You are on page 1of 10

Seminars in Cell and Developmental Biology xxx (xxxx) xxx–xxx

Contents lists available at ScienceDirect

Seminars in Cell & Developmental Biology


journal homepage: www.elsevier.com/locate/semcdb

Review

Autophagy and mitophagy in cancer metabolic remodelling


Fabio Ferroa, Stéphane Servaisa, Pierre Bessona, Sébastien Rogerb, Jean-François Dumasa,
Lucie Brissona,*
a
Université de Tours, Inserm, UMR1069 Nutrition, Croissance et Cancer, Tours, France
b
Université de Tours, EA4245 Transplantation, Immunologie et Inflammation, Tours, France

ARTICLE INFO ABSTRACT

Keywords: Metabolic reprogramming in tumours is now recognized as a hallmark of cancer, participating both in tumour
Cancer metabolism growth and cancer progression. Cancer cells develop global metabolic adaptations allowing them to survive in
Autophagy the low oxygen and nutrient tumour microenvironment. Among these metabolic adaptations, cancer cells use
Mitophagy glycolysis but also mitochondrial oxidations to produce ATP and building blocks needed for their high pro-
ROS
liferation rate. Another particular adaptation of cancer cell metabolism is the use of autophagy and specific
Mitochondria
Ion channel
forms of autophagy like mitophagy to recycle intracellular components in condition of metabolic stress or during
anticancer treatments. The plasticity of cancer cell metabolism is a major limitation of anticancer treatments and
could participate to therapy resistances. The aim of this review is to report recent advances in the understanding
of the relationship between tumour metabolism and autophagy/mitophagy in order to propose new therapeutic
strategies.

1. Introduction would be to target related pathways in order to limit the acquisition of


resistance to anticancer treatments. This review summarizes recent
In solid tumours, cancer cells are subjected to very strong and se- advances in the understanding of the regulation of autophagy by mi-
lective metabolic pressures because of limited oxygen and nutrient tochondria through the control of energy metabolism, reactive oxygen
availability. It is now recognized that the tumour microenvironment species production and mitochondrial-associated ER membranes.
has a critical effect on cancer cell selection, tumour growth and pro- Reciprocally, the regulation of mitochondrial mass and functioning by
gression. Only the most metabolically adapted cancer cells will survive mitophagy will be discussed. We will focus on mitochondrial calcium
to this inhospitable microenvironment, and to the stress factors gen- transporters as potential new therapeutic targets for autophagy/mito-
erated by anticancer treatments. Notably, the metabolic plasticity of phagy in cancer treatment.
cancer cells allows them to use various metabolic substrates.
Furthermore, the recycling of intracellular components through au- 2. Targeting mitochondrial metabolism and autophagy in cancer
tophagy participates to the production of metabolic precursors to sus-
tain cell growth. These adaptations are critical for tumour growth, 2.1. Mitochondrial metabolism in tumours
metastasis appearance and the acquisition of resistances to anticancer
treatments. One major challenge for the development of new anticancer Mitochondria are major organelles with primary roles in energy
treatments is to limit the metabolic adaptations of cancer cells which production, Ca2+ and redox homeostasis, and apoptosis. It has long
could allow better therapeutic efficacy and avoid the acquisition of been thought that cancer cells do not produce energy by mitochondrial
resistance mechanisms. Because mitochondria and autophagy partici- oxidation but through a high glycolytic rate coupled with lactic acid
pate to the metabolic adaptation of cancer cells, one potential strategy production even in presence of oxygen namely the Warburg effect (or

Abbreviations: AMPK, AMP-activated protein kinase; CAF, cancer associated fibroblast; Drp1, dynamin-related protein 1; EMT, epithelial-to-mesenchymal transi-
tion; ER, endoplasmic reticulum; FA, fatty acid; FAO, fatty acid oxidation; HIF, hypoxia-inducible factor; IMM, inner mitochondrial membrane; IMS, intermembrane
space; IP3R, inositol trisphosphate receptor calcium channel; MAM, mitochondrial-associated ER membrane; MCU, mitochondrial calcium uniporter; OMM, outer
mitochondrial membrane; OXPHOS, oxidative phosphorylation; PE, phosphatidylethanolamine; PS, phosphatidylserine; ROS, reactive oxygen species; TCA, tri-
carboxylic acid; TRP, transient receptor potential channel; VDAC, voltage-dependent anion channel

Corresponding author at: UMR Inserm 1069, Université de Tours, 10 Boulevard Tonnellé, 37032, Tours, France.
E-mail address: lucie.brisson@univ-tours.fr (L. Brisson).

https://doi.org/10.1016/j.semcdb.2019.05.029
Received 15 March 2019; Received in revised form 29 May 2019; Accepted 29 May 2019
1084-9521/ © 2019 Elsevier Ltd. All rights reserved.

Please cite this article as: Fabio Ferro, et al., Seminars in Cell and Developmental Biology, https://doi.org/10.1016/j.semcdb.2019.05.029
F. Ferro, et al. Seminars in Cell and Developmental Biology xxx (xxxx) xxx–xxx

aerobic glycolysis) [1]. Despite the initial characterization of a major primary tumours [36] and are associated with poor prognosis [37,38].
role of aerobic glycolysis in cancer cells, it is now recognized that mi- The role of autophagy in metastasis formation is complex but it is now
tochondrial oxidative phosphorylation (OXPHOS) is not impaired in all proposed that autophagy participates to all the different steps of the
cells constituting tumours and participates to ATP production [2]. In metastatic cascade. Indeed, recent evidence suggest that autophagy
fact cancer cell metabolism is more complex than initially described promotes cancer cell migration, invasiveness, epithelial-to-mesench-
related to oxygen availability, with the use of a wide range of substrates ymal transition (EMT) and anoikis resistance [39]. Autophagy regulates
including glutamine [3], lactate [4] or fatty acids [5] to fulfil metabolic focal adhesion disassembly through direct degradation of paxillin and
demands. The global reprogramming of cancer cell metabolism not only thus promotes cancer cell migration and metastasis [40]. Interestingly,
supports the production of ATP in metabolic stress conditions but also the autophagic process can promote the secretion of the pro-migratory
leads to the production of building blocks (amino acids, fatty acids, cytokine interleukin-6 (IL-6), matrix metalloproteinase 2 and WNT5A
nucleotides) needed for a high proliferation rate [6]. These building participating both in cancer cell migration and extracellular matrix
blocks are produced by the tricarboxylic acid (TCA) cycle uncoupled to degradation [41]. This unconventional function of autophagy has been
ATP production by OXPHOS (cataplerosis) providing oxaloacetate and described to promote cancer progression through secretion of proteins
α-ketoglutarate for the synthesis of amino acids and citrate as a shuttle stimulating invasiveness, angiogenesis and limiting im-
for the export of acetyl-coA to the cytosol for lipid synthesis. Further- munosurveillance [42]. Furthermore, autophagy is activated during
more an important part of the biomass is derived from glutamine and matrix detachment and promotes anoikis resistance, thus allowing
other amino acids which are able to replenish TCA cycle intermediates cancer cell survival in vessels and during metastatic colonization
(anaplerosis) for amino acid biosynthesis [7]. Metabolites from glyco- [43,44]. Although the molecular mechanism is not clearly understood
lysis and the pentose phosphate pathway are additional important and depends on cancer models and status, autophagy has been asso-
precursors for the synthesis of nucleotides and amino acids needed for ciated with EMT, a process that participates to both cancer metastasis
cancer cell proliferation [6]. and resistance to anticancer treatments [45]. Autophagy is also acti-
Through its important contribution in the production of ATP and vated during anticancer treatments by radiation therapy and che-
macromolecules, it is now well described that mitochondria sustain motherapy. Whether autophagy is associated with cell death during
tumour growth and cancer progression. Indeed mitochondrial function cancer treatment or with cell survival and resistance to anticancer
has been associated with cancer cell proliferation, resistance to cell treatments is still debated. However it is now demonstrated in different
death and metastasis (see reviews [8–11]). Consequently, targeting models that inhibition of autophagy can sensitize cancer cells to several
mitochondria in cancer cells has been proposed to reduce tumour anticancer treatments [46–52].
progression [12–14]. To date, hydroxychloroquine (HCQ), a weak base, lysosomotropic
agent, used as an anti-malarial drug, is the only autophagy inhibitor
2.2. Autophagy clinically available. HCQ has been tested in several clinical trials as a
broad spectrum inhibitor of autophagy in association with che-
Autophagy is a cellular catabolic pathway leading to the degrada- motherapy or radiotherapy. In patients with several types of solid tu-
tion and recycling of proteins and organelles, following the fusion be- mours, studies showed the safety of HCQ and its ability to target au-
tween an isolation vesicle, the autophagosome, and a lysosome pro- tophagy [53]. However, its use is limited by side effects independent of
viding hydrolytic enzymes. Macroautophagy (hereafter called autophagy [54–56]. Furthermore, HCQ activity decreases with acidity,
autophagy) is a non-selective form of autophagy, whereas several forms which is found in the extracellular compartment around tumours [57].
of autophagy are selective for intracellular organelles like mitochondria Therefore, a better understanding of the regulations of autophagy is
and therefore called mitophagy (see section mitophagy). The molecular needed to propose new strategies for targeting cancer, depending on
process of autophagy is complex and involves sequential steps of nu- tumour type, stage and metabolism.
cleation, elongation, and fusion through several proteins including the
autophagy-related (Atg) proteins (for review [15–17]). Autophagy 2.3. Mitochondrial metabolism in regulation of autophagy
connects substrate availability in the environment with cellular meta-
bolic requirements. Indeed, autophagy is activated by nutrient starva- 2.3.1. Energy metabolism
tion, as well as under oxidative stress conditions, through highly Besides the activation of autophagy by low nutrient availability in
regulated pathways linked with energy metabolism. These pathways the tumour microenvironment, the metabolic reprogramming of tu-
mostly converge on mTORC1 and the energy-sensing AMP-activated mour cells can also support autophagy activation (Fig. 1). Autophagy is
protein kinase (AMPK) [18–21]. Autophagy has two major physiolo- highly regulated by ATP production and AMPK pathway. Therefore, a
gical roles: the breakdown of dysfunctional proteins or organelles as a reduction of ATP production leads to the accumulation of AMP, the
quality control mechanism, and the recycling of macromolecules under activation of AMPK and autophagy. Indeed, mitochondrial dysfunction
nutrient stress conditions in order to sustain metabolic demands induced by the inhibition of respiratory chain and complex III knock-
[22,23]. out activates AMPK and autophagy [58,59] to promote cell survival
It is now well admitted that autophagy can have two roles in cancer: [59]. Autophagy is transcriptionally regulated during starvation by the
protective at early stage of the tumour but promoting tumour growth at induction of transcription factor TFEB which activates the transcription
later stages [24,25]. Knockout of autophagy genes in mice increases of Atg genes [60]. The acute inhibition of mitochondrial OXPHOS in-
tumour multiplicity mainly by increasing oxidative stress, indicating duces the expression of TFEB and lysosomal biogenesis [61], which can
that autophagy protects against cancer development [26,27]. However, also participate to the activation of autophagy by mitochondrial dys-
in established tumours, the activation of autophagy by the extracellular function in cancer.
conditions (hypoxia, low nutrients and growth factors, reactive oxygen Unlike the activation of autophagy by mitochondrial dysfunction, it
species (ROS), and lactate) [28–30] and by oncogenes Ras and p53 [31] has been demonstrated in yeast and in mammalian cells that mi-
sustains the survival of cancer cells and is favourable to tumour growth tochondrial activity is required for autophagy induction [62,63]. In
[24,28,32,33]. In addition, autophagy plays a prominent role in the yeast, amino acid starvation required functional mitochondria to induce
tumour microenvironment by modulating the interactions between LC3 expression and the activation of autophagic flux [62]. Interestingly,
cancer cells and non-cancer cells from the tumour [34,35]. Besides it has been demonstrated that mitochondria regulates autophagy
these apparently opposite roles in tumour initiation and tumour through the cAMP-dependent kinase PKA [62]. It is not known whether
growth, autophagy has also been associated with cancer progression. this requirement of mitochondrial activity for autophagy occurs in
Markers of autophagy are increased in metastases compared with cancer cells and how mitochondrial metabolic reprogramming in

