You are on page 1of 16

698391

research-article2017
TUB0010.1177/1010428317698391Tumor Biology X(X)Maycotte et al.

Review
Tumor Biology

Mitochondrial dynamics and cancer May 2017: 1­–16 


© The Author(s) 2017
Reprints and permissions:
sagepub.co.uk/journalsPermissions.nav
DOI: 10.1177/1010428317698391
https://doi.org/10.1177/1010428317698391
journals.sagepub.com/home/tub

Paola Maycotte1,2, Alvaro Marín-Hernández3,


Miriam Goyri-Aguirre2, Maricruz Anaya-Ruiz2, Julio Reyes-Leyva2
and Paulina Cortés-Hernández2

Abstract
Cancer is among the leading causes of death worldwide, and the number of new cases continues to rise. Despite recent
advances in diagnosis and therapeutic strategies, millions of cancer-related deaths occur, indicating the need for better
therapies and diagnostic strategies. Mitochondria and metabolic alterations have been recognized as important for
cancer progression. However, a more precise understanding of how to manipulate mitochondria-related processes for
cancer therapy remains to be established. Mitochondria are highly dynamic organelles which continually fuse and divide
in response to diverse stimuli. Participation in the aforementioned processes requires a precise regulation at many levels
that allows the cell to couple mitochondrial activity to nutrient availability, biosynthetic demands, proliferation rates,
and external stimuli. The many functions of these organelles are intimately linked to their morphology. Recent evidence
suggests an important link between mitochondrial morphology and disease, including neurodegenerative, inflammatory
diseases and cancer. Here, we review recent advances in the understanding of mitochondrial dynamics with a special
focus on its relationship to tumor progression.

Keywords
Mitochondria, cancer, mitochondrial fusion, mitochondrial fission

Date received: 28 September 2016; accepted: 23 December 2016

Introduction
Mitochondria are often considered the cell’s powerhouse, stressors. Despite a clear understanding of the importance of
not only due to their essential role in the production of mitochondrial dynamics, its precise role in the clinical out-
adenosine triphosphate (ATP) through oxidative phospho- come of cancer patients remains to be established. In this
rylation (OXPHOS) but also because of their crucial role regard, increasing evidence suggests a potential utility of
in other biochemical pathways such as pyrimidine and diverse mitochondria-related parameters for cancer diagno-
purine biosynthesis, heme synthesis, the regulation of sis and treatment, but the role of mitochondria in cancer
nitrogen balance in the urea cycle, gluconeogenesis, ketone development or progression is not simple. Here, we describe
body production, and lipid degradation and elongation.1,2
Emerging evidence has also demonstrated that mitochon-
dria play an important role in the regulation of cell signal- 1CONACYT-Centro de Investigación Biomédica de Oriente, Instituto
ing, either serving as a scaffold for protein–protein Mexicano del Seguro Social, Puebla, México
2Laboratorio de Biología Celular, Centro de Investigación Biomédica de
interactions or by regulating the levels of intracellular
messengers such as Ca2+ or reactive oxygen species (ROS). Oriente, Instituto Mexicano del Seguro Social, Puebla, México
3Departamento de Bioquímica, Instituto Nacional de Cardiología
Our understanding of mitochondrial dynamics, biogene- Ignacio Chávez, México, México
sis, and degradation has dramatically increased in recent
years and evidence has shown that alterations in these pro- Corresponding author:
Paola Maycotte, CONACYT-Centro de Investigación Biomédica de
cesses are linked to diverse pathological states including Oriente, Instituto Mexicano del Seguro Social, Km 4.5 Carretera
cancer. Mitochondrial integrity is fundamental for survival Atlixco-Metepec, 74360 Puebla, México.
in response to energetic, environmental, and genotoxic Email: pmaycottego@conacyt.mx

Creative Commons Non Commercial CC BY-NC: This article is distributed under the terms of the Creative Commons
Attribution-NonCommercial 4.0 License (http://www.creativecommons.org/licenses/by-nc/4.0/) which permits non-commercial
use, reproduction and distribution of the work without further permission provided the original work is attributed as specified on the SAGE and
Open Access pages (https://us.sagepub.com/en-us/nam/open-access-at-sage).
2 Tumor Biology

counteracts metabolic insults, preserves cellular integrity,


and protects against autophagy.6 Thus, balanced fusion and
fission events shape mitochondria to meet cellular meta-
bolic demands and to regulate the removal of damaged
organelles. Besides being involved in the maintenance of
normal cell functions as the distribution of mitochondria
during cell growth, cell division, and differentiation, mito-
chondrial dynamics has recently been considered as a cor-
nerstone for the maintenance of cellular fitness since
alterations in fusion and fission events have been closely
linked to states of health and disease (Figure 1).5,6
The molecular machinery regulating mitochondrial
dynamics comprises large dynamin-like guanosine triphos-
phatases (GTPases) mediating the fusion and fission of
both outer mitochondrial membrane (OMM) and inner
membrane (IMM; Figure 2). OMM fusion is regulated by
Figure 1.  Mitochondrial morphology and cellular processes mitofusins 1 and 2 (MFN1 and MFN2), which are required
that have been related to them. Mitochondrial dynamics
for the maintenance of a reticular mitochondrial network
are the result of a delicate balance between mitochondrial
fusion (characterized by the presence of long interconnected in cells.7 Mitofusins function through homo- and hetero-
tubules) and fission (characterized by small, fragmented typic interactions, and their synthesis is regulated by tran-
mitochondria). Diverse cellular functions have been associated scriptional and post-transcriptional mechanisms (Figure
with mitochondrial morphology, as shown in the figure and 2).6 The transcriptional mitochondrial coactivators phos-
reviewed in the text. phatidylglycerol phospholipase C-1-alpha and beta (PGC-
1-α and β), which regulate mitochondrial biogenesis and
the current knowledge of the processes regulating mito- the expression of the respiratory complex machinery, have
chondrial shape and function and review recent studies on also been involved in the regulation of mitochondrial
their role in tumorigenesis and cancer progression, focusing dynamics through control of gene expression.8,9 In this
on the potential use of mitochondrial biomarkers for cancer regard, treatment of mice with resveratrol, a known activa-
detection and prognosis as well as on the possibility of tar- tor of the protein deacetylase sirtuin 1 (SIRT1), decreased
geting mitochondrial dynamics for cancer therapy. PGC-1α acetylation and increased its activity. These
effects led to the induction of mitochondrial biogenesis
and increased mitochondrial size in skeletal muscle, con-
Mitochondrial fusion and fission
ferring metabolic advantages like increased aerobic capac-
Our view of mitochondrial dynamics has dramatically ity in vivo.6 Also, PGC-1-β has been proposed to induce
changed in the past years. Once considered to be a rigid mitochondrial fusion by increasing the transcription of
structure, mitochondria are now known to be organelles Mfn2 through estrogen-related receptor alpha (ERRα)
that migrate throughout the cell, fuse, divide, and undergo coactivation.9
regulated turnover through mitochondrial autophagy or Proteasome-mediated degradation of mitofusins is reg-
mitophagy. Mitochondria continually divide and fuse, ulated by ubiquitylation and phosphorylation in response
even in resting cells where the mitochondrial network is to cellular stimuli.6 In general, mitofusins are degraded
formed by a mixture of long and interconnected tubules.3–5 during apoptosis, while decreased proteasomal degrada-
In some cells, they fuse together in a connected network, tion induces mitochondrial fusion and cell viability
whereas in other cells or under different circumstances, (Figure 2). Deletion of either mitofusins or abrogation of
mitochondria convert into a large number of small frag- GTPase activity prevents mitochondrial fusion, inducing
ments. Fibroblast mitochondria, for example, are usually the fragmentation of mitochondria in cultured cells.
long filaments, whereas hepatocyte mitochondria are more Moreover, tissue-specific deletion of mitofusins in the
uniformly spheres or ovoids.3 The many functions of mito- central nervous system, heart, muscle, or liver compro-
chondria are intimately linked to their morphology, and mises metabolic and organ function, highlighting their
there is a delicate balance between the fusion and fission importance at both the cellular and organismal levels.6
events, which allows proper mitochondrial function in the Mitochondrial phospholipids have also been shown to be
cell. important for mitochondrial membrane fusion. It has been
Unopposed fission causes extensive mitochondrial suggested that local alterations of the phospholipid composi-
fragmentation, which has been generally associated with tion on the OMM may induce membrane curvature or desta-
cellular metabolic dysfunction and disease. However, con- bilize membrane bilayers to promote fusion or fission.6,17 In
tinuous fusion results in a hyperfused network, which this regard, phosphatidic acid (PA), a phospholipid that can
Maycotte et al. 3

