu Z, et al. The plant triterpenoid celastrol [63] Duann P, Lin PH. Mitochondria damage and kidney disease. Adv
blocks PINK1-dependent mitophagy by disrupting PINK1’s asso Exp Med Biol. 2017;982:529–551.
ciation with the mitochondrial protein TOM20. J Biol Chem. [64] Tang C, Han H, Liu Z, et al. Activation of BNIP3-mediated
2019;294(18):7472–7487. https://doi.org/10.1074/jbc.RA118. mitophagy protects against renal ischemia-reperfusion injury.
006506 Cell Death Dis. 2019;10(9):677. https://doi.org/10.1038/s41419-
[45] Jin SM, Lazarou M, Wang C, et al. Mitochondrial membrane 019-1899-0
potential regulates PINK1 import and proteolytic destabilization [65] Tang C, Han H, Yan M, et al. PINK1-PRKN/PARK2 pathway of
by PARL. J Cell Biol. 2010;191(5):933–942. https://doi.org/10. mitophagy is activated to protect against renal
1083/jcb.201008084 ischemia-reperfusion injury. Autophagy. 2018;14(5):880–897.
[46] Deas E, Plun-Favreau H, Gandhi S, et al. PINK1 cleavage at https://doi.org/10.1080/15548627.2017.1405880
position A103 by the mitochondrial protease PARL. Hum Mol [66] Liu D, Liu Y, Zheng X, et al. c-MYC-induced long noncoding
Genet. 2011;20(5):867–879. https://doi.org/10.1093/hmg/ddq526 RNA MEG3 aggravates kidney ischemia-reperfusion injury
[47] Lazarou M, Jin SM, Kane LA, et al. Role of PINK1 binding to the through activating mitophagy by upregulation of RTKN to trigger
TOM complex and alternate intracellular membranes in recruit the Wnt/beta-catenin pathway. Cell Death Dis. 2021;12(2):191.
ment and activation of the E3 ligase Parkin. Dev Cell. 2012;22 https://doi.org/10.1038/s41419-021-03466-5
(2):320–333. https://doi.org/10.1016/j.devcel.2011.12.014 [67] Tang C, Cai J, Yin XM, et al. Mitochondrial quality control in
[48] Okatsu K, Uno M, Koyano F, et al. A dimeric PINK1-containing kidney injury and repair. Nat Rev Nephrol. 2021;17(5):299–318.
complex on depolarized mitochondria stimulates Parkin https://doi.org/10.1038/s41581-020-00369-0
recruitment. J Biol Chem. 2013;288(51):36372–36384. https://doi. [68] Tang C, Livingston MJ, Liu Z, et al. Autophagy in kidney home
org/10.1074/jbc.M113.509653 ostasis and disease. Nat Rev Nephrol. 2020;16(9):489–508. https://
[49] Narendra D, Tanaka A, Suen DF, et al. Parkin is recruited selec doi.org/10.1038/s41581-020-0309-2
tively to impaired mitochondria and promotes their autophagy. [69] de Braganca Ac, Volpini RA, Mehrotra P, et al. Vitamin
J Cell Biol. 2008;183(5):795–803. https://doi.org/10.1083/jcb. D deficiency contributes to vascular damage in sustained ischemic
200809125 acute kidney injury. Physiol Rep. 2016;4(13). https://doi.org/10.
[50] Vives-Bauza C, Zhou C, Huang Y, et al. PINK1-dependent 14814/phy2.12829
recruitment of Parkin to mitochondria in mitophagy. Proc Natl [70] Hall AM, Rhodes GJ, Sandoval RM, et al. In vivo multiphoton
Acad Sci U S A. 2010;107(1):378–383. https://doi.org/10.1073/ imaging of mitochondrial structure and function during acute
pnas.0911187107 kidney injury. Kidney Int. 2013;83(1):72–83. https://doi.org/10.
[51] Shiba-Fukushima K, Inoshita T, Hattori N, et al. PINK1-mediated 1038/ki.2012.328
phosphorylation of Parkin boosts Parkin activity in Drosophila. [71] Lampert MA, Orogo AM, Najor RH, et al. BNIP3L/NIX and
PLoS Genet. 2014;10(6):e1004391. https://doi.org/10.1371/journal. FUNDC1-mediated mitophagy is required for mitochondrial net
pgen.1004391 work remodeling during cardiac progenitor cell differentiation.