2
F. Ferro, et al. Seminars in Cell and Developmental Biology xxx (xxxx) xxx–xxx

Fig. 1. Regulation of autophagy and mito-


phagy by cancer cell metabolism. Cancer cells
use a wide range of metabolic substrates in-
cluding glucose (blue arrows), lactate, fatty
acids (green arrows) and glutamine (orange
arrows) to sustain the production of ATP and
intermediate precursors for nucleotides, fatty
acids (FA) and amino acids (aa) synthesis
needed for proliferation. Autophagy and mi-
tophagy are highly regulated by nutrients
availability and reprogrammed metabolic
pathways used by cancer cells. When ATP
production is sufficient, the inactivation of
AMPK pathway represses autophagy and mi-
tophagy. The production of α-ketoglutarate (α-
KG) through glutaminolysis is a major com-
ponent of aa sensing and of autophagy re-
pression under high substrate availability.
However, ammonia, lactate and reactive
oxygen species (ROS) production are asso-
ciated with autophagy activation. Fatty acid
oxidation (FAO) can either activate or repress
autophagy. Mitochondria are also involved in
autophagy regulation through the formation of
Mitochondria-Associated endoplasmic re-
ticulum (ER) Membrane (MAM) subdomains
which are important for ion and lipid (parti-
cularly phosphatidylserine, PS) exchanges be-
tween mitochondria and ER. On the one hand, MAM formation and the remodelling of mitochondrial membrane phospholipids, with production of phosphatidy-
lethanolamine (PE) and subsequent LC3-PE promotes autophagy. On the other hand, the entry of Ca2+ from the cytosol and from the ER in MAM allows the proper
functioning of mitochondrial enzymes and therefore represses autophagy. Similarly, the specific recycling of mitochondria by mitophagy is repressed by the
maintenance of Ca2+ flux in mitochondria and by sustained ATP production. However, mitochondrial calcium channels voltage-dependent anion channel (VDAC),
transient receptor potential channel M2 (TRPM2) and mitochondrial calcium uniporter (MCU) are involved in mitophagy activation by the protein Parkin or by
hypoxia. In normal conditions, cardiolipins, specific mitochondrial phospholipids, are mostly located in the inner mitochondrial membrane (IMM). Cardiolipin
externalization to the outer mitochondrial membrane (OMM) leads to its interaction with LC3 and acts as an elimination signal for mitophagy. Yellow pointed arrows
(–>) represent a stimulation of mitophagy or autophagy. Yellow blunt-end arrows represent a repression or inhibition (——I) of mitophagy or autophagy. AcCoA is
for acetyl-coenzyme A. IMS is for inter membrane space, limited by the OMM and IMM. OXPHOS is for mitochondrial oxidative phosphorylation. TCA cycle is for
tricarboxylic acid cycle.

cancer cells can activate autophagy through PKA regulation. However, condition of metabolic stress [70]. Furthermore, ammonia is at the
the requirement of mitochondrial activity for autophagy has been de- basis of a self-fuelling system between cancer-associated fibroblasts
monstrated also in mammalian cells and involves mitochondrial phos- (CAF) and cancer cells. The glutamine produced by CAF is imported in
pholipid remodelling (see section Mitochondrial-Associated ER Mem- epithelial cancer cells to fuel the TCA cycle after its conversion to
branes) [63]. glutamate and α-ketoglutarate, while the ammonia produced diffuses
Autophagy is highly regulated by the presence or absence of me- and activates autophagy in CAF [71].
tabolic substrates in the microenvironment. Therefore, several signal- Another particular adaptation of cancer cells which depends on
ling pathways controlled by substrate availability in the tumour mi- autophagy is the metabolic symbiosis based on lactate exchanges be-
croenvironment, including glutamine, lactate or fatty acids have been tween glycolytic, lactate-producing cells, and oxidative, lactate-con-
associated with autophagy. Glutamine and leucine metabolism is linked suming cells. These lactate exchanges have been described between
with amino acid sensing via the lysosomes and mTORC1. The produc- cancer cells from different metabolic phenotypes [4] and between
tion of α-ketoglutarate through glutaminolysis has been demonstrated cancer cells and fibroblasts [72]. This cooperation requires the entry of
to be a major component of the amino acid sensing and of autophagy lactate into the oxidative cells, a process facilitated by the lactate-
regulation. Indeed α-ketoglutarate is necessary for the activity of the proton symporter monocarboxylate transporter type 1 (MCT1), and its
EGLNs/prolyl hydroxylases, leading to the activation of the small oxidation to pyruvate by lactate dehydrogenase B (LDHB). In oxidative
GTPase RRAGB and subsequent translocation and activation of cancer cells, lactate is used for OXPHOS [4] and for the production of
mTORC1 to the lysosome and inhibition of autophagy [64–66]. In lipids [73]. Interestingly the oxidation of lactate by LDHB also parti-
cancer cells, the long term activation of glutaminolysis in condition of cipates to lysosome function and activates autophagy in cancer cells
amino acid restriction induces the activation of mTORC1, represses [28]. The activation of autophagy by lactate utilisation promotes cancer
autophagy and activates apoptosis [67]. However the role of glutamine cell survival and tumour growth whereas it has no effect on autophagy
metabolism in autophagy regulation is complex. Besides fuelling the and the survival of non-cancer cells [28].
TCA cycle with α-ketoglutarate, glutaminolysis produces ammonia In cancer cells, fatty acid (FA) metabolism is central to cancer
which has long been considered as a by-product before the discovery of progression since FA synthesis provides building blocks and FA oxida-
its role in autophagy [68]. Although the mechanism is not clearly un- tion (FAO) is an important energy source [74]. The relationship be-
derstood, ammonia activates autophagy through AMPK and the un- tween FA and autophagy is not clearly understood, and both activation
folded-protein response system independently of mTOR [69]. Ammonia and inhibition of FAO have been associated to autophagy. In prostate
is found in tumours at high concentration in the interstitial fluid [68]. cancer cells, the inhibition of FAO using etomoxir (Carnitine Palmi-
Because ammonia is diffusible, it is proposed that glutaminolysis-de- toylTransferase I inhibitor) activates autophagy through the activation
rived ammonia activates autophagy even in the hypoxic core of the of AMPK and inhibition of mTOR [75]. On the opposite, in colon cancer
tumour as an adaptation mechanism to protect against cell death in cells, the activation of FAO activates autophagy through the activation

3
F. Ferro, et al. Seminars in Cell and Developmental Biology xxx (xxxx) xxx–xxx

of AMPK and promotes tumour growth [76]. In this model, surrounding first evidence that mitochondrial membrane participates to autopha-
adipocytes provide cancer cells with FA to fuel FAO and promote sur- gosome formation came from the identification of phospholipid trans-
vival under low nutrient conditions [76]. Bone marrow adipocytes can fers from mitochondria to autophagosome in starved cells [92]. The
also activate FAO and AMPK in acute leukaemia cells, leading to au- role of MAM in autophagosome formation seems to involve the con-
tophagy activation [77]. This relation of tumours with the surrounding version of phosphatidylserine (PS) from the ER to phosphatidyletha-
adipose tissue is particularly important for cancer progression and may nolamine (PE) by inner mitochondrial membrane (IMM) phosphati-
potentially limit cancer treatment. Furthermore, the relation of lipid dylserine decarboxylase [63], allowing PE-LC3 conjugation, and
metabolism and autophagy is complex because reciprocally, autophagy autophagy [93]. It has recently been demonstrated that the activation
can provide FA for FAO [78]. of autophagy by mTOR inhibition induced an increase in MAM for-
mation and the remodelling of the mitochondrial membrane phospho-
2.3.2. Reactive oxygen species lipids, with an increase in PS and PE [63]. Interestingly, the activity of
Mitochondria is the major source of ROS. An elevated level of ROS is complex I is required for MAM formation, phospholipid remodelling
an important feature of cancer cells, and could be attributed to mi- and autophagy [63].
tochondrial electron transport chain activity (mainly Complex I and On the other hand, some studies showed that the disruption of MAM
Complex III). Increased ROS production by mitochondria in cancer cells activates autophagy. MAM are important sites for the transfer of Ca2+
is associated with metastasis through the activation of Src and the Focal from the ER to the mitochondria to regulate mitochondrial enzymes as
adhesion kinase Pyk2 [12]. Although a massive ROS generation is respiratory complexes and TCA cycle enzymes. This Ca2+ flux occurs
mainly associated with cell damage and cell death, moderate ROS mainly through IP3 Receptor and TRPM8 in the ER membrane [94,95]
production is known to activate autophagy through multiple pathways. and through the Voltage-Dependent Anion Channel (VDAC), the Mi-
Autophagy is activated by ROS through the activation of the sensor tochondrial Calcium Uniporter (MCU) and the Transient Receptor
Ataxia-telangiectasia mutated (ATM) leading to mTORC1 inhibition Protein Melastatin-related 2 (TRPM2) in the mitochondrial membranes
through the LKB1/AMPK/TSC2 pathway [79]. Oxidative stress can also [96,97] (See section” mitochondrial calcium transporters as a new
induce autophagy through an NFkB-dependent upregulation of p62 target for autophagy/mitophagy modulation”). The interruption of
[80]. Furthermore, ROS production is involved in starvation-induced Ca2+ flux between the ER and mitochondria decreases OXPHOS, in-
autophagy [81]. During nutrient starvation, the ROS produced reg- duces metabolic stress and activates autophagy as a survival mechanism
ulates the activity of the HsAtg4 protease, which is important for [94]. However, the activation of autophagy by MAM disruption in
phosphatidylethanolamine (PE) conjugation to LC3 [81]. Mitochondrial cancer cells does not seem sufficient, unlike in normal cells, to maintain
ROS can also regulate autophagy through oxidation of lysosomal the energetic level needed, thus leading to cancer cell death and de-
membrane proteins and modulation of Ca2+ homeostasis [82,83]. The crease in tumour growth [98]. Furthermore, tightening ER-mitochon-
activation of TRPML1, a lysosomal Ca2+-permeable transient Receptor dria contact sites can inhibit rapamycin-induced autophagy but not
Potential (TRP) channel, by ROS induces lysosomal Ca2+ release, fol- starvation-induced autophagy suggesting that MAM play different roles
lowed by the activation of calcineurin signalling, TFEB nuclear trans- depending on autophagy stimuli [99].
location and activation of autophagy and lysosome biogenesis [82]. On Several oncoproteins and tumour suppressor proteins have been
the opposite, the activation of the lysosomal Ca2+ channel TRPM2 by identified in MAM, which highlighted the complex relationships be-
ROS induces Ca2+ CaMKII signalling, leading to autophagy inhibition tween these structures and cancer progression. Indeed, MAM is now
[83]. Among ROS species, it has been proposed that superoxide O2•− is proposed to play an important role in cancer metabolism and growth
the major regulator of autophagy since superoxide production induced [100]. However, the alterations of MAM formation or architecture in
by the dysfunction of mitochondrial complexes I and III activates au- cancer are not clearly understood. Although still controversial, the re-
tophagy [84]. cent association of MAM with autophagy regulation might be re-
Although the mechanisms are not clearly described, elevated ROS sponsible for their important roles in cancer. Pharmacological inter-
production caused by dysfunction of mitochondria can participate to vention could be a strategy to disrupt MAM and inhibit both
the regulation of autophagy in tumours, and consequently to cancer mitochondrial metabolism and autophagy [63]. A better understanding
progression. ROS are produced during extracellular matrix detachment, of the involvement of MAM in cancer progression and its involvement
which is an important step for metastatic dissemination [85,86]. It has in the regulation of autophagy is needed to propose new targeted
been demonstrated that ROS production during matrix detachment therapies.
promotes autophagy through the activation of PERK1 and promotes Altogether these studies highlight the complex regulation of au-
anoikis resistance [87]. Interestingly active PERK1 and LC3 are in- tophagy by mitochondrial metabolism in tumours. On the one hand
creased in human breast ductal carcinoma compared to normal breast autophagy is activated by metabolic changes linked with a decrease in
suggesting an important role of this signalling pathway during cancer ATP production but also by ammonia production, ROS and interruption
progression [87]. ROS are also produced following anticancer treat- of Ca2+ transfers in MAM. On the other hand recent evidences suggest
ments such as chemo and radio-therapies [88]. Although ROS play an that mitochondrial function participates to autophagy induction as a
important part for the mechanism of action of these anticancer treat- lipid source or through the activation of PKA. Since mitochondria and
ments, the activation of autophagy by ROS could participate to cancer autophagy are important players of cancer progression and resistance to
cell survival and therapy resistance. This has been demonstrated in treatment, it is crucial to better understand their mutual regulations.
bladder cancer cells in which the oxidative stress induced by capsaicin Therefore more studies are needed to clarify the regulation of autop-
activates autophagy as a pro-survival mechanism promoting drug re- hagy by mitochondria in different tumour types and during tumor-
sistance [89]. In lung adenocarcinoma, ROS induced by low dose ra- igenesis. Furthermore, the tumour microenvironment is much more
diations promote autophagy and confer radio-resistance [90]. complex than what can be reproduced in in vitro cell culture models: it
contains various cell types like cancer-associated fibroblasts, adipo-
2.3.3. Mitochondrial-Associated ER Membranes (MAM) cytes, immune cells, as well as gradients for oxygen and metabolites. All
The juxtaposition of membranes from the ER and mitochondria, of these features of the tumours microenvironment require being as-
including specific proteins, forms a subdomain called MAM (Fig. 1). sessed in vivo. Interestingly the microenvironment of tumours can also
This interface plays an important role for ion and lipid exchanges be- play a role in autophagy regulation in cancer cells by providing lactate,
tween mitochondria and ER allowing the proper function of the two glutamine or FA and by regulating tumour-stroma interaction [34,35].
organelles [91]. Mitochondrial membranes and MAM have been sug- Therefore it is important to clarify the effects of metabolic drugs on
gested to participate to autophagosome formation as lipid source. The autophagy in tumours and to consider their association with autophagy