Figure 2.  Molecular machinery involved in the shaping of mitochondria. Mitochondrial shape is regulated by a family of GTPases
with structural homologies to dynamin proteins. Mitofusins are the GTPases required for OMM fusion and they are regulated by
ubiquitylation and phosphorylation. The E3-ubiquitin ligase PARKIN marks both MFN1 and MFN2 with ubiquitin (ub) regulating
their activity and levels. However, the deubiquitylase USP30 antagonizes PARKIN activity promoting mitochondrial fusion.
PINK1 is a kinase able to phosphorylate ubiquitin chains on OMM proteins including MFN2 which can then recruit PARKIN.
Phosphorylation of MFN2 by JNK in response to cellular stress signals the recruitment of the E3-ubiquitin ligase HUWE1, triggering
MFN2 degradation by the proteasome. Also, miR-140, which can be induced by oxidative or genotoxic insults, negatively regulates
MFN1 translation. The transcriptional coactivator PGC-1-β has been proposed to regulate mitochondrial fusion via Mfn2 and
other mitochondrial genes like OPA1. MitoPLD is a member of the phospholipase D family which is bound to the outer membrane
and converts cardiolipin to phosphatidic acid. This catalytic activity is required for mitochondrial fusion. In mammalian cells, the
major player of mitochondrial fission is DRP1 which has a cytoplasmic localization (inactive form) and reversibly associates with
OMM at sites of mitochondrial division, where the fission occurs preferentially at ER-contact regions. DRP1 is subject to several
post-translational modifications in response to cellular cues. Phosphorylation of DRP1 at Ser637 represents the signal that controls
protein translocation from the mitochondria to the cytoplasm. Following mitochondrial depolarization, a rise in cytosolic Ca2+
activates cytosolic calcineurin (CaN) that dephosphorylates DRP1 on Ser637, inducing its translocation to mitochondria. CaN itself
is subject to translational regulation by miR-499 in a p53-dependent manner. Conversely, protein kinase A (PKA), after cyclic
AMP activation, phosphorylates this residue, leading to the elongation of mitochondria and resistance to various pro-apoptotic
insults. DRP1 localization to mitochondria is a process requiring adaptors on the OMM (FIS1, MFF, MiD49, MiD51, and Mcl-1L).
The mitochondrial ubiquitin ligase MITOL/MARCH5 promotes mitochondrial division in a DRP1- and MiD49-dependent manner.
MITOL/MARCH5 also targets MFN1 and FIS1. The mitochondrial anchored protein ligase (MAPL) induces SUMOylation of DRP1
targeting it to mitochondria and accelerating mitochondrial fission. SUMOylation can be reversed by the SUMO protease SENP5.
During mitosis, CDK1/cyclin B phosphorylates DRP1 at Ser616 to induce mitochondrial fission and proper organelle segregation.
ERK2 can also phosphorylate DRP1 at Ser616 during MAPK-mediated transformation. Enforced expression of MTP18 correlates
with the recruitment of DRP1 to mitochondria. DRP1 can also be S-nitrosylated at Cys644 in response to an increase in NO
inducing mitochondrial fission. Finally, an interaction of DRP1 with phosphatidic acid (PA) restrains DRP1 fission activity and
shifts the balance toward mitochondrial fusion. OPA1 is involved in inner membrane processing. Alternative splicing of OPA1
gives rise to long forms (L-OPA) that are proteolytically cleaved to short forms (S-OPA) by inner membrane peptidases, OMA1
and YME1L. While L-OPA1 induces mitochondrial fusion and maintains tubular mitochondria, excessive accumulation of S-OPA1
can either induce mitochondrial fusion or accelerate fission and promote mitochondrial fragmentation. The IMM peptidase
YME1L constitutively cleaves OPA1 at the S2 site inducing mitochondrial fusion. However, OMA1 cleaves OPA1 at the S1 site
constitutively or in response to stress, inducing mitochondrial fragmentation. The expression of OPA1 is under transcriptional
control of the NF-κB transcription factor, which governs diverse aspects of cell division and metabolic reprogramming. Post-
translational modifications—P: phosphorylation; ub: ubiquitination; SUMO: sumoylation; PA: phosphatidic acid.6,7,10–16

generate negative curvature of lipid membranes, has an phosphatase decreases PA levels and induces mitochondrial
important role in mitochondrial fusion. Phosphatidic acid– fragmentation.17 However, mitoPLD, a mitochondrion-
preferring phospholipase A1 (PA-PLA1) or Lipin 1 localized phospholipase D able to produce PA from
4 Tumor Biology

cardiolipin, facilitates MFN-mediated fusion and ablation of do survive in culture, some of their mitochondria display a
mitoPLD induced mitochondrial fragmentation in vitro.6 reduced mtDNA copy number and decreased membrane
Inner membrane fusion is mediated by optic atrophy 1 potential, causing problems with ATP synthesis.6 Therefore,
(OPA1), another dynamin-like GTPase anchored to the although an organism is unable to develop normally without
IMM and mostly exposed to the intermembrane space mitochondrial fusion, some cells can survive without this
(IMS).6 Multiple forms of OPA1 have been described, process. These differential requirements of mitochondrial
which are the result of alternative splicing and proteolytic fusion for survival may stem from the distinctive need to
cleavage by mitochondrial proteases that target OPA1 upon distribute mitochondria in large cells,19 from higher demands
dissipation of membrane potential, mitochondrial DNA on oxidative metabolism in different cell types and develop-
(mtDNA) loss, or induction of apoptosis.4 Alternative splic- mental stages, or from demands on other functions that are
ing of OPA1 produces long isoforms (L-OPA1) that are indirectly affected by fusion.3 In this regard, cancer cells
cleaved to short forms (S-OPA1) with different functions on have been proposed to harbor altered mitochondrial metab-
mitochondrial morphology (Figure 2).10,11 The expression of olism, a phenomenon related to the increased glycolytic
OPA1 is under transcriptional control of the nuclear factor- activity in cancer cells known as the Warburg effect. It will
κB (NF-κB) transcription factor, which governs diverse thus be interesting to understand how dependent are cancer
aspects of cell division and metabolic reprogramming.18 cells on mitochondrial fusion and fission and whether this is
The inverse process of mitochondrial fragmentation or particular to one type of cancer.
fission has been observed in response to nutrient excess Mitochondrial shape is controlled by changes in metab-
and cellular dysfunction in cardiovascular and neuromus- olism. In general, mitochondria fuse when they are forced
cular disorders, obesity, and cancer. In normal cells, mito- to rely on oxidative phosphorylation by withdrawing glu-
chondrial fission facilitates the autophagic clearance of cose as a carbon source, thus maximizing oxidative phos-
mitochondria (mitophagy) and allows the adaption of phorylation by stimulating complementation among
mitochondrial activities to physiological demands.6 OMM mitochondria. Fusion is also enhanced by treatments that
fission is mediated by a cytosolic dynamin family member inhibit protein synthesis, by starvation, and by autophagy
dynamin-related protein 1 (DRP1) that translocates from induction. Starvation-induced autophagy may enhance
the cytosol and binds its receptors on the OMM (fission fusion by increasing reliance on oxidative phosphorylation
protein 1 (FIS1), mitochondrial fission factor (MFF), through the metabolism of lipids and proteins.3 More spe-
mitochondrial dynamics of 49 kDa protein (MiD49), and cifically, autophagy induced by starvation or mammalian
mitochondrial dynamics of 51 kDa protein (MiD51)) often target of rapamycin (mTOR) inhibition induces increased
at sites where mitochondria make contact with the endo- cyclic adenosine monophosphate (cAMP) and protein
plasmic reticulum.6 DRP1 forms spirals around mitochon- kinase A (PKA) activation, which phosphorylates DRP1 (at
dria, and GTP binding and hydrolysis promote a Ser637) and increases its cytoplasmic localization, leading
conformational change in DRP1 that results in membrane to unopposed mitochondrial fusion.20 Then, elongated
constriction to sever both IMM and OMM.3,6 DRP1 is mitochondria are spared from autophagic degradation
regulated post-translationally at different levels including (mitophagy). Conversely, when elongation is blocked dur-
protein phosphorylation, sumoylation, ubiquitination, and ing starvation, mitochondria consume ATP provoking star-
S-nitrosylation (Figure 2).12 vation-induced cell death.20 Mitochondrial fusion is
Mitochondrial fission is essential for growing and divid- required for mtDNA maintenance, probably because it
ing cells to populate them with adequate numbers of mito- allows metabolite and mtDNA exchange between mito-
chondria. Although it is less evident why mitochondrial chondria. Thus, mitochondrial fragmentation due to
fission and fusion are also needed for non-proliferating impaired fusion is often accompanied by bioenergetic
cells, it is probably due to fusion-induced complementation defects due to a loss of mtDNA.21 In agreement with the
between damaged mitochondria. This allows sharing of previous observations linking mitochondrial elongation to
RNA, protein components, and lipids with other mitochon- cellular survival, stress stimuli leading to protein synthesis
dria and mitigation of the effects of mtDNA mutations inhibition or serum and amino acid deprivation has been
caused by environmental damage. While mitochondrial shown to produce stress-induced mitochondrial hyperfu-
fusion would prevent mitochondrial damage through com- sion (SIMH), defined as a highly interconnected network
plementation, the lack of fusion would favor mitochondrial that induces increased mitochondrial ATP production and
degradation through autophagy.3 Thus, non-proliferating has been proposed to work as an adaptive pro-survival
neurons cannot survive without mitochondrial fission, and response against stress.21 Importantly, this mechanism was
two human diseases, dominant optic atrophy and Charcot– independent of MFN2, BCL-2-associated X protein (BAX)/
Marie–Tooth disease type 2A, are caused by fusion defects.4 BCL-2 homologous antagonist/killer (BAK), and prohib-
Mfn1 and Mfn2 knock-out mice die in utero, and although itins but required L-OPA1, MFN1, and the IMM protein
mouse embryo fibroblasts (MEFs) derived from these mice stomatin-like protein 2 (SLP-2).21
Maycotte et al. 5