AUTOPHAGY 413
[111] Isaka Y, Suzuki C, Abe T, et al. Bcl-2 protects tubular epithelial [124] Abdalkader M, Lampinen R, Kanninen KM, et al. Targeting Nrf2
cells from ischemia/reperfusion injury by dual mechanisms. to suppress ferroptosis and Mitochondrial dysfunction in
Transplant Proc. 2009;41(1):52–54. https://doi.org/10.1016/j.trans neurodegeneration. Front Neurosci. 2018;12:466.
proceed.2008.https://doi.org/10.026 [125] Battaglia AM, Chirillo R, Aversa I, et al. Ferroptosis and cancer:
[112] Xu YJ, Zheng L, Hu YW, et al. Pyroptosis and its relationship to Mitochondria meet the “Iron Maiden” cell death. Cells. 2020;9
atherosclerosis. Clin Chim Acta. 2018;476:28–37. (6):1505. https://doi.org/10.3390/cells9061505
[113] Singh LP, Devi TS, Yumnamcha T. The role of txnip in mitophagy [126] Tanaka T, Saotome M, Katoh H, et al. Glycogen synthase
dysregulation and inflammasome activation in diabetic retinopa kinase-3beta opens mitochondrial permeability transition pore
thy: a new perspective. JOJ Ophthalmol. 2017;4(4). https://doi. through mitochondrial hexokinase II dissociation. J Physiol Sci.
org/10.19080/JOJO.2017.04.555643 2018;68(6):865–871. https://doi.org/10.1007/s12576-018-0611-y
[114] Zhang Y, Yao Y, Qiu X, et al. Listeria hijacks host mitophagy through [127] Wang Z, Ge Y, Bao H, et al. Redox-sensitive glycogen synthase
a novel mitophagy receptor to evade killing. Nat Immunol. 2019;20 kinase 3beta-directed control of mitochondrial permeability tran
(4):433–446. https://doi.org/10.1038/s41590-019-0324-2 sition: rheostatic regulation of acute kidney injury. Free Radic Biol
[115] Zhen Y, Zhang H. NLRP3 inflammasome and inflammatory Med. 2013;65:849–858.
bowel disease. Front Immunol. 2019;10:276. [128] Sun Y, Vashisht AA, Tchieu J, et al. Voltage-dependent anion
[116] Zhong Z, Umemura A, Sanchez-Lopez E, et al. NF-kappaB channels (VDACs) recruit Parkin to defective mitochondria to
restricts inflammasome activation via elimination of damaged promote mitochondrial autophagy. J Biol Chem. 2012;287
mitochondria. Cell. 2016;164(5):896–910. https://doi.org/10.1016/ (48):40652–40660. https://doi.org/10.1074/jbc.M112.419721
j.cell.2015.12.057 [129] Moras M, Hattab C, Gonzalez-Menendez P, et al. Downregulation
[117] Yu J, Nagasu H, Murakami T, et al. Inflammasome activation of Mitochondrial tspo inhibits mitophagy and reduces enucleation
leads to Caspase-1-dependent mitochondrial damage and block during human terminal erythropoiesis. Int J Mol Sci. 2020;21
of mitophagy. Proc Natl Acad Sci U S A. 2014;111 (23):9066. https://doi.org/10.3390/ijms21239066
(43):15514–15519. https://doi.org/10.1073/pnas.1414859111 [130] Wang H, Liu C, Zhao Y, et al. Mitochondria regulation in
[118] Zhou R, Yazdi AS, Menu P, et al. A role for mitochondria in ferroptosis. Eur J Cell Biol. 2020;99(1):151058. https://doi.org/10.
NLRP3 inflammasome activation. Nature. 2011;469 1016/j.ejcb.2019.151058
(7329):221–225. https://doi.org/10.1038/nature09663 [131] Geldenhuys WJ, TC L, Carroll RT. Carroll RT. mitoNEET as
[119] Su LJ, Zhang JH, Gomez H, et al. Reactive oxygen species-induced a novel drug target for mitochondrial dysfunction. Drug Discov
lipid peroxidation in apoptosis, autophagy, and ferroptosis. Oxid Today. 2014;19(10):1601–1606.
Med Cell Longev. 2019;2019:5080843. [132] Lipper CH, Stofleth JT, Bai F, et al. Redox-dependent gating of
[120] Xie Y, Hou W, Song X, et al. Ferroptosis: process and function. Cell VDAC by mitoNEET. Proc Natl Acad Sci U S A. 2019;116
Death Differ. 2016;23(3):369–379. https://doi.org/10.1038/cdd.2015.158 (40):19924–19929. https://doi.org/10.1073/pnas.1908271116
[121] V S-B, De S MVDAC. the Na(+)/Ca(2+) exchanger, and the Ca(2 [133] McElroy GS, Reczek CR, Reyfman PA, et al. NAD+ regenera
+) uniporter in ca(2+) dynamics and signaling. Adv Exp Med tion rescues lifespan, but not ataxia, in a mouse model of
Biol. 2017;981:323–347. brain Mitochondrial complex i dysfunction. Cell Metab.
[122] Quan J, Fitch MD, Fleming SE. Rate at which glutamine enters 2020;32(2):301–308 e306. https://doi.org/10.1016/j.cmet.2020.
TCA cycle influences carbon atom fate in intestinal epithelial cells. 06.003
Am J Physiol. 1998;275(6):G1299–1308. [134] Gao M, Yi J, Zhu J, et al. Role of Mitochondria in ferroptosis.
[123] Neitemeier S, Jelinek A, Laino V, et al. Bid links ferroptosis to Mol Cell. 2019;73(2):354–363 e353. https://doi.org/10.1016/j.
mitochondrial cell death pathways. Redox Biol. 2017;12:558–570. molcel.2018