4
F. Ferro, et al. Seminars in Cell and Developmental Biology xxx (xxxx) xxx–xxx

inhibitors. limit ROS production, its function has been associated with tumour
suppression. Indeed, Parkin or PINK1 deletion in mice leads to the
3. Targeting mitophagy in cancer spontaneous development of hepatocellular carcinoma [121] and in-
creases Kras-driven pancreatic tumorigenesis [122]. In human, Parkin
3.1. Mitophagy regulation and function deletion has been identified in tumours including colorectal cancer
[123], glioblastoma [124], melanoma [125], lung cancer [126] and
Mitochondrial mass is highly dynamic and tightly regulated through breast cancer [127,128]. Loss of Parkin increases proinflammatory
fusion/fission, but also by mitobiogenesis and mitophagy. Mitophagy is signals, promotes genomic instability [126], increases cancer cell pro-
a specific form of autophagy that selectively degrades mitochondria liferation and resistance to apoptosis [121,125]. Interestingly the ac-
through PINK1/Parkin and BNIP3/NIX/FUNDC1 pathways. It is trig- cumulation of mitochondrial dysfunctions induced by Parkin deficiency
gered by mitochondrial depolarization, hypoxia and metabolic stress. decreases mitochondrial OXPHOS, increases ROS production and in-
The PINK1/Parkin pathway has been originally identified in creases glycolysis, therefore possibly contributing to the Warburg ef-
Parkinson’s disease where mutations in corresponding genes impair fect, and consequently increases tumorigenesis [129]. Furthermore,
mitochondrial clearance. The activation of mitophagy by membrane Parkin has been identified as an E3 ubiquitin ligase for HIF-1α to
depolarization involves the mitochondrial kinase PINK1, the localisa- promote its proteasomal degradation. Therefore, the downregulation of
tion of which depends on mitochondrial membrane potential [101]. In Parkin in breast cancer cells stabilizes HIF-1α and promotes metastasis
normal conditions, the voltage-dependent translocation of PINK1 to the [130]. The BNIP3 pathway has also been linked with a tumour sup-
IMM induces its cleavage by the mitochondrial protease PARL. When pression function of mitophagy. A decrease of BNIP3 is found in pan-
IMM is depolarized, PINK1 accumulates in the outer mitochondrial creatic cancers and associated with chemoresistance and poor prognosis
membrane (OMM) where it phosphorylates the E3 ubiquitin ligase [131,132]. In mammary tumours, BNIP3 decreases with cancer pro-
Parkin, leading to its recruitment at the OMM [102]. Several mi- gression, leading to an accumulation of mitochondria associated with
tochondrial proteins, including VDAC1, Miro, Mfn-1 and Mfn-2, are an increase ROS production and HIF-1α stabilization [133]. The au-
ubiquitinylated by Parkin [103] and recognized by LC3-interacting thors demonstrated that the loss of BNIP3 promotes mammary tumour
proteins p62, NDP52, optineurin, TAX1BP1 or NBR-1 to recruit the growth, progression to metastasis and can be used as a prognostic
autophagic machinery [104–108]. Mitophagy is also activated by hy- marker in human triple-negative breast cancer [133]. Lastly, FUNDC1-
poxia through the induction of adaptor proteins: BNIP3, NIX (or deficient mice showed an increase in tumour initiation, also suggesting
BNIP3L) and FUNDC1. These proteins are localized in the OMM and a tumour suppressor role for FUNDC1 [134]. Altogether, these studies
contain an LC3-interacting motif to promote the recruitment of the highlighted the role of mitophagy as a tumour suppressor and the po-
autophagic machinery [109]. The expression of BNIP3 and NIX is in- tential benefit of its induction to limit the consequences of mitochon-
creased by hypoxia through Hypoxia-inducible factor (HIF-1) drial dysfunction in cancer.
[110,111] whereas FUNDC1 is dephosphorylated in hypoxia by Src Like non-selective autophagy, the role of mitophagy is complex and
kinase inactivation, increasing its affinity for LC3 compared to phos- can depend on tumour type and stage. Although mitophagy has been
phorylated FUNDC1 [112]. In addition, AMPK activation by low nu- associated with tumour suppression in early stages of the tumour de-
trient availability can directly activate mitophagy through phosphor- velopment, it can promote tumour growth at advanced stages. Indeed,
ylation of Ulk1 [20]. mitophagy can be induced when AMPK is activated by low nutrient
Besides the activation of PINK1/Parkin and BNIP3/NIX/FUNDC1 conditions [20], a condition found in tumours. Hypoxia, a common
pathways, mitophagy is highly linked to mitochondrial function, en- feature of solid tumours, has been shown to activate mitophagy in
abling to equilibrate mitochondrial degradation with mitochondrial cancer cells through BNIP3 and NIX induction [110]. Furthermore, the
activity requirement. Mitophagy is activated by OXPHOS activity to induction of mitophagy depends on mitochondrial metabolism, such as
stimulate the renewal of mitochondria and maintain their efficiency glutamine availability and use which can increase BNIP3 expression
[113]. In addition, mitophagy induces the translocation of Nrf2 and and mitophagy in melanoma cells [135]. While few studies investigated
TFEB transcription factors to the nucleus, inducing the transcription of mitophagy in tumours, its activity seems dependent on tumour meta-
autophagy, lysosome and mitochondria-related genes to renew lyso- bolism and microenvironment.
somal and mitochondrial compartments [114]. Furthermore, the re- In several cancer types, the degradation of damaged mitochondria
cognition of injured mitochondria for degradation through mitophagy and the recycling of metabolic precursors by mitophagy can promote
is regulated by mitochondrial membrane composition. In normal con- cell survival and protect from cell death [110,136]. In Parkin-deficient
dition, cardiolipins, specific mitochondrial phospholipids, are mostly mice melanoma growth and metastasis are suppressed, suggesting a
located in the IMM. Cardiolipin externalization to the OMM leads to its pro-tumour role of Parkin-dependent mitophagy [137]. However,
interaction with LC3 and acts as an elimination signal for mitophagy Parkin is often downregulated in many tumour types. It has been re-
[115]. The hexameric intermembrane space protein, NDPK-D, has been cently demonstrated that besides this frequent loss of parkin, PINK1-
identified as the cardiolipin translocation system, in case of mi- dependent mitophagy can be functional through the induction of the E3
tochondrial depolarisation, which is needed for the elimination of da- ubiquitin ligase ARIH1/HHAR [138]. Interestingly ARIH1 is over-
maged mitochondria by mitophagy [116]. expressed in breast and lung cancer cells while Parkin expression is low
Mitophagy cooperates with mitochondrial dynamic and it is now [138]. In addition, BNIP3 induction has been reported to promote cell
admitted that mitochondrial fission precedes mitophagy. The mito- migration through cytoskeleton remodelling [139], resistance to an-
phagy proteins Parkin and BNIP3 interact with the mitochondrial fis- oikis [140] and invasion [141], suggesting a role of BNIP3-dependent
sion factor dynamin-related protein (Drp) 1 [117,118]. This coopera- mitophagy in metastasis formation. The pro-survival role of mitophagy
tion is further enhanced in hypoxia with the recruitment of FUNDC1 in has also been associated with resistance to anti-cancer treatments, such
MAM due to its interaction with the ER protein calnexin, leading to the as the ARIH1-PINK1 mitophagy which protects against cisplatin-in-
recruitment of Drp1 to allow mitochondrial fission and mitophagy duced cell death [138]. A role of mitophagy in resistance to anti-cancer
[119]. Furthermore, during mitophagy induction, PINK1 and Beclin1 treatment is also suggested in a study showing that doxorubicin induces
relocalize to the MAM and promote autophagosome formation [120]. the expression of NIX in cancer stem cells and promotes resistance to
treatment through limitation of ROS production [142]. Interestingly,
3.2. Mitophagy in cancer FUNDC1 silencing has been proposed to improve chemotherapy effi-
ciency by increasing cisplatin-induced cell death [143].
Because mitophagy can degrade dysfunctional mitochondria and Consistently with the dual role of mitophagy in cancer progression,