Cancer cell metabolism total cellular ATP supply has been estimated to range
between 10% and 70% depending on the cell type,26 indi-
In cancer, cellular metabolism may be subverted to support cating that some tumor cell lines have high levels of oxi-
tumor growth. For instance, glycolysis is upregulated in dative phosphorylation and that this pathway might be the
almost all cancer cells since it is needed for the synthesis predominant energy source. Regarding oncogene-medi-
of macromolecules that are required for active prolifera- ated cell transformation, most of the research in cancer
tion.22,23 This characteristic, known as the Warburg effect, cell metabolism has focused on the increased glycolysis in
has found clinical utility for imaging of tumors using cancer cells. However, recent evidence indicates that
18F-fluorodeoxyglucose (18F-FDG) which accumulates some tumor suppressor genes and oncogenes also have an
in highly glycolytic cancer cells and can be visualized by important role in the modulation of mitochondrial func-
positron emission tomography.23 Moreover, this upregula- tion to support carcinogenesis.
tion in glycolysis is currently being explored for cancer
therapeutics utilizing the non-metabolizable glucose Metabolic effects of oncogenes and
analog 2-deoxyglucose to block glycolysis and cancer
tumor suppressors
growth.24 Warburg assumed that “aerobic glycolysis” was
a universal property of malignant cells and suggested that Glycolysis is stimulated in hypoxic regions of the tumor
cancer is caused by impaired mitochondrial metabolism. where oxygen levels are low. This occurs by the induction
However, further studies have challenged this idea, and of hypoxia-inducible factor 1 (HIF-1), which is considered
evidence supporting the idea that dysfunctional mitochon- a master regulator for the initiation and maintenance of the
dria is the main cause of the Warburg effect is limited.25 Warburg effect at the level of gene expression of glycoly-
So, although there indeed seems to be a diminished oxida- sis-related enzymes and glucose transporters.22,23,28 HIF-1
tive metabolism in many tumor types, it has been shown induction can be triggered by the accumulation of ROS
that tumor mitochondria do respire and produce ATP. produced by dysfunctional mitochondria or by Krebs cycle
Evidence thus suggests that there is no oxidative phospho- metabolites like succinate that can inhibit HIF-1α prolyl
rylation to glycolysis switch but rather an upregulation of hydroxylases in the cytosol, leading to the stabilization
glycolysis that has also been observed not only in cancer and activation of HIF-1α.22,23 Succinate accumulation in
cells but in rapidly proliferating normal cells due to the mitochondria can be caused by mutations in succinate
temporary higher demands in metabolic material and dehydrogenase, which has been documented in a familial
energy for completing the cell proliferation process.23,26 predisposition to benign tumors.22 Interestingly, HIF-1
Evidence in support of a decreased oxidative metabo- levels are high in certain tumors even under oxygen-rich
lism in cancer cells has been explained in several ways for conditions, indicating that hormones or growth factors
different tumor types, including decreased pyruvate trans- might cause stabilization of HIF-1 expression and that it
porter activity, lower pyruvate oxidation, truncated Krebs has important roles in tumorigenesis.22,23 Additionally,
cycle, reduced transfer of reducing equivalents to mito- HIF-1 itself can also function as a negative regulator of
chondria, lower mitochondrial content, or defective res- mitochondrial biogenesis and oxidative phosphorylation.
piratory chain.26 Perhaps the most documented cause of a For example, HIF-1 activation upregulates several mito-
partial inhibition of oxidative phosphorylation in tumor chondrial proteins including pyruvate dehydrogenase
cells is the Crabtree effect or the inhibition by high con- kinase 1, which inhibits the conversion of pyruvate to
centrations of glucose and other hexoses. Also, the acidic acetyl-CoA by pyruvate dehydrogenase resulting in
pH induced by lactate generation may affect pH-sensitive decreased mitochondrial respiration.22,29 HIF-1 activation
oxidative enzymes decreasing their function.26 A general also inhibits MYC transcription and activates its degrada-
property of transformed cells with dysfunctional mito- tion by the proteasome pathway. Since MYC is a positive
chondria is increased ROS production.27 Therefore, it has regulator of PGC-1α and PGC-1β, the mitochondrial coac-
been suggested that these cells might show a lower thresh- tivators that regulate mitochondrial biogenesis and the
old of sensitivity to ROS-induced apoptosis. In agreement expression of the respiratory machinery, its repression by
with this idea, therapies able to specifically target the res- HIF-1 results in the inhibition of mitochondrial biogenesis
piratory chain to further elevate ROS production have and oxidative phosphorylation.30,31 Interestingly, the
been proposed to be useful in cancer cells which show human von Hippel–Lindau (VHL) tumor suppressor (a
elevated levels of ROS. Indeed, this has been shown with negative regulator of HIF-1) promoter has a nuclear res-
mitochondrial inhibitors like mitocans, As2O3, antimycin piratory factor 1 (NRF-1, a nuclear transcription factor
A, or rotenone in cancer cells with defective mitochondrial associated with the expression of many genes required for
function.27 expression and function of the respiratory chain) recogni-
However, although there are several examples of tumor tion site. Since NRF-1 is a target for the PGC-1 family
cell lines which certainly exhibit a decreased mitochon- coactivators, these proteins may promote mitochondrial
drial function, that observation does not seem to apply to oxidative metabolism in part through the upregulation of
all tumor cell types. The contribution of glycolysis to the VHL expression.30
6 Tumor Biology

MYC regulates genes involved in glucose metabolism to oxidative metabolism mediated by PGC-1α upregulation
by binding to and regulating virtually all glycolytic enzyme and was overcome by mTOR inhibition which repressed
genes as well as numerous genes involved in mitochon- PGC-1α expression.33 Also, in a mouse model of KRas-
drial function.23 Deregulated MYC expression is a hall- mutant pancreatic ductal adenocarcinoma (PDAC), cells
mark of a large fraction of all human tumors acting as an that survive oncogene ablation showed increased PGC-1α
oncogene in approximately 30% of all human cancers.32 expression and mitochondrial function. Interestingly, the
Concerning mitochondria, MYC regulates the expression reliance on mitochondrial respiration resulted in sensitivity
of over 400 mitochondrial genes, including mitochondrial to oxidative phosphorylation inhibitors.38
transcription factor A (TFAM), an mtDNA transcription Therefore, oncogenes commonly activated in cancer
and maintenance factor, as well as PGC-1α and PGC-1β.33 not only activate glycolysis as Warburg initially proposed
Therefore, gain/loss of MYC increases/reduces mitochon- but can also activate mitochondrial metabolism. Whether
drial mass, respectively. In cancer, oncogenic MYC this mitochondrial activation is particular to certain cells
increases cellular biosynthetic and respiratory capacity by within a tumor (those that are not in hypoxic regions where
upregulating mitochondrial metabolism to support rapid HIF-1 is stimulated or to those cells which are resistant to
cell proliferation and by regulating the expression of pro- therapy) or whether this is a metabolic effect in tumor cells
teins that modulate mitochondrial dynamics.33,34 with certain oncogenic mutations of specific cancer types
Other oncogenes commonly activated in cancers affect or tissues remains to be determined.
metabolism, for example, activation of the phosphoi- Of special interest to cancer cell metabolism is the fact
nositide 3-kinase (PI3K)-Akt-mTOR pathway can increase that p53, a crucial tumor suppressor, long recognized to
cell size, glycolytic activity, metabolism, and cell sur- suppress cancer through the induction of cell-cycle-arrest
vival.22 In this regard, recent data show that mTOR can act or apoptosis programs in response to a plethora of different
as a major regulator of energy production in mitochon- cellular stress signals, has been closely related to tumor cell
dria.35 mTOR regulates mitochondria at multiple levels; metabolism. These functions include the regulation of
transcriptionally, it regulates PGC-1α, ERR-α (a transcrip- metabolism, autophagy, and the oxidative status of the
tion factor downstream of PGC-1α involved in the expres- cell.28 Concerning tumor cell metabolism, p53 can modu-
sion of components of the respiratory chain and control of late the balance between glycolysis and oxidative phospho-
fatty acid oxidation), ERR-α target genes, and the expres- rylation. It can induce the expression of the protein synthesis
sion of nuclear-encoded mitochondrial regulators such as of cytochrome c oxidase 2 (SCO2) that is required for the
TFAM, mitochondrial ribosomal proteins, and compo- assembly of cytochrome c oxidase (COX).22 p53 mutation
nents of complex I and V.35 All these changes result in in tumors causes downregulation of mitochondrial respira-
increased mitochondrial biogenesis and oxidative phos- tion as a result of COX deficiency and a shift of cellular
phorylation. Translationally, mTOR regulates mitochon- metabolism toward glycolysis. Another target of p53 is
drial metabolism and biogenesis via repression of TP53-induced glycolysis and apoptosis regulator (TIGAR),
inhibitory 4E-binding proteins (4E-BPs) that downregu- which decreases fructose-2,6-bisphospate levels in cells,
late translation of nuclear-encoded mitochondrial proteins. resulting in the inhibition of glycolysis, while stimulating
Finally, mTOR inhibits autophagy, a major catabolic pro- nicotinamide adenine dinucleotide phosphate (NADPH)
cess in the cell that serves the cellular function of eliminat- generation through the pentose-phosphate shunt.22 These
ing damaged mitochondria.33,35 studies suggest that p53 is an important regulator of tumor
RAS is an oncogene whose pathway is activated in a cell metabolism and that cells with wild-type p53 and cells
variety of tumors, and its activation has been commonly with mutations or deletions in this gene probably have dif-
linked to alterations in glycolysis. In this regard, RAS ferences in glycolytic and mitochondrial metabolism.
transformation, as well as increased mutant KRas copy Importantly, more than half of human cancers of a wide
number, has been shown to induce metabolic changes in variety of types harbor p53 mutations and even more have
immortalized cells characterized by increased expression alterations in the pathways that regulate p53 stability within
of glycolytic enzymes.36,37 Also, RAS transformation has the cell.28 Importantly, p53 has an important relationship
been shown to increase sensitivity to glucose depletion with HIF-1, and the amplification of HIF-1-dependent
since RAS-transformed cells were unable to upregulate responses to hypoxia via the loss of p53 function contrib-
mitochondrial activity after glucose deprivation due to a utes to the angiogenic switch, promoting cancer progres-
partially uncoupled mitochondrial respiration.36 In contrast, sion.39 Since HIF-1 can downregulate mitochondrial
increased mitochondrial activity has been linked to resist- activity; those tumors with HIF-1 activation and p53 muta-
ance to therapy and recurrence in RAS-transformed can- tions or deletions could be the ones showing a dramatic
cers probably due to upregulation of PGC-1α.33 In BRAF increase in glycolysis as well as an impaired mitochondrial
or NRAS mutant melanomas, resistance to mitogen-acti- metabolism. In other tumors, the series of mutations driv-
vated protein kinase (MAPK) inhibitors (which are acti- ing tumorigenesis could have a specific effect on tumor cell
vated by RAS transformation) was partially due to a switch metabolism.
Maycotte et al. 7

Mitochondrial dynamics and mitochondrial fission and fusion. In this regard, mitochon-
cell death drial fission has been generally associated with apoptotic
cell death. In healthy cells (i.e. in the absence of apoptotic
Apoptosis is triggered in response to various types of stress stimuli), the combined deletion of BAK and BAX results in
that cancer cells experience during tumorigenesis such as reduced fusion. In this setting, non-activated BAX may pro-
high levels of oncogene signaling, DNA damage associated mote fusion by altering MFN2 distribution, mobility, and/or
with hyper proliferation, or anti-cancer therapy. Also, apop- complex assembly.43,45 Also, SIMH, which leads to highly
tosis has been shown to be attenuated in tumors, and a elongated mitochondria, has been shown to delay BAX acti-
resistance to cell death has been listed as one of the hall- vation upon exposure of cells to apoptotic stimuli.43
marks of cancer cells.40 Mitochondria are a focal point of During apoptosis, mitochondrial fission occurs as an
apoptotic signaling. Diverse death stimuli ranging from early event, occurring within the same time frame as acti-
DNA damage to metabolic stress due to immune cell-­ vation of BAX and MOMP. In this regard, diverse interac-
mediated receptor ligation converge on mitochondria to tions have been described for DRP1 and pro-apoptotic
trigger the release of cytochrome c and other death-promot- BCL2 family proteins. In apoptotic cells with activated
ing proteins from the IMS. The release of cytochrome c is BAX (recruited to the mitochondrial membrane), DRP1,
considered a critical decision point in apoptosis and occurs which normally cycles between the cytosol and the mito-
through mitochondrial outer membrane permeabilization chondrial membrane, stably associates with the OMM due
(MOMP), which is regulated by the B-cell lymphoma 2 to BAX/BAK dependent SUMOylation and precedes
(BCL2) protein family members,41 and alterations in the MOMP.46 Also, DRP1 binds and is required for BAX mito-
intrinsic or mitochondrial apoptotic pathway have been chondrial translocation after an apoptotic stimulus.47 The
implicated as a barrier to anti-cancer therapy.40 constricting ability of DRP1 may contribute to the mito-
The BCL2 family of proteins is divided into pro- and chondrial membrane curvature required for the efficient
anti-apoptotic members, and they all share combinations insertion of BAX and mitochondrial cristae remodeling,
of BCL2 homology (BH) domains. BCL2-like proteins suggesting a pro-apoptotic role for DRP1. Interference
(BCL-XL, BCL-W, A1, and MCL-1) are pro-survival and with DRP1 function during apoptosis results in a block of
antagonize the pro-apoptotic members of the family mitochondrial fission, leading to long interconnected orga-
(BAX, BAK, and BCL2 related ovarian killer (BOK)), nelles as well as a delay in cell death.46 In this study, the
which mediate mitochondrial apoptosis through permea- authors suggest that during apoptosis, cycling of DRP1
bilization of the OMM, allowing efflux of apoptogenic between the cytoplasm and the mitochondria mediates
proteins.42 BH3-only proteins (BCL2-like 11 (BIM), mitochondrial fission, whereas BAX/BAK dependent
BCL2 associated agonist of cell death (BAD), BH3- membrane-associated DRP1 is involved in the subsequent
interacting domain death agonist (BID), p53 upregulated apoptotic events, such as the remodeling of the mitochon-
modulator of apoptosis (PUMA), and NOXA) of the drial membrane that leads to MOMP and a loss of mem-
BCL2 family share this BH3 interaction domain and brane potential. Interestingly, DRP1 functions in apoptosis
antagonize BCL2 like proteins. The regulation of interac- could be independent of or complementary to mitochon-
tions between pro-apoptotic and anti-apoptotic members drial fission, since DRP1-induced mitochondrial fragmen-
of the BCL2 family of proteins determines whether a cell tation by itself is not sufficient to induce apoptosis.46,47
lives or dies in response to a stimulus.42 Once in the cyto- It has also been suggested that hyperfragmented mito-
sol, cytochrome c induces the oligomerization of cas- chondria are not involved in apoptosis and that MFN1
pase-9, apoptotic protease activating factor 1 (APAF-1), establishes a mitochondrial size (fragmented, but not
and ATP to form the apoptosome that goes on to activate hyperfragmented) that is permissive for pro-apoptotic
the executioner caspases, caspase-3 and caspase-7. These BCL2 family function.48 In this study, cells with hyper-
terminal caspases cleave various cellular substrates that fragmented mitochondria (less than 0.5 µm) failed to sup-
lead to cellular dismantling through apoptosis.41–43 port BAX-dependent membrane association and
Interestingly, highly glycolytic cells like neurons and can- permeabilization due to an inability to stabilize BAX–
cer cells can be refractory to cytochrome c–induced cas- membrane interactions. Bioenergetics of large and small
pase activation. In these cells, glucose-stimulated mitochondria were similar but only larger mitochondria
production of glutathione as a result of NADPH produc- supported BAX integration to the OMM and mitochon-
tion through the pentose-­phosphate pathway inactivated drial pore formation. The authors suggest that while mito-
cytochrome c by keeping it in its reduced state.41 chondrial hyperfusion can protect from cell death, a
The BCL2 family proteins also have important effects on balanced, non–hyperfragmented mitochondrial network is
metabolism. For instance, phosphorylated BAD can pro- required for BAX-dependent MOMP after terminal ER
mote hepatic glycolysis by activating glucokinase in stress or chemotherapeutic drug treatment. In agreement
response to the fed state.44 Also, proteins from the BCL2 with the previous findings, increased sensitization to cispl-
family have been shown to modulate the balance between atin- or paclitaxel-induced apoptosis was observed after
8 Tumor Biology

DRP1 inhibition with mitochondrial division inhibitor-1 As in normal cells, mitochondrial dynamics in cancer
(mDIVI-1), which induced mitochondrial fusion in the cells have important implications for cellular function as
human melanoma cell line A375, which normally displays well as for sustaining proliferation and resistance to diverse
hyperfragmented mitochondria.48 types of stress. In general, the majority of studies that have
explored mitochondrial morphology in tumor cells support
a pro-tumorigenic role for mitochondrial fission (Table 1).51
Mitochondrial dynamics and cell cycle Reduced cancer cell growth and/or spontaneous apoptosis
progression induced by DRP1 inhibition alone or in combination with
Mitochondria grow continuously throughout the cell cycle chemotherapy have been observed both in vitro and in
and their dynamics and organization are tightly controlled vivo in several cancer types, including breast, colon, lung,
across its different phases to ensure proper cell division.49 pancreatic, hepatocellular carcinoma, and melanoma.49,52–58
In mammalian cells, evidence suggests that as cells pro- Moreover, alterations in the levels of proteins regulating
gress through G1, mitochondrial membrane potential and mitochondrial dynamics have been suggested as potential
respiration markedly increase.50 Then, at G1/S, mitochon- biomarkers of malignancy or as a target for therapy in
dria undergo a marked alteration in morphology, forming a diverse tumor types (Table 2). In this regard, inhibition of
giant and tubular network with hyperpolarized and highly mitochondrial fragmentation by silencing of DRP1 has
coupled mitochondria.49,50 This network is associated with been associated with increased genomic instability49 and
increased energy production to prepare for DNA synthesis. reduced migration and invasion59 in breast cancer cell lines
At this stage, mitochondrial hyperfusion also facilitates the which normally have a high level of fragmented mitochon-
mixing of mitochondrial contents to promote their homoge- dria. The mechanism underlying cellular dysfunction and
neity within the cell, ensuring proper mitochondrial inherit- cell death induced by decreased mitochondrial fission
ance following cellular division. Mitochondrial hyperfusion seems to be the replicative stress and mitotic defects that
triggers S-phase initiation and this alone is sufficient to severely affect the genomic integrity of cancer cells.
drive G0 quiescent cells into S phase of the cell cycle.49 Impaired mitochondrial fission also causes mitochondrial
During the following S, G2, and M phases, mitochon- dysfunction characterized by accumulation of mtDNA
dria become increasingly fragmented, reaching the highest mutations and generation of ROS. This combined effect of
fragmentation at mitosis to assure equal segregation of genome instability and mitochondrial dysfunction may
mitochondrial contents between daughter cells.49 Mitotic eventually result in the activation of the mitochondrial
mitochondrial fission is directly regulated by kinases apoptotic pathway, suggesting that inhibition of mitochon-
involved in mitosis, like Aurora A kinase that promotes drial fission could be a potential mechanism to enhance
RalA relocalization to mitochondrial membranes and cancer cell apoptosis, to increase sensitivity to therapy, or
recruits the effector RalBP1. RalBP1 functions as a scaf- to potentially circumvent resistance of cancer cells to con-
fold for cyclin B/cyclin-dependent kinase 1 (cdk1) that ventional chemotherapy.49,60
promotes phosphorylation of DRP1 at Ser616 resulting in In agreement with the previous observations, it has also
mitochondrial fission. Sustained mitochondrial hyperfu- been reported that several cancer cell lines or cells that have
sion beyond the G1/S border induces replication stress been transformed with oncogenes have more fragmented
causing ATM-dependent G2/M delay and chromosomal mitochondria than their non-transformed controls.48,55,56,59
instability during mitosis. Also, prolonged mitochondrial At least one of the molecular mechanisms involved in the
fusion causes mitochondrial bridges between daughter maintenance of fragmented mitochondria in cancer cells
cells resulting in a defective cytokinesis, unequal distribu- has been recently elucidated and it has been proposed to
tion of mitochondria, incorrect segregation of chromo- involve the MAPK signaling pathway, a proliferation path-
somes, and aneuploidy. Cells that experience persistent way activated in a variety of tumors. In normal cells, the
mitochondrial hyperfusion during mitosis present genomic RAS-RAF-MEK-ERK (MAPK) pathway is activated by
instability characterized by the presence of micronuclei growth factors and hormone signaling and its precise regu-
and disorganized nuclear structure.49 lation controls proliferation and cell death. In cancer cells,
excess signaling by growth factors, hormones, or onco-
genic mutations (like RASG12V or BRAFV600E) activate this
Mitochondrial dynamics and cancer pathway which results in aberrant signaling, uncontrolled
Cancer is a disease characterized by inappropriate cell pro- proliferation, and silencing of the cell death machinery. A
liferation, dysregulated cell cycle control, and defects in hallmark feature of cancer cells with MAPK oncogenic
apoptosis. Since mitochondrial dynamics has been shown activation is an increase in anaerobic glycolysis (Warburg
to be involved in the aforementioned mechanisms, it is not effect) and glutaminolysis. More recently, mitochondrial
surprising to find recent evidence that strongly relate the defects characterized by mitochondrial fragmentation with
processes of mitochondrial fusion and fission to cancer decreased mitochondrial oxygen consumption rate and
progression in diverse types of cancer. mitochondrial ATP production have been described in
Maycotte et al. 9