5
F. Ferro, et al. Seminars in Cell and Developmental Biology xxx (xxxx) xxx–xxx

the expression of mitophagy proteins has been found to be either Therefore the inhibition of cation flux could have a dual effect in re-
downregulated or overexpressed in different cancer types. BNIP3 has ducing mitochondrial function but also mitophagy by preventing mi-
been found downregulated in invasive ductal carcinoma and metastases tochondrial depolarization.
[144] and in pancreatic cancers [131,132] but upregulated in breast Among cations, Ca2+ exchanges have been associated with autop-
ductal carcinoma in situ [145] and in other human cancer tissues hagy and mitophagy regulation. VDAC is the most abundant protein in
(ovarian, head and neck squamous cell carcinoma) where BNIP3 loca- the OMM, and is ubiquitously expressed in all eukaryotic cells. In its
lized in perinecrotic regions [111]. It has been suggested that some of open state it becomes an aspecific pore, allowing the flux of many
these mitophagy proteins can be used as prognosis markers in specific substrates and ions smaller than 5 kDa. In this condition, VDAC is
cancers. A high expression of FUNDC1 has been associated with poor highly Ca2+ permeable and allows Ca2+ flux to IMS, thus modulating
prognosis in cervical cancer [143]. High BNIP3 expression can be used Ca2+ availability to Ca2+ permeant-channels in the IMM. MCU is a
as a prognosis marker in specific cancers such as uveal melanoma where Ca2+ channel present in the IMM, ubiquitously expressed and generally
a high BNIP3 expression is associated with a poor prognosis [146]. considered as the main Ca2+ transporter in the matrix. Two pools of
However, there is a controversy concerning the role of BNIP3-depen- calcium can be imported into the mitochondria: one from the cytosol
dent mitophagy in cancer that could be explained by an alternatively and another one from the ER. The juxtaposition of ER and mitochondria
spliced BNIP3 expressed preferentially in cancer cells: whereas the full- in MAM facilitates the generation of a high Ca2+ concentration that
length BNIP3 promotes cell death, the Bnip3Δex3 isoform promotes cell could be efficiently transferred to mitochondrial compartments. This
survival [147]. Interestingly, a switch towards a glycolytic metabolism Ca2+ flux in MAM has important roles for the regulation of mi-
increases the expression of the Bnip3Δex3 isoform [147]. tochondrial function since its interruption decreases OXPHOS and ac-
Altogether, these studies highlighted the complex role of mitophagy tivates autophagy [94] [98]. On the opposite, the disruption of MAM
in cancer progression, showing either pro-tumour or anti-tumour ac- has been associated with a decrease in mitochondrial PE production
tivities depending on tumour type, stage, or its metabolic activity. and therefore a reduction of autophagy [63]. Although the mechanisms
However, recent evidence suggest an important role of mitophagy in linked with autophagy are not clearly understood, VDAC and MCU
tumour growth, metastasis and therapy resistance. Since mitophagy is could be interesting targets to disrupt Ca2+ and lipid exchanges in
an important regulator of mitochondrial number and function, its MAM in cancer cells, decreasing mitochondrial function and inducing
pharmacological modulation in tumours could be a promising antic- cell death. Furthermore, VDAC is involved in Parkin recruitment to
ancer strategy. However, the proposition of mitophagy inhibition or defective mitochondria, triggering mitophagy [155]. The Ca2+ fluxes in
activation is limited by the little knowledge of its role and regulation in MAM through MCU is also important for Parkin-dependent mitophagy
preclinical models and in human tumours. Therefore, a major challenge [156]. Because of their double role in mitochondrial Ca2+ flux and
will be to identify the specific roles of the different mitophagy pathways Parkin recruitment, VDAC and MCU could be used to target mitophagy
in human cancers before being able to propose mitophagy modulators and autophagy regulation in tumours. VDAC is overexpressed in several
for anti-cancer therapy. The identification of mitophagy biomarkers tumour types and pre-clinical models, suggesting that it could be an
will be helpful to design mitophagy modulators for tumours having a interesting anticancer target [157,158]. Although the mechanism has
high requirement for mitophagy. It is important to clarify the role of not been linked to autophagy, VDAC downregulation decreases mi-
mitophagy in tumours and to investigate the effect of its modulation in tochondrial membrane potential, cellular ATP content, cancer cell mi-
tumour-bearing mice models to reflect the behaviour of tumours de- gration, proliferation and tumour growth in lung cancer xenografts
tected in patients. It is also important to develop specific tools to [159]. MCU has also been found to be altered in tumours from different
modulate mitophagy in vivo and not using general autophagy inhibitors. tissues [160]. In particular, in human breast [161,162] and hepato-
This issue could be addressed using mitochondrial targeted molecules, cellular [163] carcinomas, the expression of MCU is associated with
for example using triphenylphosphonium derivatives structures [148], cancer progression and metastasis.
to deliver autophagy inhibitors. Interestingly using this strategy with The MCU regulator 1 (MCUR1) is a secondary subunit of MCU
mitochondria-targeted redox agents can activate mitophagy selectively complex allowing ion flux into the matrix [164,165]. MCUR1 down-
in breast cancer cells compared to non-cancer cells [149]. regulation decreases Ca2+ flux, OXPHOS, ATP production and activates
AMPK-dependent autophagy [164,165]. MCUR1 is upregulated in he-
4. Mitochondrial calcium transporters as new targets for patocellular carcinoma and its expression is associated with cancer cell
autophagy/mitophagy modulation survival and tumour growth [166]. In this model, MCUR1 induces
mitochondrial Ca2+ accumulation and ROS production, showing that
The OXPHOS activity leads to a strong proton concentration in the MCUR1 association with MCU is necessary for a functional MCU com-
intermembrane space (IMS) and is the main factor that determines the plex and its Ca2+-associated entry into mitochondria is a pro-survival
very negative voltage (Ψm) of the IMM. Therefore the modulation of factor for cancer cells [166].
OXPHOS activity can influence the proton gradient and consequently Several other ion channels have been identified in the IMM of dif-
the IMM potential. The electrochemical gradient in the IMS is favour- ferent cell type and could compensate for MCU-dependent Ca2+ flux.
able to cation influx (mainly K+ and Ca2+) into the mitochondrial Among them, two splice variants of TRPM2 are expressed in neuro-
matrix and regulates the activity of mitochondrial enzymes, ROS and blastoma. The functional TRPM2-L and the dominant negative TRPM2-
ATP production. This cation fluxes is tightly regulated by the expression S are both localized in mitochondria [167]. In xenograft models of SH-
of ion channels in the IMM: BKCa, KATP, Kv1.3, MCU, SK-family mem- SY5Y glioblastoma cell line, the expression of the dominant negative
bers, TRP-family members, CLIC4 and others, while the main actor in isoform decreases mitophagy and led to smaller tumour growth,
the OMM that allows ion flux is VDAC [96,97,150,151] (Fig. 1). An showing that a functional mitochondrial TRPM2 is necessary for mi-
interruption of cation fluxes decreases OXPHOS activity and on the tophagy and cancer growth [167]. In this model, TRPM2-S was asso-
opposite a high cation influx induces mitochondrial membrane depo- ciated with a reduction in HIF-1/2α and BNIP3 expression [167]. The
larization [152,153]. Since autophagy and mitophagy are linked with role of mitochondrial TRMP2 in mitophagy has also been demonstrated
mitochondrial function, the modulation of mitochondrial ion channels in gastric cancer cell lines where the downregulation of TRPM2 de-
could be an interesting strategy to modulate autophagy or mitophagy in creases mitochondrial oxygen consumption and ATP production asso-
tumours. Indeed, mitochondrial depolarization induces mitophagy ciated with a reduction in mitophagy and autophagy through a JNK-
through the PINK1/Parkin pathway [101,102] and the disruption of dependent and mTOR-independent pathway [168]. The reduction of
mitochondrial potential by FCCP and CCCP activates autophagy and mitophagy and autophagy by the lack of functional TRPM2 is also as-
mitophagy through Nrf2 pathway and TFEB transcription factor [154]. sociated with enhanced efficacies of doxorubicin, tamoxifen and

6
F. Ferro, et al. Seminars in Cell and Developmental Biology xxx (xxxx) xxx–xxx

paclitaxel chemotherapies, showing the potential benefit of targeting [9] P.E. Porporato, N. Filigheddu, J.M.B. Pedro, G. Kroemer, L. Galluzzi,
mitochondrial TRPM2 for cancer therapies [167–169]. Interestingly, Mitochondrial metabolism and cancer, Cell Res. 28 (3) (2018) 265–280.
[10] W.X. Zong, J.D. Rabinowitz, E. White, Mitochondria and Cancer, Mol. Cell 61 (5)
TRPM2 inhibition or silencing did not increase doxorubicin cytotoxicity (2016) 667–676.