RAS-transformed cells.55 While the RalGEF pathway has MYC is another oncogene whose effects seem to be
been shown to promote mitochondrial fission during mito- mediated by the regulation of mitochondrial dynamics, and
sis through DRP1 phosphorylation by Cdk1, RAS- in contrast to RAS, MYC has been suggested to induce
transformed cells and tumors show mitochondrial mitochondrial fusion.32,34 Graves et al.34 reported augmented
fragmentation independent of the cell cycle, and this con- mitochondrial mass with increased membrane polarization, a
stitutive mitochondrial fission has been attributed to the correction of OXPHOS deficiency, and mitochondrial fusion
activation of ERK2.55,56 DRP1 has been shown to be phos- upon Myc re-expression in myc−/− fibroblasts. Myc re-­
phorylated by ERK2 on Ser616, and this phosphorylation is expression altered the levels of proteins involved in mito-
required for mitochondrial fission and tumor growth in chondrial dynamics ultimately favoring fusion and
RAS-transformed tumors, emphasizing the need for MAPK increasing mitochondrial mass.34 Also, in a recent study,
activation and consequent mitochondrial fragmentation in MYC induced PLD6 (a phospholipase of the OMM) expres-
tumors expressing oncogenic RAS.56 Interestingly, RAS is sion and mitochondrial fusion to promote biosynthesis of
mutated in up to 30% of all tumors and in 90% of PDACs, metabolic precursors and MYC-driven cell growth.32 Since
and BRAFV600E mutations have been identified in approxi- MYC potently stimulates cell growth and proliferation, the
mately 66% of melanomas and in a smaller percentage in authors suggest that MYC-induced mitochondrial fusion is
other tumors (thyroid, colon, ovarian cancer, and sarco- responsible for increased lipid, nucleotide, and protein
mas).61 Tumors or cell lines from these types of cancer are metabolism, which strain cellular energetic resources and
known to have an altered metabolism and increased mito- hence activate adenosine monophosphate–activated protein
chondrial fragmentation55,56. Besides, DRP1 knockdown, kinase (AMPK, which is activated in response to high AMP
expression of the dominant-negative DRP1-K38A or inhi- and low ATP levels) to inhibit yes-associated protein (YAP)/
bition of DRP1 phosphorylation on Ser616 decreased tumor TAZ coactivator activity. In this work, induction of mito-
growth in a RAS-driven mouse tumor model, suggesting chondrial fusion by mDIVI-1 was sufficient to activate
that DRP1 is a potential therapeutic target in tumors AMPK indicating that MYC exerts at least some of its
expressing oncogenic RAS. Also, DRP1S616 phosphoryla- effects on metabolism and cell proliferation by inducing
tion was found to correlate with BRAFV600E in human mel- mitochondrial fusion.32 Interestingly, MYC has been found
anoma samples,55 and DRP1 phosphorylation or RAS to be highly expressed in triple-negative breast tumors and
transformation led to decreased mitochondrial membrane has been associated with poor prognosis in breast cancer
potential, increased production of mitochondrial ROS, and patients,32 suggesting a potential use for inhibitors of mito-
reduced basal and maximal oxygen consumption rates,55 chondrial fusion for this type of cancer.
indicating that ERK2-mediated DRP1 phosphorylation is Regarding metastasis, human breast carcinomas and
responsible for decreased mitochondrial function in RAS- metastasis to lymph nodes have been found to have higher
transformed cells. In mouse tumors, DRP1 inhibition levels of DRP1 than ductal carcinoma in situ samples or
induced a decrease in tumor vasculature and decreased vas- than the normal/adjacent tissue.59 Also, invasive breast
cular endothelial growth factor (VEGF) messenger RNA cancer cell lines have been found to have fragmented mito-
(mRNA).56 The authors suggest that one of the possible chondria, higher DRP1 and DRP1S616 levels, as well as
mechanisms of tumorigenesis induced by MAPK-mediated lower MFN1 than a non-metastatic cell line.59 In the same
mitochondrial fission could be the activation of proangio- study, DRP1 knockdown, DRP1 inhibition with mDIVI-1,
genic signaling pathways, which are sensitive to mitochon- or MFN-1/2 expression decreased migration and invasion
dria-derived ROS. Also, with regard to MAPK-induced in the metastatic cell lines without affecting cell cycle or
mitochondrial fragmentation, c-Jun N-terminal protein viability. Mitochondrial fission induced cell spreading
kinase (JNK) activation during chemotherapy-induced and increased lamellipodia formation as well as mito-
apoptosis has been related to proteasomal degradation of chondrial distribution to the lamellipodia region at the
MFN2 through the activation of Huwe1 ubiquitin ligase, leading edge of invading cells in response to chemoat-
decreasing mitochondrial fusion and inducing apoptosis.13 tractants, suggesting a crucial role of mitochondrial fis-
Therefore, MAPK activation in cancer has been linked to sion in the migration and invasion of breast cancer cells.59
mitochondrial fission and can induce transformation in Also, since hypoxia is a major stimulator of migration and
some cases (RAS transformation) and apoptosis in other invasion in cancer cells, another study found that hypoxia
contexts (chemotherapy-induced apoptosis). Importantly, increased the expression of DRP1 and mitochondrial fis-
in the first case, DRP1 inhibition (inhibition of fission) sion in a glioblastoma cell line. In the same study, hypoxia-
decreased tumor growth in a mouse model and in patient- induced migration was decreased with mDIVI-1 treatment
derived pancreatic cancer cell lines;56 while in the second or a dominant-negative DRP1-K38A.62 These studies sug-
case, MFN2 knockdown (inhibition of fusion) increased gest that targeting mitochondrial fission is likely to have a
apoptosis in doxorubicin-treated cells, highlighting the therapeutic benefit in highly invasive cancers like glio-
need to precisely understand how to manipulate mitochon- blastoma, which is one of the most aggressive cerebral
drial dynamics for cancer therapy. gliomas, or in other highly metastatic cancers.
10 Tumor Biology

Other studies have found that tumor cells with increased increased PGC-1-α and mitochondrial mass, increased
mitochondrial fragmentation are more sensitive to meta- oxygen consumption rate as well as a strong reliance on
bolic stress and have proposed mitochondrial inhibition as mitochondrial respiration for survival, and a decreased
a therapeutic target in these types of cancers. For example, dependence on glycolysis for cellular energetics.38 In the
in prostate cancer, androgens and androgen receptors play same study, mitochondria from TICs had an altered mor-
critical roles in the proliferation of prostate cancer through phology, were hyperpolarized, and produced more ROS.
transcriptional regulation of target genes. Androgens have The authors suggest that altered metabolic and mito-
been found to upregulate the expression of DRP1, facilitat- chondrial functions are critical features of TICs in this
ing mitochondrial fragmentation and apoptosis in prostate model, making them more resistant to apoptosis.
cancer cell lines in response to mitochondrial stress. Interestingly, OXPHOS inhibition by oligomycin
Interestingly, in the same study, clinical tissue samples of decreased ATP content and induced cell death in TICs,
prostate carcinoma showed high levels of DRP1 in andro- which were unable to upregulate glycolysis when com-
gen-dependent cancers but not in androgen independent pared to KRAS expressing cells from the same model.
cancers. The authors suggest that androgens might func- Altered metabolism in these TICs was attributed to
tion as a pro-apoptotic stimulus when combined with increased protein and fatty acid metabolism, making
agents that affect mitochondrial function in androgen- them more sensitive to autophagy (which mediates lipid
dependent cancers.63 Also, with regard to hormonal regu- droplet degradation) and β-oxidation inhibitors. These
lation of mitochondrial dynamics, estradiol treatment has findings indicate that TICs are more dependent on mito-
been shown to affect the levels of proteins regulating mito- chondrial respiration than the highly glycolytic bulk of
chondrial dynamics and to induce mitochondrial fusion in tumor cells.38
the MCF-7, estrogen receptor positive cell line.64 The In agreement with the previous report, in a different
above-mentioned studies indicate that hormones have a model, brain TICs isolated from tumor xenografts or from
strong influence on mitochondrial dynamics and the over- primary tumors have been found to have fragmented
all effect might involve hormone-specific effects, the type mitochondria and increased levels of activating DRP1S616
of malignancy, or the type of hormone receptors present in phosphorylation relative to non-TICs, which showed
the cancer cell or tumor tissue,65 underscoring the impor- higher levels of DRP1S637 inactivating phosphorylation.
tance of understanding how to manipulate mitochondrial Interestingly, transfection with a phosphomimetic,
dynamics for the treatment of hormone-related malignan- ­inactivation-incompetent double-mutant DRP1S616E,    S637A
cies. Besides decreasing mitochondrial metabolism, it has induced expression of stem cell markers in non-TIC tumor
also been shown that mitochondrial fragmentation cells, and DRP1 knockdown or its pharmacological inhibi-
increases glycolytic metabolism and makes cancer cells tion with mDIVI-1 decreased tumor growth in two brain
more sensitive to glycolysis inhibitors.66 In this study, sur- tumor mouse models. Furthermore, while total DRP1 lev-
vivin, a member of the Inhibitor of Apoptosis Protein fam- els were similar in normal and neoplastic brain tissues,
ily which is overexpressed in neuroblastoma, induced activating phosphorylation of DRP1 (DRP1S616) was
increased DRP1 levels and mitochondrial fragmentation in increased in glioblastomas, and a strong inverse correla-
a neuroblastoma cell line. Mitochondrial fission increased tion was found between DRP1S616 and poor survival in
glycolysis and lactate production and made cells more sen- glioblastoma patients.69 In this work, the authors suggest
sitive to 2-deoxyglucose alone or in combination with that cyclin-dependent kinase 5 (CDK5) was responsible
chemotherapy. These studies suggest that mitochondrial for DRP1 activation in brain TICs and that calcium-/calm-
fragmentation could be a biomarker for treatment with odulin-dependent protein kinase type 2 (CAMK2) was
either or both mitochondrial and glycolytic metabolic responsible for inhibitory phosphorylation of DRP1 in
inhibitors. Ser637 in non-TICs.69 Since glioblastomas rank among the
It has also been suggested that mitochondrial inhibi- most lethal cancers, it will be interesting to explore DRP1
tion could be a potential therapeutic target for the highly inhibition as a therapeutic strategy for these tumors.
tumorigenic tumor-initiating cells (TICs). TICs or cancer In addition, mammospheres of breast cancer cell lines,
stem cells have been defined as cells within the tumor which are known to enrich TICs, as well as breast cancer
that possess clonal long-term repopulation and self- tumors, overexpress mitochondrial enzymes, proteins
renewal capacity. These cells have been proposed to be involved in mitochondrial biogenesis, and protein inhibitors
responsible for therapy failure and the formation of of mitophagy when compared to their surrounding tissue.70
metastasis.67,68 In a PDAC model, dormant tumor cells The previous findings suggest that tumor cells, and more
surviving oncogene-ablation (KRAS withdrawal) were specifically TICs, resist stress by increasing their ATP pro-
responsible for tumor relapse and had TIC features. duction through oxidative mitochondrial metabolism.
These TICs showed prominent expression of genes gov- Regarding mitochondrial dynamics, MYC-induced prolif-
erning electron transport chain, autophagy, lysosome eration was shown to be mediated by the induction of
activity, mitochondrial and peroxisomal oxidation, PLD6-mediated mitochondrial fusion and resulted in a
Maycotte et al. 11

decrease in mammosphere forming ability in CD44high/ shapes,72 suggesting alterations in function when com-
CD24low mammary epithelial cells.32 This work, together pared to normal cells, but the functional implications of
with the previously mentioned studies, suggests a relation- these observations or their importance for cancer treatment
ship between mitochondrial fragmentation and TIC fea- are not clear. Moreover, despite an increased understand-
tures or between mitochondrial fusion and a decrease in ing of the molecular mechanisms regulating mitochondrial
TIC characteristics. fusion and fission and their role in certain cancer types or
Other studies suggest that mitochondrial fusion could cancer-related situations, a precise understanding of a
be involved in resistance to therapy. Cervical and ovarian mechanism to manipulate mitochondrial dynamics is not
cancer cell lines resistant to cisplatin showed a higher pro- being used for cancer therapy.
portion of tubular mitochondria and reduced sensitivity to The main complication for effectively targeting mito-
cisplatin-induced mitochondrial fragmentation and apop- chondrial fusion or fission for therapy or for using mito-
tosis induction than their non-resistant controls. In this chondrial morphology as a cancer biomarker is the
work, the authors show that p53 re-expression induces context-dependent role of mitochondria in diverse types of
mitochondrial fragmentation in p53 mut or p53 null cell cancer or settings (Table 1). However, with a few excep-
lines. The mechanism by which p53 regulates apoptosis in tions, most of the evidence suggests that increased mito-
cisplatin-sensitive cells involves cisplatin-mediated p53 chondrial fragmentation can be used as a diagnostic
phosphorylation which translocates to the mitochondria biomarker for tumor progression or for certain oncogene
and binds prohibitin1, releasing OPA1 and allowing its mutations (Table 2), although whether this is a general fea-
processing to S-OPA1 by Oma1, whose proteolytic activa- ture for all tumor types remains to be determined.
tion is also induced by p53, inducing mitochondrial frag- Oncogene transformation has been shown to have
mentation and apoptosis.10 This will be an interesting diverse effects on mitochondrial morphologies: while
hypothesis since, as we mentioned previously, more than RAS transformation was shown to mediate mitochondrial
half of human cancers harbor p53 mutations and even fragmentation,55,56 MYC-induced oncogenesis induced
more have alterations in the pathways that regulate its sta- mitochondrial elongation.32,34 Importantly, these changes
bility within the cell,28 suggesting that the induction of in mitochondrial dynamics were shown to be relevant for
mitochondrial fragmentation could help restore the sensi- oncogene-mediated transformation and proliferation.
bility to therapy. Besides, p53, the most frequently mutated tumor suppres-
Other links have been described between the mitochon- sor gene, induced mitochondrial fragmentation and apop-
drial fusion–fission machinery and p53. p53 has been tosis when re-expressed in p53-mutated or null ovarian
shown to bind DRP1, which mediates the monoubiquitina- cancer cell lines.10 The aforementioned evidence suggests
tion of p53 by mouse double minute 2 homolog (MDM2). that the array of mutations in oncogenes or tumor suppres-
This allows p53 to translocate to the mitochondria and ini- sors could define mitochondrial morphology and probably
tiate necrosis in response to oxidative stress. Also, p53 mitochondrial function within a particular tumor. In this
directly promotes DRP1 expression in conditions of oxida- regard, steroid hormones were also shown to affect mito-
tive stress, which induces mitochondrial fission and the chondrial dynamics in hormone-related malignancies:
activation of caspase-3, and the enforced expression of while androgens increased mitochondrial fragmentation in
miR-30 attenuated mitochondrial fission by repressing androgen-sensitive prostate cancer, estradiol induced
p53.71 Since p53 also transcriptionally regulates the mitochondrial fusion in an estrogen receptor–positive
expression of proteins that promote mitochondrial apopto- breast cancer cell line. Thus, it will be important to identify
sis, it will be interesting to determine whether its role in whether changes in mitochondrial morphology are specific
the induction of mitochondrial fragmentation causes cell to an array of cancer mutations or to cancer cells in a tis-
death by itself or if it is only a mechanism to facilitate cell sue, whether these changes contribute to tumor progres-
dismissal during apoptosis. sion, and whether mitochondrial dynamics can be
effectively targeted for therapy.
Regarding treatment, the induction of mitochondrial
Discussion fragmentation is generally associated with a pro-apoptotic
Since changes in mitochondrial morphology are closely effect in cancer cells treated with chemotherapy or ultravio-
linked to important cellular functions, including those let (UV) radiation, suggesting that the induction of fission
altered in cancer, it is not surprising that alterations in or inhibition of fusion could be potentially used in combi-
mitochondrial dynamics are associated with tumor pro- nation with other therapies during cancer treatment in sev-
gression or resistance to therapy. Several alterations in eral malignancies. However, the proposal that mitochondria
mitochondrial mass and ultrastructure have been reported in need a minimal size permissive for apoptosis and that
neoplastic tissues, including ultrastructural heterogeneity, hyperfragmented mitochondria are not involved in apopto-
cristal disorganization, altered mitochondrial membranes, sis48 raises important questions as to how to induce mito-
matricial vacuoles, mitochondrial swelling, and distorted chondrial fragmentation and to what extent. Importantly, in
12 Tumor Biology

Table 1.  Mitochondrial dynamics and its relationship with cancer-related processes.

Mitochondrial Cancer-related Observation Treatment Cancer cell line Ref.