in non-cancerous HMEC cell line [169]. [11] J.F. Dumas, L. Brisson, S. Chevalier, K. Maheo, G. Fromont, D. Moussata,
P. Besson, S. Roger, Metabolic reprogramming in cancer cells, consequences on pH
and tumour progression: integrated therapeutic perspectives with dietary lipids as
5. Conclusion adjuvant to anticancer treatment, Semin. Cancer Biol. 43 (2017) 90–110.
[12] P.E. Porporato, V.L. Payen, J. Perez-Escuredo, C.J. De Saedeleer, P. Danhier,
Altogether, these studies highlighted the complex relation between T. Copetti, S. Dhup, M. Tardy, T. Vazeille, C. Bouzin, O. Feron, C. Michiels,
B. Gallez, P. Sonveaux, A mitochondrial switch promotes tumor metastasis, Cell
mitochondria (ATP and ROS production, MAM and calcium transpor- Rep. 8 (3) (2014) 754–766.
ters) and autophagy. Autophagy could result, as a metabolic adaptation, [13] S.E. Weinberg, N.S. Chandel, Targeting mitochondria metabolism for cancer
from chemotherapies targeting mitochondria. In this context, targeting therapy, Nat. Chem. Biol. 11 (1) (2015) 9–15.
[14] Y. Zhu, A.E. Dean, N. Horikoshi, C. Heer, D.R. Spitz, D. Gius, Emerging evidence
both autophagy and mitochondria could limit chemoresistance me-
for targeting mitochondrial metabolic dysfunction in cancer therapy, J. Clin.
chanisms. However, recent discoveries suggest that mitochondria are Invest. 128 (9) (2018) 3682–3691.
also involved in autophagic activity, mainly by providing lipids for [15] Y. Ichimura, T. Kirisako, T. Takao, Y. Satomi, Y. Shimonishi, N. Ishihara,
autophagosome formation. Because of these two effects on autophagy, N. Mizushima, I. Tanida, E. Kominami, M. Ohsumi, T. Noda, Y. Ohsumi, A ubi-
quitin-like system mediates protein lipidation, Nature 408 (6811) (2000)
mitochondria could be considered as a target for cancer therapy. 488–492.
Mitophagy seems to play an important role in cancer cells, but its effects [16] N. Mizushima, T. Noda, T. Yoshimori, Y. Tanaka, T. Ishii, M.D. George,
on cancer cell growth could be different depending on carcinogenesis D.J. Klionsky, M. Ohsumi, Y. Ohsumi, A protein conjugation system essential for
autophagy, Nature 395 (6700) (1998) 395–398.
stage and cancer types. Therefore, mitophagy modulation could be [17] G. Zaffagnini, S. Martens, Mechanisms of selective autophagy, J. Mol. Biol. 428 (9
beneficial for metastasis prevention and improve therapy efficacy. Pt A) (2016) 1714–1724.
Among mitochondrial proteins, calcium transporters have a key role in [18] N. Hosokawa, T. Hara, T. Kaizuka, C. Kishi, A. Takamura, Y. Miura, S. Iemura,
T. Natsume, K. Takehana, N. Yamada, J.L. Guan, N. Oshiro, N. Mizushima,
the regulation of mitochondrial function. These mitochondrial calcium Nutrient-dependent mTORC1 association with the ULK1-Atg13-FIP200 complex
transporters have been recently associated with autophagy and mito- required for autophagy, Mol. Biol. Cell 20 (7) (2009) 1981–1991.
phagy regulation in cancer cells. Consequently they also represent at- [19] C.H. Jung, C.B. Jun, S.H. Ro, Y.M. Kim, N.M. Otto, J. Cao, M. Kundu, D.H. Kim,
ULK-Atg13-FIP200 complexes mediate mTOR signaling to the autophagy ma-
tractive anticancer targets to modulate mitochondrial dysfunction and chinery, Mol. Biol. Cell 20 (7) (2009) 1992–2003.
autophagy/mitophagy in tumours. Studies are still needed to better [20] D.F. Egan, D.B. Shackelford, M.M. Mihaylova, S. Gelino, R.A. Kohnz, W. Mair,
understand the regulations of autophagy and mitophagy by cancer D.S. Vasquez, A. Joshi, D.M. Gwinn, R. Taylor, J.M. Asara, J. Fitzpatrick, A. Dillin,
B. Viollet, M. Kundu, M. Hansen, R.J. Shaw, Phosphorylation of ULK1 (hATG1) by
metabolism, and develop strategies targeting both cancer metabolism
AMP-activated protein kinase connects energy sensing to mitophagy, Science 331
and autophagy to avoid adaptation and resistance mechanisms. (6016) (2011) 456–461.
[21] J. Kim, M. Kundu, B. Viollet, K.L. Guan, AMPK and mTOR regulate autophagy
Conflict of interests through direct phosphorylation of Ulk1, Nat. Cell Biol. 13 (2) (2011) 132–141.
[22] K.H. Kim, M.S. Lee, Autophagy–a key player in cellular and body metabolism, Nat.
Rev. Endocrinol. 10 (6) (2014) 322–337.
Authors declare no conflict of interest. [23] N. Mizushima, M. Komatsu, Autophagy: renovation of cells and tissues, Cell 147
(4) (2011) 728–741.
[24] S. Rao, L. Tortola, T. Perlot, G. Wirnsberger, M. Novatchkova, R. Nitsch,
Acknowledgements P. Sykacek, L. Frank, D. Schramek, V. Komnenovic, V. Sigl, K. Aumayr,
G. Schmauss, N. Fellner, S. Handschuh, M. Glosmann, P. Pasierbek, M. Schlederer,
We thank all our colleagues from the Université de Tours-Inserm G.P. Resch, Y. Ma, H. Yang, H. Popper, L. Kenner, G. Kroemer, J.M. Penninger, A
dual role for autophagy in a murine model of lung cancer, Nat. Commun. 5 (2014)
UMR1069 for all constructive discussion and Ms. Catherine Le Roy for 3056.
secretary and administrative assistance. The work performed at the [25] E. White, The role for autophagy in cancer, J. Clin. Invest. 125 (1) (2015) 42–46.
Université de Tours-Inserm UMR1069 was supported by the Inserm, the [26] Z. Yue, S. Jin, C. Yang, A.J. Levine, N. Heintz, Beclin 1, an autophagy gene es-
sential for early embryonic development, is a haploinsufficient tumor suppressor,
"Ligue Nationale Contre le Cancer – Inter-région Grand-Ouest", the
Proc. Natl. Acad. Sci. U. S. A. 100 (25) (2003) 15077–15082.
Fondation ARC and the “Cancéropole Grand-Ouest”. [27] X. Qu, J. Yu, G. Bhagat, N. Furuya, H. Hibshoosh, A. Troxel, J. Rosen,
Finally, we wish to apologize to all researchers whose relevant E.L. Eskelinen, N. Mizushima, Y. Ohsumi, G. Cattoretti, B. Levine, Promotion of
tumorigenesis by heterozygous disruption of the beclin 1 autophagy gene, J. Clin.
works, owing to the limited length of this article format, could not be
Invest. 112 (12) (2003) 1809–1820.
cited in this review. [28] L. Brisson, P. Banski, M. Sboarina, C. Dethier, P. Danhier, M.J. Fontenille, V.F. Van
Hee, T. Vazeille, M. Tardy, J. Falces, C. Bouzin, P.E. Porporato, R. Frederick,
References C. Michiels, T. Copetti, P. Sonveaux, Lactate dehydrogenase B controls lysosome
activity and autophagy in Cancer, Cancer Cell 30 (3) (2016) 418–431.
[29] I. Papandreou, A.L. Lim, K. Laderoute, N.C. Denko, Hypoxia signals autophagy in
[1] O. Warburg, On the origin of cancer cells, Science 123 (3191) (1956) 309–314. tumor cells via AMPK activity, independent of HIF-1, BNIP3, and BNIP3L, Cell
[2] R. Moreno-Sanchez, S. Rodriguez-Enriquez, A. Marin-Hernandez, E. Saavedra, Death Differ. 15 (10) (2008) 1572–1581.
Energy metabolism in tumor cells, FEBS J. 274 (6) (2007) 1393–1418. [30] L. Poillet-Perez, G. Despouy, R. Delage-Mourroux, M. Boyer-Guittaut, Interplay
[3] R.J. DeBerardinis, T. Cheng, Q’s next: the diverse functions of glutamine in me- between ROS and autophagy in cancer cells, from tumor initiation to cancer
tabolism, cell biology and cancer, Oncogene 29 (3) (2010) 313–324. therapy, Redox Biol. 4 (2015) 184–192.
[4] P. Sonveaux, F. Vegran, T. Schroeder, M.C. Wergin, J. Verrax, Z.N. Rabbani, [31] M.C. Maiuri, E. Tasdemir, A. Criollo, E. Morselli, J.M. Vicencio, R. Carnuccio,
C.J. De Saedeleer, K.M. Kennedy, C. Diepart, B.F. Jordan, M.J. Kelley, B. Gallez, G. Kroemer, Control of autophagy by oncogenes and tumor suppressor genes, Cell
M.L. Wahl, O. Feron, M.W. Dewhirst, Targeting lactate-fueled respiration selec- Death Differ. 16 (1) (2009) 87–93.
tively kills hypoxic tumor cells in mice, J. Clin. Invest. 118 (12) (2008) [32] A.M. Strohecker, J.Y. Guo, G. Karsli-Uzunbas, S.M. Price, G.J. Chen, R. Mathew,
3930–3942. M. McMahon, E. White, Autophagy sustains mitochondrial glutamine metabolism
[5] K.M. Nieman, H.A. Kenny, C.V. Penicka, A. Ladanyi, R. Buell-Gutbrod, and growth of BrafV600E-driven lung tumors, Cancer Discov. 3 (11) (2013)
M.R. Zillhardt, I.L. Romero, M.S. Carey, G.B. Mills, G.S. Hotamisligil, S.D. Yamada, 1272–1285.
M.E. Peter, K. Gwin, E. Lengyel, Adipocytes promote ovarian cancer metastasis [33] J.Y. Guo, X. Teng, S.V. Laddha, S. Ma, S.C. Van Nostrand, Y. Yang, S. Khor,
and provide energy for rapid tumor growth, Nat. Med. 17 (11) (2011) 1498–1503. C.S. Chan, J.D. Rabinowitz, E. White, Autophagy provides metabolic substrates to
[6] M.G. Vander Heiden, L.C. Cantley, C.B. Thompson, Understanding the Warburg maintain energy charge and nucleotide pools in Ras-driven lung cancer cells,
effect: the metabolic requirements of cell proliferation, Science 324 (5930) (2009) Genes Dev. 30 (15) (2016) 1704–1717.
1029–1033. [34] H. Maes, N. Rubio, A.D. Garg, P. Agostinis, Autophagy: shaping the tumor mi-
[7] A.M. Hosios, V.C. Hecht, L.V. Danai, M.O. Johnson, J.C. Rathmell, croenvironment and therapeutic response, Trends Mol. Med. 19 (7) (2013)
M.L. Steinhauser, S.R. Manalis, M.G. Vander Heiden, Amino acids rather than 428–446.
glucose account for the majority of cell mass in proliferating mammalian cells, [35] E.E. Mowers, M.N. Sharifi, K.F. Macleod, Functions of autophagy in the tumor
Dev. Cell 36 (5) (2016) 540–549. microenvironment and cancer metastasis, FEBS J. 285 (10) (2018) 1751–1766.
[8] C.C. Hsu, L.M. Tseng, H.C. Lee, Role of mitochondrial dysfunction in cancer pro- [36] R. Lazova, R.L. Camp, V. Klump, S.F. Siddiqui, R.K. Amaravadi, J.M. Pawelek,
gression, Exp. Biol. Med. (Maywood) 241 (12) (2016) 1281–1295. Punctate LC3B expression is a common feature of solid tumors and associated with