process process
Fission Pro-apoptotic DRP1 associates with OMM Staurosporine HeLa cervical cancer Wasiak et al.46
due to BAX/BAK dependent or anti-Fas- cell line
SUMOylation and precedes treated HeLa
MOMP. cells
Fission Pro-apoptotic DRP1-induced mitochondrial Staurosporine, HeLa and DLBCL Wasiak et al.46
membrane curvature is required anti-Fas, and UV (diffuse large B-cell and Wang
for efficient insertion of BAX. irradiation lymphoma) cell lines et al.47
Fission Pro-apoptotic JNK induced MFN2 proteasomal Doxorubicin U2OS osteosarcoma Leboucher
degradation decreasing cell line Guillaume
mitochondrial fusion during et al.13
chemotherapy-induced apoptosis.
Fission Pro-apoptotic Cisplatin resistant cervical and Cisplatin Cervical (OV2008 and Kong et al.10
ovarian cancer cell lines have more C13*) and ovarian
tubular mitochondria. P53 re- (A2780, SKOV3, and
expression in p53 mutant or null HEY) cancer cell lines
cell lines induced mitochondrial
fragmentation and apoptosis.
Fission Apoptosis DRP1-induced changes in – DLBCL cell lines Wang et al.47
mitochondrial fragmentation are
not sufficient to induce apoptosis.
Fission Apoptosis Hyperfragmented mitochondria Terminal Mfn1−/− MEFs and Renault et al.48
are not involved in apoptosis unfolded protein A375 human melanoma
but they need a minimal size response, cell line
permissive for pro-apoptotic BCL2 cisplatin, and
protein family function. paclitaxel
Fission Anti-apoptotic DRP1 inhibition sensitized to Cisplatin and A375 human melanoma Renault et al.48
treatment in a cancer cell line paclitaxel cell line
that normally displays fragmented
mitochondria.
Fission Pro- DRP1 knockdown induced – MDA-MB-231 breast Qian et al.49
tumorigenic genomic instability, cellular cancer cell line
dysfunction, and cell death in a
cancer cell line which normally
displays fragmented mitochondria.
Fission Pro- RAS-transformed cells have – E1A + RasG12V Serasinghe
tumorigenic fragmented mitochondria, and expressing MEFs, HEK- et al.55 and
inhibition of DRP1 decreased TtH HRASG12V, A375, Kashatus et al.56
tumor growth in a mouse model. or SK-MEL-28 BRAFV600E
cell lines, BT-474 ErbB2
overexpressing cell line,
and patient-derived
KRAS mutant pancreatic
cell lines
Fission Pro- Invasive breast cancer cell – MDA-MB-231 and Zhao et al.59
tumorigenic lines have more fragmented MDA-MB-436 breast
mitochondria, and DRP1 inhibition cancer cell lines
decreased migration and invasion
without affecting viability.
Fission Pro- Hypoxia-induced migration was Hypoxia (1% U251 glioblastoma cell Wan et al.62
tumorigenic decreased with mDIVI-1 treatment O2 or CoCl2 line
or expression of a dominant- treatment)
negative DRP1-K38A.
Fission Pro- Androgens increase DRP1 and Mitochondrial LNCaP and VCaP Choudhary
tumorigenic mitochondrial fragmentation stress prostate cancer cell line et al.63
in androgen-sensitive prostate
cancers, making them more
sensitive to mitochondrial stress.
(Continued)
Maycotte et al. 13

Table 1. (Continued)

Mitochondrial Cancer-related Observation Treatment Cancer cell line Ref.


process process
Fission Pro- Brain tumor–initiating cells – CD133+ brain tumor– Xie et al.69
tumorigenic, from tumor xenografts or initiating cells from
increased TIC primary tumors have fragmented tumors or xenografts
markers mitochondria and increased
DRP1S616. Phospho-mimetic
DRP1S616 induced TIC markers.
Fusion Increased MYC transformed cells have – MYC expressing von Eyss et al.32
proliferation increased mitochondrial fusion CD44high/CD24low
and decreased which promotes mitochondrial mammary epithelial cells
TIC features metabolism, cell proliferation and
decreases TIC markers.

UV: ultraviolet; TIC: tumor-initiating cells; OMM: outer mitochondrial membrane.


Some of the recent studies regarding mitochondrial fusion or fission and the suggested role in different aspects of apoptosis or cancer progression
are mentioned.

Table 2.  Alterations in proteins regulating mitochondrial dynamics in tumor samples and their suggested use as prognostic or
diagnostic biomarkers.

Alterations in Type of cancer Suggested use as a prognostic or diagnostic biomarker Ref.


proteins related to
mitochondrial dynamics
Increased DRP1 Lung Higher recurrence rate than patients with cytoplasmic DRP1. Nuclear Chiang
nuclear staining adenocarcinomas DRP1 was associated with advanced tumor stage, patient’s smoking et al.73
habits, and hypoxia. No difference in survival was found between
patients with high DRP1 levels and those with low DRP1 levels.
Increased DRP1 and Lung Increased DRP1 and decreased MFN2 staining was found in lung cancer Rehman
decreased MFN2 adenocarcinomas tissue when compared to healthy lung. This study used a small number et al.53
of tumor samples.
Increased p-DRP1S616 Melanoma Phosphorylation of DRP1 at Ser616 was significantly associated with Serasinghe
BRAFV600E mutations when compared to BRAFwt melanoma samples or et al.55 and
normal skin suggesting its use as a biomarker for BRAF mutations for Wieder
targeting MAPK or as a pharmacological target in this type of cancer. et al.74
Increased p-DRP1S616 Pancreatic ductal DRP1S616 correlated with ERK phosphorylation and mitochondrial Kashatus
adenocarcinoma fragmentation suggesting that mitochondrial fragmentation or DRP1S616 et al.56
could be used as a marker of MAPK pathway activation or as a marker
for targeting DRP1 in this type of cancer. This study used a small
number of tumor samples.
Increased DRP1 Breast cancer Increased DRP1 expression in breast cancer specimens when Zou et al.57
staining compared to adjacent normal tissues. Increased DRP1 staining in and Zhao
noninvasive ductal carcinoma in situ which was much more intense in et al.59
invasive carcinoma and metastases to lymph nodes.
Increased DRP1 Prostate cancer High DRP1 levels in post-surgery samples from prostate cancer Choudhary
staining patients correlated with the expression of the androgen receptor (AR) et al.63
suggesting a potential use for targeting mitochondria in AR+ prostate
cancers. This study used a small number of tumor samples.
Increased p-DRP1S616 Glioblastoma Increased DRP1S616 phosphorylation was increased in glioblastoma Xie et al.69
samples and correlated with worse patient survival.
Increased DRP1 Hepatocellular DRP1 mRNA was upregulated in hepatocellular carcinoma when Zhan et al.52
mRNA and protein carcinoma compared with non-tumor tissues. DRP1 protein levels were increased and Huang
and decreased MFN1 and MFN1 protein levels were decreased in tumor samples when et al.58
protein levels compared to peritumor area. Patients with high DRP1 expression had
a worse overall survival, while patients with high expression of MFN1
had a better overall survival. A lower ratio of DRP1/MFN1 was a
better predictor for better overall survival.

MAPK: mitogen-activated protein kinase; ERK: extracellular signal–regulated protein kinase; mRNA: messenger RNA.
14 Tumor Biology

cancer cells that normally display fragmented mitochondria, Funding


DRP1 inhibition sensitized them to treatment10,48 or induced This work was supported by the following project from the
cell death per se,49 underscoring the importance of under- Instituto Mexicano del Seguro Social (IMSS):
standing whether the induction of mitochondrial fragmenta- CTFIS/1ORD/012/2011. P.M. is a recipient of a Cátedra
tion or inhibition of fusion will only be useful to target CONACYT (Consejo Nacional de Ciencia y Tecnología) for
cancer cells with tubular mitochondria. young investigators (Cátedras CONACYT/3103).
The suggestion that TICs, those cells within a tumor that
have tumor-initiating capacity and that have been sug- References
gested to be responsible for tumor relapse, display increased 1. Dominy JE and Puigserver P. Mitochondrial biogen-
mitochondrial fragmentation and are more sensitive to esis through activation of nuclear signaling proteins. Cold
mitochondrial stress in several tissues (i.e. a pancreatic can- Spring Harb Perspect Biol 2013; 5: a015008.
cer model,38 breast cancer mammospheres,70 or brain tumor 2. Tait SW and Green DR. Mitochondria and cell signalling.
TICs from tumor xenografts or primary tumors69) raises an J Cell Sci 2012; 125: 807–815.
important suggestion for targeting mitochondria in combi- 3. Youle RJ and van der Bliek AM. Mitochondrial fission,
nation with cancer therapy in order to eliminate TICs in the fusion, and stress. Science 2012; 337: 1062–1065.
4. Palmer CS, Osellame LD, Stojanovski D, et al. The regu-
tumor. Importantly, some of the above-mentioned studies
lation of mitochondrial morphology: intricate mechanisms
suggest targeting mitochondrial oxidative metabolism,38,70 and dynamic machinery. Cell Signal 2011; 23: 1534–1545.
and regarding mitochondrial dynamics, Xie et al.69 showed 5. van der Bliek AM, Shen Q and Kawajiri S. Mechanisms
that DRP1 knockdown or mDIVI-1 treatment decreased of mitochondrial fission and fusion. Cold Spring Harb
brain TICs. Perhaps one of the most important findings of Perspect Biol 2013; 5: a011072.
this study is the fact that expression of DRP1 with two 6. Wai T and Langer T. Mitochondrial dynamics and meta-
mutations (Ser616E that mimics an activating phosphoryla- bolic regulation. Trends Endocrinol Metabol 2016; 27:
tion and Ser637A that blocks inhibitory phosphorylation) 105–117.
induced mitochondrial fragmentation and the expression of 7. Senft D and Ronai ZA. Regulators of mitochondrial dynam-
some core stem cell regulators and repression of differen- ics in cancer. Curr Opin Cell Biol 2016; 39: 43–52.
tiation markers in non-TICs. Therefore, mitochondrial 8. Lagouge M, Argmann C, Gerhart-Hines Z, et al. Resveratrol
improves mitochondrial function and protects against meta-
fragmentation by itself can induce TIC features in cancer,
bolic disease by activating SIRT1 and PGC-1alpha. Cell
making mitochondrial fission a potential target in these 2006; 127: 1109–1122.
malignancies. Whether this is particular for brain TICs or 9. Liesa M, Borda-d’Agua B, Medina-Gomez G, et al.
whether it is a general feature of TICs in other tissues Mitochondrial fusion is increased by the nuclear coactivator
remains to be established. PGC-1beta. PLoS ONE 2008; 3: e3613.
Finally, several questions remain to be answered. Are 10. Kong B, Wang Q, Fung E, et al. p53 is required for cispl-
there four mitochondrial states (hyperfused, fused, frag- atin-induced processing of the mitochondrial fusion protein
mented, and hyperfragmented) rather than just two (fused L-Opa1 that is mediated by the mitochondrial metallopepti-
and fragmented) and do they have different functions? Are dase Oma1 in gynecologic cancers. J Biol Chem 2014; 289:
there particular biomarkers for each of these morpholo- 27134–27145.
gies? How is this relevant to the different cancer cells? Is 11. Anand R, Langer T and Baker MJ. Proteolytic control