7
F. Ferro, et al. Seminars in Cell and Developmental Biology xxx (xxxx) xxx–xxx

proliferation, metastasis, and poor outcome, Clin. Cancer Res. 18 (2) (2012) biogenesis, Sci. Rep. 7 (2017) 45076.
370–379. [62] M. Graef, J. Nunnari, Mitochondria regulate autophagy by conserved signalling
[37] A. Giatromanolaki, E. Sivridis, S. Mendrinos, A.V. Koutsopoulos, M.I. Koukourakis, pathways, EMBO J. 30 (11) (2011) 2101–2114.
Autophagy proteins in prostate cancer: relation with anaerobic metabolism and [63] H.E. Thomas, Y. Zhang, J.A. Stefely, S.R. Veiga, G. Thomas, S.C. Kozma,
Gleason score, Urol. Oncol. 32 (1) (2014) p. 39 e11-8. C.A. Mercer, Mitochondrial complex I activity is required for maximal autophagy,
[38] H. Zhao, M. Yang, J. Zhao, J. Wang, Y. Zhang, Q. Zhang, High expression of LC3B Cell Rep. 24 (9) (2018) 2404–2417 e8.
is associated with progression and poor outcome in triple-negative breast cancer, [64] R.V. Duran, E.D. MacKenzie, H. Boulahbel, C. Frezza, L. Heiserich, S. Tardito,
Med. Oncol. 30 (1) (2013) 475. O. Bussolati, S. Rocha, M.N. Hall, E. Gottlieb, HIF-independent role of prolyl hy-
[39] E.E. Mowers, M.N. Sharifi, K.F. Macleod, Autophagy in cancer metastasis, droxylases in the cellular response to amino acids, Oncogene 32 (38) (2013)
Oncogene (2016). 4549–4556.
[40] M.N. Sharifi, E.E. Mowers, L.E. Drake, C. Collier, H. Chen, M. Zamora, S. Mui, [65] R.V. Duran, W. Oppliger, A.M. Robitaille, L. Heiserich, R. Skendaj, E. Gottlieb,
K.F. Macleod, Autophagy promotes focal adhesion disassembly and cell motility of M.N. Hall, Glutaminolysis activates Rag-mTORC1 signaling, Mol. Cell 47 (3)
metastatic tumor cells through the direct interaction of Paxillin with LC3, Cell Rep. (2012) 349–358.
15 (8) (2016) 1660–1672. [66] S. Lorin, M.J. Tol, C. Bauvy, A. Strijland, C. Pous, A.J. Verhoeven, P. Codogno,
[41] R. Lock, C.M. Kenific, A.M. Leidal, E. Salas, J. Debnath, Autophagy-dependent A.J. Meijer, Glutamate dehydrogenase contributes to leucine sensing in the reg-
production of secreted factors facilitates oncogenic RAS-driven invasion, Cancer ulation of autophagy, Autophagy 9 (6) (2013) 850–860.
Discov. 4 (4) (2014) 466–479. [67] V.H. Villar, T.L. Nguyen, V. Delcroix, S. Teres, M. Bouchecareilh, B. Salin,
[42] T.G. Keulers, M.B. Schaaf, K.M. Rouschop, Autophagy-dependent secretion: con- C. Bodineau, P. Vacher, M. Priault, P. Soubeyran, R.V. Duran, mTORC1 inhibition
tribution to tumor progression, Front. Oncol. 6 (2016) 251. in cancer cells protects from glutaminolysis-mediated apoptosis during nutrient
[43] Y.F. Peng, Y.H. Shi, Y.H. Shen, Z.B. Ding, A.W. Ke, J. Zhou, S.J. Qiu, J. Fan, limitation, Nat. Commun. 8 (2017) 14124.
Promoting colonization in metastatic HCC cells by modulation of autophagy, PLoS [68] C.H. Eng, K. Yu, J. Lucas, E. White, R.T. Abraham, Ammonia derived from glu-
One 8 (9) (2013) p. e74407. taminolysis is a diffusible regulator of autophagy, Sci. Signal. 3 (119) (2010) ra31.
[44] C. Fung, R. Lock, S. Gao, E. Salas, J. Debnath, Induction of autophagy during [69] L.M. Harder, J. Bunkenborg, J.S. Andersen, Inducing autophagy: a comparative
extracellular matrix detachment promotes cell survival, Mol. Biol. Cell 19 (3) phosphoproteomic study of the cellular response to ammonia and rapamycin,
(2008) 797–806. Autophagy 10 (2) (2014) 339–355.
[45] M. Gugnoni, V. Sancisi, G. Manzotti, G. Gandolfi, A. Ciarrocchi, Autophagy and [70] G. Marino, G. Kroemer, Ammonia: a diffusible factor released by proliferating cells
epithelial-mesenchymal transition: an intricate interplay in cancer, Cell Death Dis. that induces autophagy, Sci. Signal. 3 (124) (2010) pe19.
7 (12) (2016) e2520. [71] Y.H. Ko, Z. Lin, N. Flomenberg, R.G. Pestell, A. Howell, F. Sotgia, M.P. Lisanti,
[46] X. Sui, R. Chen, Z. Wang, Z. Huang, N. Kong, M. Zhang, W. Han, F. Lou, J. Yang, U.E. Martinez-Outschoorn, Glutamine fuels a vicious cycle of autophagy in the
Q. Zhang, X. Wang, C. He, H. Pan, Autophagy and chemotherapy resistance: a tumor stroma and oxidative mitochondrial metabolism in epithelial cancer cells:
promising therapeutic target for cancer treatment, Cell Death Dis. 4 (2013) e838. implications for preventing chemotherapy resistance, Cancer Biol. Ther. 12 (12)
[47] A.V. Knizhnik, W.P. Roos, T. Nikolova, S. Quiros, K.H. Tomaszowski, (2011) 1085–1097.
M. Christmann, B. Kaina, Survival and death strategies in glioma cells: autophagy, [72] D. Whitaker-Menezes, U.E. Martinez-Outschoorn, Z. Lin, A. Ertel, N. Flomenberg,
senescence and apoptosis triggered by a single type of temozolomide-induced DNA A.K. Witkiewicz, R.C. Birbe, A. Howell, S. Pavlides, R. Gandara, R.G. Pestell,
damage, PLoS One 8 (1) (2013) p. e55665. F. Sotgia, N.J. Philp, M.P. Lisanti, Evidence for a stromal-epithelial "lactate shuttle"
[48] R.K. Amaravadi, D. Yu, J.J. Lum, T. Bui, M.A. Christophorou, G.I. Evan, in human tumors: MCT4 is a marker of oxidative stress in cancer-associated fi-
A. Thomas-Tikhonenko, C.B. Thompson, Autophagy inhibition enhances therapy- broblasts, Cell Cycle 10 (11) (2011) 1772–1783.
induced apoptosis in a Myc-induced model of lymphoma, J. Clin. Invest. 117 (2) [73] Y.J. Chen, N.G. Mahieu, X. Huang, M. Singh, P.A. Crawford, S.L. Johnson,
(2007) 326–336. R.W. Gross, J. Schaefer, G.J. Patti, Lactate metabolism is associated with mam-
[49] W. Guo, Y. Wang, Z. Wang, Y.P. Wang, H. Zheng, Inhibiting autophagy increases malian mitochondria, Nat. Chem. Biol. 12 (11) (2016) 937–943.
epirubicin’s cytotoxicity in breast cancer cells, Cancer Sci. 107 (11) (2016) [74] C. Corbet, O. Feron, Emerging roles of lipid metabolism in cancer progression,
1610–1621. Curr. Opin. Clin. Nutr. Metab. Care 20 (4) (2017) 254–260.
[50] C. Bellodi, M.R. Lidonnici, A. Hamilton, G.V. Helgason, A.R. Soliera, M. Ronchetti, [75] I.R. Schlaepfer, L. Rider, L.U. Rodrigues, M.A. Gijon, C.T. Pac, L. Romero,
S. Galavotti, K.W. Young, T. Selmi, R. Yacobi, R.A. Van Etten, N. Donato, A. Cimic, S.J. Sirintrapun, L.M. Glode, R.H. Eckel, S.D. Cramer, Lipid catabolism
A. Hunter, D. Dinsdale, E. Tirro, P. Vigneri, P. Nicotera, M.J. Dyer, T. Holyoake, via CPT1 as a therapeutic target for prostate cancer, Mol. Cancer Ther. 13 (10)
P. Salomoni, B. Calabretta, Targeting autophagy potentiates tyrosine kinase in- (2014) 2361–2371.
hibitor-induced cell death in Philadelphia chromosome-positive cells, including [76] Y.A. Wen, X. Xing, J.W. Harris, Y.Y. Zaytseva, M.I. Mitov, D.L. Napier, H.L. Weiss,
primary CML stem cells, J. Clin. Invest. 119 (5) (2009) 1109–1123. B. Mark Evers, T. Gao, Adipocytes activate mitochondrial fatty acid oxidation and
[51] M. Selvakumaran, R.K. Amaravadi, I.A. Vasilevskaya, P.J. O’Dwyer, Autophagy autophagy to promote tumor growth in colon cancer, Cell Death Dis. 8 (2) (2017)
inhibition sensitizes colon cancer cells to antiangiogenic and cytotoxic therapy, e2593.
Clin. Cancer Res. 19 (11) (2013) 2995–3007. [77] Y. Tabe, S. Yamamoto, K. Saitoh, K. Sekihara, N. Monma, K. Ikeo, K. Mogushi,
[52] Y. Chen, X. Li, L. Guo, X. Wu, C. He, S. Zhang, Y. Xiao, Y. Yang, D. Hao, Combining M. Shikami, V. Ruvolo, J. Ishizawa, N. Hail Jr., S. Kazuno, M. Igarashi,
radiation with autophagy inhibition enhances suppression of tumor growth and H. Matsushita, Y. Yamanaka, H. Arai, I. Nagaoka, T. Miida, Y. Hayashizaki,
angiogenesis in esophageal cancer, Mol. Med. Rep. 12 (2) (2015) 1645–1652. M. Konopleva, M. Andreeff, Bone marrow adipocytes facilitate fatty acid oxidation
[53] A.V. Onorati, M. Dyczynski, R. Ojha, R.K. Amaravadi, Targeting autophagy in activating AMPK and a transcriptional network supporting survival of acute
cancer, Cancer 124 (16) (2018) 3307–3318. monocytic leukemia cells, Cancer Res. 77 (6) (2017) 1453–1464.
[54] H. Maes, A. Kuchnio, A. Peric, S. Moens, K. Nys, K. De Bock, A. Quaegebeur, [78] A.S. Rambold, S. Cohen, J. Lippincott-Schwartz, Fatty acid trafficking in starved
S. Schoors, M. Georgiadou, J. Wouters, S. Vinckier, H. Vankelecom, M. Garmyn, cells: regulation by lipid droplet lipolysis, autophagy, and mitochondrial fusion
A.C. Vion, F. Radtke, C. Boulanger, H. Gerhardt, E. Dejana, M. Dewerchin, dynamics, Dev. Cell 32 (6) (2015) 678–692.
B. Ghesquiere, W. Annaert, P. Agostinis, P. Carmeliet, Tumor vessel normalization [79] A. Alexander, S.L. Cai, J. Kim, A. Nanez, M. Sahin, K.H. MacLean, K. Inoki,
by chloroquine independent of autophagy, Cancer Cell 26 (2) (2014) 190–206. K.L. Guan, J. Shen, M.D. Person, D. Kusewitt, G.B. Mills, M.B. Kastan, C.L. Walker,
[55] P. Maycotte, S. Aryal, C.T. Cummings, J. Thorburn, M.J. Morgan, A. Thorburn, ATM signals to TSC2 in the cytoplasm to regulate mTORC1 in response to ROS,
Chloroquine sensitizes breast cancer cells to chemotherapy independent of au- Proc. Natl. Acad. Sci. U. S. A. 107 (9) (2010) 4153–4158.
tophagy, Autophagy 8 (2) (2012) 200–212. [80] C. Song, S.K. Mitter, X. Qi, E. Beli, H.V. Rao, J. Ding, C.S. Ip, H. Gu, D. Akin,
[56] M. Mauthe, I. Orhon, C. Rocchi, X. Zhou, M. Luhr, K.J. Hijlkema, R.P. Coppes, W.A. Dunn Jr., C. Bowes Rickman, A.S. Lewin, M.B. Grant, M.E. Boulton,
N. Engedal, M. Mari, F. Reggiori, Chloroquine inhibits autophagic flux by de- Oxidative stress-mediated NFkappaB phosphorylation upregulates p62/SQSTM1
creasing autophagosome-lysosome fusion, Autophagy 14 (8) (2018) 1435–1455. and promotes retinal pigmented epithelial cell survival through increased autop-
[57] P. Pellegrini, A. Strambi, C. Zipoli, M. Hagg-Olofsson, M. Buoncervello, S. Linder, hagy, PLoS One 12 (2) (2017) p. e0171940.
A. De Milito, Acidic extracellular pH neutralizes the autophagy-inhibiting activity [81] R. Scherz-Shouval, E. Shvets, E. Fass, H. Shorer, L. Gil, Z. Elazar, Reactive oxygen
of chloroquine: implications for cancer therapies, Autophagy 10 (4) (2014) species are essential for autophagy and specifically regulate the activity of Atg4,
562–571. EMBO J. 26 (7) (2007) 1749–1760.
[58] Y. Chen, E. McMillan-Ward, J. Kong, S.J. Israels, S.B. Gibson, Mitochondrial [82] X. Zhang, X. Cheng, L. Yu, J. Yang, R. Calvo, S. Patnaik, X. Hu, Q. Gao, M. Yang,
electron-transport-chain inhibitors of complexes I and II induce autophagic cell M. Lawas, M. Delling, J. Marugan, M. Ferrer, H. Xu, MCOLN1 is a ROS sensor in
death mediated by reactive oxygen species, J. Cell. Sci. 120 (Pt 23) (2007) lysosomes that regulates autophagy, Nat. Commun. 7 (2016) 12109.
4155–4166. [83] Q. Wang, L. Huang, J. Yue, Oxidative stress activates the TRPM2-Ca(2+)-CaMKII-
[59] B. Zhao, L. Qiang, J. Joseph, B. Kalyanaraman, B. Viollet, Y.Y. He, Mitochondrial ROS signaling loop to induce cell death in cancer cells, Biochim Biophys Acta Mol
dysfunction activates the AMPK signaling and autophagy to promote cell survival, Cell Res 1864 (6) (2017) 957–967.
Genes Dis. 3 (1) (2016) 82–87. [84] Y. Chen, M.B. Azad, S.B. Gibson, Superoxide is the major reactive oxygen species
[60] C. Settembre, R. De Cegli, G. Mansueto, P.K. Saha, F. Vetrini, O. Visvikis, regulating autophagy, Cell Death Differ. 16 (7) (2009) 1040–1052.
T. Huynh, A. Carissimo, D. Palmer, T.J. Klisch, A.C. Wollenberg, D. Di Bernardo, [85] A.E. Li, H. Ito, I.I. Rovira, K.S. Kim, K. Takeda, Z.Y. Yu, V.J. Ferrans, T. Finkel, A
L. Chan, J.E. Irazoqui, A. Ballabio, TFEB controls cellular lipid metabolism role for reactive oxygen species in endothelial cell anoikis, Circ. Res. 85 (4) (1999)
through a starvation-induced autoregulatory loop, Nat. Cell Biol. 15 (6) (2013) 304–310.
647–658. [86] Z.T. Schafer, A.R. Grassian, L. Song, Z. Jiang, Z. Gerhart-Hines, H.Y. Irie, S. Gao,
[61] L. Fernandez-Mosquera, C.V. Diogo, K.F. Yambire, G.L. Santos, M. Luna Sanchez, P. Puigserver, J.S. Brugge, Antioxidant and oncogene rescue of metabolic defects
P. Benit, P. Rustin, L.C. Lopez, I. Milosevic, N. Raimundo, Acute and chronic caused by loss of matrix attachment, Nature 461 (7260) (2009) 109–113.
mitochondrial respiratory chain deficiency differentially regulate lysosomal [87] A. Avivar-Valderas, E. Salas, E. Bobrovnikova-Marjon, J.A. Diehl, C. Nagi,

8
F. Ferro, et al. Seminars in Cell and Developmental Biology xxx (xxxx) xxx–xxx

J. Debnath, J.A. Aguirre-Ghiso, PERK integrates autophagy and oxidative stress [114] D. Ivankovic, K.Y. Chau, A.H. Schapira, M.E. Gegg, Mitochondrial and lysosomal
responses to promote survival during extracellular matrix detachment, Mol. Cell. biogenesis are activated following PINK1/parkin-mediated mitophagy, J.
Biol. 31 (17) (2011) 3616–3629. Neurochem. 136 (2) (2016) 388–402.
[88] T. Ozben, Oxidative stress and apoptosis: impact on cancer therapy, J. Pharm. Sci. [115] C.T. Chu, J. Ji, R.K. Dagda, J.F. Jiang, Y.Y. Tyurina, A.A. Kapralov, V.A. Tyurin,
96 (9) (2007) 2181–2196. N. Yanamala, I.H. Shrivastava, D. Mohammadyani, K.Z.Q. Wang, J. Zhu, J. Klein-
[89] C. Amantini, M.B. Morelli, M. Nabissi, C. Cardinali, M. Santoni, A. Gismondi, Seetharaman, K. Balasubramanian, A.A. Amoscato, G. Borisenko, Z. Huang,
G. Santoni, Capsaicin triggers autophagic cell survival which drives epithelial A.M. Gusdon, A. Cheikhi, E.K. Steer, R. Wang, C. Baty, S. Watkins, I. Bahar,
mesenchymal transition and chemoresistance in bladder cancer cells in an H. Bayir, V.E. Kagan, Cardiolipin externalization to the outer mitochondrial
Hedgehog-dependent manner, Oncotarget 7 (31) (2016) 50180–50194. membrane acts as an elimination signal for mitophagy in neuronal cells, Nat. Cell
[90] N. Chen, L. Wu, H. Yuan, J. Wang, ROS/Autophagy/Nrf2 pathway mediated low- Biol. 15 (10) (2013) 1197–1205.
dose radiation induced radio-resistance in human lung adenocarcinoma A549 cell, [116] V.E. Kagan, J. Jiang, Z. Huang, Y.Y. Tyurina, C. Desbourdes, C. Cottet-Rousselle,
Int. J. Biol. Sci. 11 (7) (2015) 833–844. H.H. Dar, M. Verma, V.A. Tyurin, A.A. Kapralov, A. Cheikhi, G. Mao, D. Stolz,
[91] D. Naon, L. Scorrano, At the right distance: ER-mitochondria juxtaposition in cell C.M. St Croix, S. Watkins, Z. Shen, Y. Li, M.L. Greenberg, M. Tokarska-Schlattner,
life and death, Biochim. Biophys. Acta 1843 (10) (2014) 2184–2194. M. Boissan, M.L. Lacombe, R.M. Epand, C.T. Chu, R.K. Mallampalli, H. Bayir,
[92] D.W. Hailey, A.S. Rambold, P. Satpute-Krishnan, K. Mitra, R. Sougrat, P.K. Kim, U. Schlattner, NDPK-D (NM23-H4)-mediated externalization of cardiolipin enables
J. Lippincott-Schwartz, Mitochondria supply membranes for autophagosome bio- elimination of depolarized mitochondria by mitophagy, Cell Death Differ. 23 (7)
genesis during starvation, Cell 141 (4) (2010) 656–667. (2016) 1140–1151.
[93] Y. Kabeya, N. Mizushima, A. Yamamoto, S. Oshitani-Okamoto, Y. Ohsumi, [117] Y. Lee, H.Y. Lee, R.A. Hanna, A.B. Gustafsson, Mitochondrial autophagy by Bnip3
T. Yoshimori, LC3, GABARAP and GATE16 localize to autophagosomal membrane involves Drp1-mediated mitochondrial fission and recruitment of Parkin in cardiac
depending on form-II formation, J. Cell. Sci. 117 (Pt 13) (2004) 2805–2812. myocytes, Am. J. Physiol. Heart Circ. Physiol. 301 (5) (2011) p. H1924-31.
[94] C. Cardenas, R.A. Miller, I. Smith, T. Bui, J. Molgo, M. Muller, H. Vais, [118] L. Buhlman, M. Damiano, G. Bertolin, R. Ferrando-Miguel, A. Lombes, A. Brice,
K.H. Cheung, J. Yang, I. Parker, C.B. Thompson, M.J. Birnbaum, K.R. Hallows, O. Corti, Functional interplay between Parkin and Drp1 in mitochondrial fission
J.K. Foskett, Essential regulation of cell bioenergetics by constitutive InsP3 re- and clearance, Biochim. Biophys. Acta 1843 (9) (2014) 2012–2026.
ceptor Ca2+ transfer to mitochondria, Cell 142 (2) (2010) 270–283. [119] W. Wu, C. Lin, K. Wu, L. Jiang, X. Wang, W. Li, H. Zhuang, X. Zhang, H. Chen,
[95] G. Bidaux, D. Gordienko, G. Shapovalov, V. Farfariello, A.S. Borowiec, S. Li, Y. Yang, Y. Lu, J. Wang, R. Zhu, L. Zhang, S. Sui, N. Tan, B. Zhao, J. Zhang,
O. Iamshanova, L. Lemonnier, M. Gueguinou, R. Guibon, G. Fromont, M. Paillard, L. Li, D. Feng, FUNDC1 regulates mitochondrial dynamics at the ER-mitochondrial
Y. Gouriou, C. Chouabe, E. Dewailly, D. Gkika, P. Lopez-Alvarado, J. Carlos contact site under hypoxic conditions, EMBO J. 35 (13) (2016) 1368–1384.
Menendez, L. Heliot, C. Slomianny, N. Prevarskaya, 4TM-TRPM8 channels are new [120] V. Gelmetti, P. De Rosa, L. Torosantucci, E.S. Marini, A. Romagnoli, M. Di Rienzo,
gatekeepers of the ER-mitochondria Ca(2+) transfer, Biochim Biophys Acta Mol G. Arena, D. Vignone, G.M. Fimia, E.M. Valente, PINK1 and BECN1 relocalize at
Cell Res 1865 (7) (2018) 981–994. mitochondria-associated membranes during mitophagy and promote ER-mi-
[96] B. O’Rourke, Mitochondrial ion channels, Annu. Rev. Physiol. 69 (2007) 19–49. tochondria tethering and autophagosome formation, Autophagy 13 (4) (2017)
[97] I. Szabo, M. Zoratti, Mitochondrial channels: ion fluxes and more, Physiol. Rev. 94 654–669.
(2) (2014) 519–608. [121] M. Fujiwara, H. Marusawa, H.Q. Wang, A. Iwai, K. Ikeuchi, Y. Imai, A. Kataoka,
[98] C. Cardenas, M. Muller, A. McNeal, A. Lovy, F. Jana, G. Bustos, F. Urra, N. Smith, N. Nukina, R. Takahashi, T. Chiba, Parkin as a tumor suppressor gene for hepa-
J. Molgo, J.A. Diehl, T.W. Ridky, J.K. Foskett, Selective vulnerability of Cancer tocellular carcinoma, Oncogene 27 (46) (2008) 6002–6011.
cells by inhibition of Ca(2+) transfer from endoplasmic reticulum to mitochon- [122] C. Li, Y. Zhang, X. Cheng, H. Yuan, S. Zhu, J. Liu, Q. Wen, Y. Xie, J. Liu,
dria, Cell Rep. 14 (10) (2016) 2313–2324. G. Kroemer, D.J. Klionsky, M.T. Lotze, H.J. Zeh, R. Kang, D. Tang, PINK1 and
[99] P. Gomez-Suaga, S. Paillusson, R. Stoica, W. Noble, D.P. Hanger, C.C.J. Miller, The PARK2 suppress pancreatic tumorigenesis through control of mitochondrial iron-
ER-Mitochondria tethering complex VAPB-PTPIP51 regulates autophagy, Curr. mediated immunometabolism, Dev. Cell 46 (4) (2018) 441–455 e8.
Biol. 27 (3) (2017) 371–385. [123] G. Poulogiannis, R.E. McIntyre, M. Dimitriadi, J.R. Apps, C.H. Wilson, K. Ichimura,
[100] M.S. Herrera-Cruz, T. Simmen, Cancer: Untethering Mitochondria from the F. Luo, L.C. Cantley, A.H. Wyllie, D.J. Adams, M.J. Arends, PARK2 deletions occur
Endoplasmic Reticulum? Front. Oncol. 7 (2017) 105. frequently in sporadic colorectal cancer and accelerate adenoma development in
[101] S.M. Jin, M. Lazarou, C. Wang, L.A. Kane, D.P. Narendra, R.J. Youle, Apc mutant mice, Proc. Natl. Acad. Sci. U. S. A. 107 (34) (2010) 15145–15150.
Mitochondrial membrane potential regulates PINK1 import and proteolytic de- [124] S. Veeriah, B.S. Taylor, S. Meng, F. Fang, E. Yilmaz, I. Vivanco, M. Janakiraman,
stabilization by PARL, J. Cell Biol. 191 (5) (2010) 933–942. N. Schultz, A.J. Hanrahan, W. Pao, M. Ladanyi, C. Sander, A. Heguy, E.C. Holland,
[102] D.P. Narendra, S.M. Jin, A. Tanaka, D.F. Suen, C.A. Gautier, J. Shen, P.B. Paty, P.S. Mischel, L. Liau, T.F. Cloughesy, I.K. Mellinghoff, D.B. Solit,
M.R. Cookson, R.J. Youle, PINK1 is selectively stabilized on impaired mitochon- T.A. Chan, Somatic mutations of the Parkinson’s disease-associated gene PARK2 in
dria to activate Parkin, PLoS Biol. 8 (1) (2010) e1000298. glioblastoma and other human malignancies, Nat. Genet. 42 (1) (2010) 77–82.
[103] S.A. Sarraf, M. Raman, V. Guarani-Pereira, M.E. Sowa, E.L. Huttlin, S.P. Gygi, [125] H.H. Hu, C. Kannengiesser, S. Lesage, J. Andre, S. Mourah, L. Michel, V. Descamps,
J.W. Harper, Landscape of the PARKIN-dependent ubiquitylome in response to N. Basset-Seguin, M. Bagot, A. Bensussan, C. Lebbe, L. Deschamps, P. Saiag,
mitochondrial depolarization, Nature 496 (7445) (2013) 372–376. M.T. Leccia, B. Bressac-de-Paillerets, A. Tsalamlal, R. Kumar, S. Klebe,
[104] S. Geisler, K.M. Holmstrom, D. Skujat, F.C. Fiesel, O.C. Rothfuss, P.J. Kahle, B. Grandchamp, N. Andrieu-Abadie, L. Thomas, A. Brice, N. Dumaz, N. Soufir,
W. Springer, PINK1/Parkin-mediated mitophagy is dependent on VDAC1 and p62/ PARKIN inactivation links parkinson’s disease to melanoma, J. Natl. Cancer Inst.
SQSTM1, Nat. Cell Biol. 12 (2) (2010) 119–131. 108 (3) (2016).
[105] J.Y. Lee, Y. Nagano, J.P. Taylor, K.L. Lim, T.P. Yao, Disease-causing mutations in [126] S. Lee, J. She, B. Deng, J. Kim, M. de Andrade, J. Na, Z. Sun, J.A. Wampfler,
parkin impair mitochondrial ubiquitination, aggregation, and HDAC6-dependent J.M. Cunningham, Y. Wu, A.H. Limper, M.C. Aubry, C. Wendt, P. Biterman,
mitophagy, J. Cell Biol. 189 (4) (2010) 671–679. P. Yang, Z. Lou, Multiple-level validation identifies PARK2 in the development of
[106] E. Hollville, R.G. Carroll, S.P. Cullen, S.J. Martin, Bcl-2 family proteins participate lung cancer and chronic obstructive pulmonary disease, Oncotarget 7 (28) (2016)
in mitochondrial quality control by regulating Parkin/PINK1-dependent mito- 44211–44223.
phagy, Mol. Cell 55 (3) (2014) 451–466. [127] A. Letessier, S. Garrido-Urbani, C. Ginestier, G. Fournier, B. Esterni, F. Monville,
[107] M. Lazarou, D.A. Sliter, L.A. Kane, S.A. Sarraf, C. Wang, J.L. Burman, D.P. Sideris, J. Adelaide, J. Geneix, L. Xerri, P. Dubreuil, P. Viens, E. Charafe-Jauffret,
A.I. Fogel, R.J. Youle, The ubiquitin kinase PINK1 recruits autophagy receptors to J. Jacquemier, D. Birnbaum, M. Lopez, M. Chaffanet, Correlated break at PARK2/
induce mitophagy, Nature 524 (7565) (2015) 309–314. FRA6E and loss of AF-6/Afadin protein expression are associated with poor out-
[108] B. Richter, D.A. Sliter, L. Herhaus, A. Stolz, C. Wang, P. Beli, G. Zaffagnini, P. Wild, come in breast cancer, Oncogene 26 (2) (2007) 298–307.
S. Martens, S.A. Wagner, R.J. Youle, I. Dikic, Phosphorylation of OPTN by TBK1 [128] R. Cesari, E.S. Martin, G.A. Calin, F. Pentimalli, R. Bichi, H. McAdams, F. Trapasso,
enhances its binding to Ub chains and promotes selective autophagy of damaged A. Drusco, M. Shimizu, V. Masciullo, G. D’Andrilli, G. Scambia, M.C. Picchio,
mitochondria, Proc. Natl. Acad. Sci. U. S. A. 113 (15) (2016) 4039–4044. H. Alder, A.K. Godwin, C.M. Croce, Parkin, a gene implicated in autosomal re-
[109] L.E. Drake, M.Z. Springer, L.P. Poole, C.J. Kim, K.F. Macleod, Expanding per- cessive juvenile parkinsonism, is a candidate tumor suppressor gene on chromo-
spectives on the significance of mitophagy in cancer, Semin. Cancer Biol. 47 some 6q25-q27, Proc. Natl. Acad. Sci. U. S. A. 100 (10) (2003) 5956–5961.
(2017) 110–124. [129] C. Zhang, M. Lin, R. Wu, X. Wang, B. Yang, A.J. Levine, W. Hu, Z. Feng, Parkin, a
[110] G. Bellot, R. Garcia-Medina, P. Gounon, J. Chiche, D. Roux, J. Pouyssegur, p53 target gene, mediates the role of p53 in glucose metabolism and the Warburg
N.M. Mazure, Hypoxia-induced autophagy is mediated through hypoxia-inducible effect, Proc. Natl. Acad. Sci. U. S. A. 108 (39) (2011) 16259–16264.
factor induction of BNIP3 and BNIP3L via their BH3 domains, Mol. Cell. Biol. 29 [130] J. Liu, C. Zhang, Y. Zhao, X. Yue, H. Wu, S. Huang, J. Chen, K. Tomsky, H. Xie,
(10) (2009) 2570–2581. C.A. Khella, M.L. Gatza, D. Xia, J. Gao, E. White, B.G. Haffty, W. Hu, Z. Feng,
[111] H.M. Sowter, P.J. Ratcliffe, P. Watson, A.H. Greenberg, A.L. Harris, HIF-1-de- Parkin targets HIF-1alpha for ubiquitination and degradation to inhibit breast
pendent regulation of hypoxic induction of the cell death factors BNIP3 and NIX in tumor progression, Nat. Commun. 8 (1) (2017) 1823.
human tumors, Cancer Res. 61 (18) (2001) 6669–6673. [131] M. Akada, T. Crnogorac-Jurcevic, S. Lattimore, P. Mahon, R. Lopes, M. Sunamura,
[112] L. Liu, D. Feng, G. Chen, M. Chen, Q. Zheng, P. Song, Q. Ma, C. Zhu, R. Wang, S. Matsuno, N.R. Lemoine, Intrinsic chemoresistance to gemcitabine is associated
W. Qi, L. Huang, P. Xue, B. Li, X. Wang, H. Jin, J. Wang, F. Yang, P. Liu, Y. Zhu, with decreased expression of BNIP3 in pancreatic cancer, Clin. Cancer Res. 11 (8)
S. Sui, Q. Chen, Mitochondrial outer-membrane protein FUNDC1 mediates hy- (2005) 3094–3101.
poxia-induced mitophagy in mammalian cells, Nat. Cell Biol. 14 (2) (2012) [132] M. Erkan, J. Kleeff, I. Esposito, T. Giese, K. Ketterer, M.W. Buchler, N.A. Giese,
177–185. H. Friess, Loss of BNIP3 expression is a late event in pancreatic cancer contributing
[113] S. Melser, E.H. Chatelain, J. Lavie, W. Mahfouf, C. Jose, E. Obre, S. Goorden, to chemoresistance and worsened prognosis, Oncogene 24 (27) (2005)
M. Priault, Y. Elgersma, H.R. Rezvani, R. Rossignol, G. Benard, Rheb regulates 4421–4432.
mitophagy induced by mitochondrial energetic status, Cell Metab. 17 (5) (2013) [133] A.H. Chourasia, K. Tracy, C. Frankenberger, M.L. Boland, M.N. Sharifi, L.E. Drake,
719–730. J.R. Sachleben, J.M. Asara, J.W. Locasale, G.S. Karczmar, K.F. Macleod,

9
F. Ferro, et al. Seminars in Cell and Developmental Biology xxx (xxxx) xxx–xxx

Mitophagy defects arising from BNip3 loss promote mammary tumor progression [153] M. Laskowski, B. Augustynek, B. Kulawiak, P. Koprowski, P. Bednarczyk,
to metastasis, EMBO Rep. 16 (9) (2015) 1145–1163. W. Jarmuszkiewicz, A. Szewczyk, What do we not know about mitochondrial
[134] W. Li, Y. Li, S. Siraj, H. Jin, Y. Fan, X. Yang, X. Huang, X. Wang, J. Wang, L. Liu, potassium channels? Biochim. Biophys. Acta 1857 (8) (2016) 1247–1257.
L. Du, Q. Chen, FUN14 domain-containing 1-Mediated mitophagy suppresses he- [154] M.S. Kane, A. Paris, P. Codron, J. Cassereau, V. Procaccio, G. Lenaers, P. Reynier,
patocarcinogenesis by inhibition of inflammasome activation in mice, Hepatology A. Chevrollier, Current mechanistic insights into the CCCP-induced cell survival
(2018). response, Biochem. Pharmacol. 148 (2018) 100–110.
[135] M. Vara-Perez, H. Maes, S. Van Dingenen, P. Agostinis, BNIP3 contributes to the [155] Y. Sun, A.A. Vashisht, J. Tchieu, J.A. Wohlschlegel, L. Dreier, Voltage-dependent
glutamine-driven aggressive behavior of melanoma cells, Biol. Chem. (2018). anion channels (VDACs) recruit Parkin to defective mitochondria to promote
[136] L. Hui, H. Wu, T.W. Wang, N. Yang, X. Guo, X.J. Jang, Hydrogen peroxide-induced mitochondrial autophagy, J. Biol. Chem. 287 (48) (2012) 40652–40660.
mitophagy contributes to laryngeal cancer cells survival via the upregulation of [156] T.D. MacVicar, L.V. Mannack, R.M. Lees, J.D. Lane, Targeted siRNA screens
FUNDC1, Clin. Transl. Oncol. (2018). identify ER-to-Mitochondrial calcium exchange in Autophagy and mitophagy re-
[137] Y.S. Lee, Y.Y. Jung, M.H. Park, I.J. Yeo, H.S. Im, K.T. Nam, H.D. Kim, S.K. Kang, sponses in RPE1 cells, Int. J. Mol. Sci. 16 (6) (2015) 13356–13380.
J.K. Song, Y.R. Kim, D.Y. Choi, P.H. Park, S.B. Han, J.S. Yun, J.T. Hong, Deficiency [157] N.M. Mazure, VDAC in cancer, Biochim Biophys Acta Bioenerg 1858 (8) (2017)
of parkin suppresses melanoma tumor development and metastasis through in- 665–673.
hibition of MFN2 ubiquitination, Cancer Lett. 433 (2018) 156–164. [158] V. Shoshan-Barmatz, Y. Krelin, A. Shteinfer-Kuzmine, T. Arif, Voltage-dependent
[138] E. Villa, E. Proics, C. Rubio-Patino, S. Obba, B. Zunino, J.P. Bossowski, anion channel 1 As an emerging drug target for novel anti-cancer therapeutics,
R.M. Rozier, J. Chiche, L. Mondragon, J.S. Riley, S. Marchetti, E. Verhoeyen, Front. Oncol. 7 (2017) 154.
S.W.G. Tait, J.E. Ricci, Parkin-independent mitophagy controls chemotherapeutic [159] T. Arif, L. Vasilkovsky, Y. Refaely, A. Konson, V. Shoshan-Barmatz, Silencing
response in Cancer cells, Cell Rep. 20 (12) (2017) 2846–2859. VDAC1 expression by siRNA inhibits Cancer cell proliferation and tumor growth in
[139] H. Maes, S. Van Eygen, D.V. Krysko, P. Vandenabeele, K. Nys, K. Rillaerts, vivo, Mol. Ther. Nucleic Acids 3 (2014) e159.
A.D. Garg, T. Verfaillie, P. Agostinis, BNIP3 supports melanoma cell migration and [160] A. Vultur, C.S. Gibhardt, H. Stanisz, I. Bogeski, The role of the mitochondrial
vasculogenic mimicry by orchestrating the actin cytoskeleton, Cell Death Dis. 5 calcium uniporter (MCU) complex in cancer, Pflugers Arch. 470 (8) (2018)
(2014) e1127. 1149–1163.
[140] L. Sun, T. Li, Q. Wei, Y. Zhang, X. Jia, Z. Wan, L. Han, Upregulation of BNIP3 [161] S. Tang, X. Wang, Q. Shen, X. Yang, C. Yu, C. Cai, G. Cai, X. Meng, F. Zou,
mediated by ERK/HIF-1alpha pathway induces autophagy and contributes to an- Mitochondrial Ca(2)(+) uniporter is critical for store-operated Ca(2)(+) entry-
oikis resistance of hepatocellular carcinoma cells, Future Oncol. 10 (8) (2014) dependent breast cancer cell migration, Biochem. Biophys. Res. Commun. 458 (1)
1387–1398. (2015) 186–193.
[141] H. Wu, S. Huang, Z. Chen, W. Liu, X. Zhou, D. Zhang, Hypoxia-induced autophagy [162] C. Yu, Y. Wang, J. Peng, Q. Shen, M. Chen, W. Tang, X. Li, C. Cai, B. Wang, S. Cai,
contributes to the invasion of salivary adenoid cystic carcinoma through the HIF- X. Meng, F. Zou, Mitochondrial calcium uniporter as a target of microRNA-340
1alpha/BNIP3 signaling pathway, Mol. Med. Rep. 12 (5) (2015) 6467–6474. and promoter of metastasis via enhancing the Warburg effect, Oncotarget 8 (48)
[142] C. Yan, L. Luo, C.Y. Guo, S. Goto, Y. Urata, J.H. Shao, T.S. Li, Doxorubicin-induced (2017) 83831–83844.
mitophagy contributes to drug resistance in cancer stem cells from HCT8 human [163] T. Ren, H. Zhang, J. Wang, J. Zhu, M. Jin, Y. Wu, X. Guo, L. Ji, Q. Huang,
colorectal cancer cells, Cancer Lett. 388 (2017) 34–42. H. Zhang, H. Yang, J. Xing, MCU-dependent mitochondrial Ca(2+) inhibits NAD
[143] H. Hou, P. Er, J. Cheng, X. Chen, X. Ding, Y. Wang, X. Chen, Z. Yuan, Q. Pang, (+)/SIRT3/SOD2 pathway to promote ROS production and metastasis of HCC
P. Wang, D. Qian, High expression of FUNDC1 predicts poor prognostic outcomes cells, Oncogene 36 (42) (2017) 5897–5909.
and is a promising target to improve chemoradiotherapy effects in patients with [164] K. Mallilankaraman, C. Cardenas, P.J. Doonan, H.C. Chandramoorthy,
cervical cancer, Cancer Med. 6 (8) (2017) 1871–1881. K.M. Irrinki, T. Golenar, G. Csordas, P. Madireddi, J. Yang, M. Muller, R. Miller,
[144] E.A. Koop, T. van Laar, D.F. van Wichen, R.A. de Weger, E. Wall, P.J. van Diest, J.E. Kolesar, J. Molgo, B. Kaufman, G. Hajnoczky, J.K. Foskett, M. Madesh,
Expression of BNIP3 in invasive breast cancer: correlations with the hypoxic re- MCUR1 is an essential component of mitochondrial Ca2+ uptake that regulates
sponse and clinicopathological features, BMC Cancer 9 (2009) 175. cellular metabolism, Nat. Cell Biol. 14 (12) (2012) 1336–1343.
[145] H.M. Sowter, M. Ferguson, C. Pym, P. Watson, S.B. Fox, C. Han, A.L. Harris, [165] D. Tomar, Z. Dong, S. Shanmughapriya, D.A. Koch, T. Thomas, N.E. Hoffman,
Expression of the cell death genes BNip3 and NIX in ductal carcinoma in situ of the S.A. Timbalia, S.J. Goldman, S.L. Breves, D.P. Corbally, N. Nemani,
breast; correlation of BNip3 levels with necrosis and grade, J. Pathol. 201 (4) J.P. Fairweather, A.R. Cutri, X. Zhang, J. Song, F. Jana, J. Huang, C. Barrero,
(2003) 573–580. J.E. Rabinowitz, T.S. Luongo, S.M. Schumacher, M.E. Rockman, A. Dietrich,
[146] Z. Jiang, F. Yu, M. Li, Upregulation of BCL2 19 kD protein-interacting protein 3 S. Merali, J. Caplan, P. Stathopulos, R.S. Ahima, J.Y. Cheung, S.R. Houser,
(BNIP3) is predictive of unfavorable prognosis in Uveal Melanoma, Med. Sci. W.J. Koch, V. Patel, V.M. Gohil, J.W. Elrod, S. Rajan, M. Madesh, MCUR1 is a
Monit. 24 (2018) 4711–4717. scaffold factor for the MCU complex function and promotes mitochondrial bioe-
[147] H. Gang, R. Dhingra, J. Lin, Y. Hai, Y. Aviv, V. Margulets, M. Hamedani, nergetics, Cell Rep. 15 (8) (2016) 1673–1685.
T. Thanasupawat, E. Leygue, T. Klonisch, J.R. Davie, L.A. Kirshenbaum, PDK2- [166] T. Ren, J. Wang, H. Zhang, P. Yuan, J. Zhu, Y. Wu, Q. Huang, X. Guo, J. Zhang,
mediated alternative splicing switches Bnip3 from cell death to cell survival, J. L. Ji, J. Li, H. Zhang, H. Yang, J. Xing, MCUR1-mediated mitochondrial calcium
Cell Biol. 210 (7) (2015) 1101–1115. signaling facilitates cell survival of hepatocellular carcinoma via reactive oxygen
[148] E. Gazzano, L. Lazzarato, B. Rolando, J. Kopecka, S. Guglielmo, C. Costamagna, species-dependent P53 degradation, Antioxid. Redox Signal. 28 (12) (2018)
K. Chegaev, C. Riganti, Mitochondrial Delivery of Phenol Substructure Triggers 1120–1136.
Mitochondrial Depolarization and Apoptosis of Cancer Cells, Front. Pharmacol. 9 [167] S.J. Chen, N.E. Hoffman, S. Shanmughapriya, L. Bao, K. Keefer, K. Conrad,
(2018) 580. S. Merali, Y. Takahashi, T. Abraham, I. Hirschler-Laszkiewicz, J. Wang,
[149] T.G. Biel, V.A. Rao, Mitochondrial dysfunction activates lysosomal-dependent X.Q. Zhang, J. Song, C. Barrero, Y. Shi, Y.I. Kawasawa, M. Bayerl, T. Sun,
mitophagy selectively in cancer cells, Oncotarget 9 (1) (2018) 995–1011. M. Barbour, H.G. Wang, M. Madesh, J.Y. Cheung, B.A. Miller, A splice variant of
[150] S. Feng, H. Li, Y. Tai, J. Huang, Y. Su, J. Abramowitz, M.X. Zhu, L. Birnbaumer, the human ion channel TRPM2 modulates neuroblastoma tumor growth through
Y. Wang, Canonical transient receptor potential 3 channels regulate mitochondrial hypoxia-inducible factor (HIF)-1/2alpha, J. Biol. Chem. 289 (52) (2014)
calcium uptake, Proc. Natl. Acad. Sci. U. S. A. 110 (27) (2013) 11011–11016. 36284–36302.
[151] B.A. Miller, N.E. Hoffman, S. Merali, X.Q. Zhang, J. Wang, S. Rajan, [168] S. Almasi, B.E. Kennedy, M. El-Aghil, A.M. Sterea, S. Gujar, S. Partida-Sanchez,
S. Shanmughapriya, E. Gao, C.A. Barrero, K. Mallilankaraman, J. Song, T. Gu, Y. El Hiani, TRPM2 channel-mediated regulation of autophagy maintains mi-
I. Hirschler-Laszkiewicz, W.J. Koch, A.M. Feldman, M. Madesh, J.Y. Cheung, tochondrial function and promotes gastric cancer cell survival via the JNK-sig-
TRPM2 channels protect against cardiac ischemia-reperfusion injury: role of mi- naling pathway, J. Biol. Chem. 293 (10) (2018) 3637–3650.
tochondria, J. Biol. Chem. 289 (11) (2014) 7615–7629. [169] D.W. Koh, D.P. Powell, S.D. Blake, J.L. Hoffman, M.M. Hopkins, X. Feng, Enhanced
[152] E.J. Griffiths, G.A. Rutter, Mitochondrial calcium as a key regulator of mi- cytotoxicity in triple-negative and estrogen receptorpositive breast adenocarci-
tochondrial ATP production in mammalian cells, Biochim. Biophys. Acta 1787 noma cells due to inhibition of the transient receptor potential melastatin-2
(11) (2009) 1324–1333. channel, Oncol. Rep. 34 (3) (2015) 1589–1598.

10

You might also like