of mitochondrial function and morphogenesis. Biochim
mitochondrial fission only a feature of MAPK-transformed
Biophys Acta 2013; 1833: 195–204.
cancers and fusion a feature of MYC transformation? How 12. Chang CR and Blackstone C. Dynamic regulation of mito-
do other oncogenes affect mitochondrial dynamics? How chondrial fission through modification of the dynamin-
does mitochondrial dynamics manipulation combine with related protein Drp1. Ann N Y Acad Sci 2010; 1201: 34–39.
conventional chemotherapies used to treat these types of 13. Leboucher Guillaume P, Tsai Yien C, Yang M, et al. Stress-
cancer? Is mitochondrial inhibition useful for TICs in all induced phosphorylation and proteasomal degradation of
cancer types? Is mitochondrial fission necessary for migra- mitofusin 2 facilitates mitochondrial fragmentation and
tion in all cancer cells? It is very likely that a more precise apoptosis. Mol Cell 2012; 47: 547–557.
understanding of the mechanisms governing mitochon- 14. Morciano G, Pedriali G, Sbano L, et al. Intersection of mito-
drial dynamics and their role in cancer will be available chondrial fission and fusion machinery with apoptotic path-
soon, allowing for an improvement in cancer-related thera- ways: role of Mcl-1. Biol Cell 2016; 108(10): 279–293.
15. Kashatus DF. Restraining the divider: a Drp1-phospholipid
pies as well as for their use as cancer-related biomarkers.
interaction inhibits Drp1 activity and shifts the balance
from mitochondrial fission to fusion. Mol Cell 2016; 63:
Declaration of conflicting interests 913–915.
The author(s) declared no potential conflicts of interest with 16. Adachi Y, Itoh K, Yamada T, et al. Coincident phosphatidic
respect to the research, authorship, and/or publication of this acid interaction restrains Drp1 in mitochondrial division.
article. Mol Cell 2016; 63: 1034–1043.
Maycotte et al. 15

17. Baba T, Kashiwagi Y, Arimitsu N, et al. Phosphatidic acid 36. Chiaradonna F, Sacco E, Manzoni R, et al. Ras-dependent
(PA)-preferring phospholipase A1 regulates mitochondrial carbon metabolism and transformation in mouse fibroblasts.
dynamics. J Biol Chem 2014; 289: 11497–11511. Oncogene 2006; 25: 5391–5404.
18. Parra V, Verdejo HE, Iglewski M, et al. Insulin stimulates 37. Kerr EM, Gaude E, Turrell FK, et al. Mutant Kras copy
mitochondrial fusion and function in cardiomyocytes via the number defines metabolic reprogramming and therapeutic
Akt-mTOR-NFkappaB-Opa-1 signaling pathway. Diabetes susceptibilities. Nature 2016; 531: 110–113.
2014; 63: 75–88. 38. Viale A, Pettazzoni P, Lyssiotis CA, et al. Oncogene abla-
19. Chen H and Chan DC. Mitochondrial dynamics—fusion, tion-resistant pancreatic cancer cells depend on mitochon-
fission, movement, and mitophagy—in neurodegenerative drial function. Nature 2014; 514: 628–632.
diseases. Hum Mol Genet 2009; 18: R169–R176. 39. Amelio I and Melino G. The p53 family and the hypoxia-
20. Gomes LC, Di Benedetto G and Scorrano L. During
inducible factors (HIFs): determinants of cancer progres-
autophagy mitochondria elongate, are spared from degra- sion. Trends Biochem Sci 2015; 40: 425–434.
dation and sustain cell viability. Nat Cell Biol 2011; 13: 40. Hanahan D and Weinberg RA. Hallmarks of cancer: the
589–598. next generation. Cell 2011; 144: 646–674.
21. Tondera D, Grandemange S, Jourdain A, et al. SLP-2 is 41. Andersen JL and Kornbluth S. The tangled circuitry of
required for stress-induced mitochondrial hyperfusion. metabolism and apoptosis. Mol Cell 2013; 49: 399–410.
EMBO J 2009; 28: 1589–1600. 42. Adams JM. Ways of dying: multiple pathways to apoptosis.
22. Gogvadze V, Orrenius S and Zhivotovsky B. Mitochondria Genes Dev 2003; 17: 2481–2495.
in cancer cells: what is so special about them? Trends Cell 43. Chipuk JE, Moldoveanu T, Llambi F, et al. The BCL-2
Biol 2008; 18: 165–173. ­family reunion. Mol Cell 2010; 37: 299–310.
23. Chen X, Qian Y and Wu S. The Warburg effect: evolving 44. Giménez-Cassina A, Garcia-Haro L, Choi Cheol S, et al.
interpretations of an established concept. Free Radic Biol Regulation of hepatic energy metabolism and gluconeogen-
Med 2015; 79: 253–263. esis by BAD. Cell Metab 2014; 19: 272–284.
24. Raez LE, Papadopoulos K, Ricart AD, et al. A phase I dose- 45. Karbowski M, Norris KL, Cleland MM, et al. Role of Bax
escalation trial of 2-deoxy-D-glucose alone or combined and Bak in mitochondrial morphogenesis. Nature 2006;
with docetaxel in patients with advanced solid tumors. 443: 658–662.
Cancer Chemother Pharmacol 2013; 71: 523–530. 46. Wasiak S, Zunino R and McBride HM. Bax/Bak promote
25. Boland ML, Chourasia AH and Macleod KF. Mitochondrial sumoylation of DRP1 and its stable association with mito-
dysfunction in cancer. Front Oncol 2013; 3: 292. chondria during apoptotic cell death. J Cell Biol 2007; 177:
26. Moreno-Sanchez R, Rodriguez-Enriquez S, Marin-
439–450.
Hernandez A, et al. Energy metabolism in tumor cells. 47. Wang P, Liu B, Zhao J, et al. Dynamin-related protein
FEBS J 2007; 274: 1393–1418. Drp1 is required for Bax translocation to mitochondria in
27. Ralph SJ, Rodriguez-Enriquez S, Neuzil J, et al. The causes response to irradiation-induced apoptosis. Oncotarget 2015;
of cancer revisited: “mitochondrial malignancy” and ROS- 6: 22598–22612.
induced oncogenic transformation–why mitochondria are tar- 48. Renault TT, Floros KV, Elkholi R, et al. Mitochondrial
gets for cancer therapy. Mol Aspects Med 2010; 31: 145–170. shape governs BAX-induced membrane permeabilization
28. Brady CA and Attardi LD. p53 at a glance. J Cell Sci 2010; and apoptosis. Mol Cell 2015; 57: 69–82.
123: 2527–2532. 49. Qian W, Wang J and Van Houten B. The role of dynamin-
29. Papandreou I, Cairns RA, Fontana L, et al. HIF-1 mediates related protein 1 in cancer growth: a promising therapeutic
adaptation to hypoxia by actively downregulating mitochon- target? Expert Opin Ther Targets 2013; 17: 997–1001.
drial oxygen consumption. Cell Metab 2006; 3: 187–197. 50. Lee IH and Finkel T. Metabolic regulation of the cell cycle.
30. Scarpulla RC. Metabolic control of mitochondrial biogen- Curr Opin Cell Biol 2013; 25: 724–729.
esis through the PGC-1 family regulatory network. Biochim 51. Corrado M, Scorrano L and Campello S. Mitochondrial
Biophys Acta 2011; 1813: 1269–1278. dynamics in cancer and neurodegenerative and neuroin-
31. Zhang H, Gao P, Fukuda R, et al. HIF-1 inhibits mitochon- flammatory diseases. Int J Cell Biol 2012; 2012: 729290.
drial biogenesis and cellular respiration in VHL-deficient 52. Zhan L, Cao H, Wang G, et al. Drp1-mediated mitochon-
renal cell carcinoma by repression of C-MYC activity. drial fission promotes cell proliferation through crosstalk
Cancer Cell 2007; 11: 407–420. of p53 and NF-κB pathways in hepatocellular carcinoma.
32. von Eyss B, Jaenicke LA, Kortlever RM, et al. A MYC- Oncotarget 2016; 7: 65001–65011.
driven change in mitochondrial dynamics limits YAP/TAZ 53. Rehman J, Zhang HJ, Toth PT, et al. Inhibition of mitochon-
function in mammary epithelial cells and breast cancer. drial fission prevents cell cycle progression in lung cancer.
Cancer Cell 2015; 28: 743–757. FASEB J 2012; 26: 2175–2186.
33. Vyas S, Zaganjor E and Haigis MC. Mitochondria and can- 54. Inoue-Yamauchi A and Oda H. Depletion of mitochondrial
cer. Cell 2016; 166: 555–566. fission factor DRP1 causes increased apoptosis in human
34. Graves JA, Wang Y, Sims-Lucas S, et al. Mitochondrial colon cancer cells. Biochem Biophys Res Commun 2012;
structure, function and dynamics are temporally controlled 421: 81–85.
by C-MYC. PLoS ONE 2012; 7: e37699. 55. Serasinghe MN, Wieder SY, Renault TT, et al. Mitochondrial
35. Morita M, Gravel SP, Hulea L, et al. mTOR coordinates division is requisite to RAS-induced transformation and tar-
protein synthesis, mitochondrial activity and proliferation. geted by oncogenic MAPK pathway inhibitors. Mol Cell
Cell Cycle 2015; 14: 473–480. 2015; 57: 521–536.
16 Tumor Biology

56. Kashatus JA, Nascimento A, Myers LJ, et al. Erk2 phospho- 65. Sastre-Serra J, Nadal-Serrano M, Pons DG, et al. The over-
rylation of drp1 promotes mitochondrial fission and MAPK- expression of ERbeta modifies estradiol effects on mito-
driven tumor growth. Mol Cell 2015; 57: 537–551. chondrial dynamics in breast cancer cell line. Int J Biochem
57. Zou P, Liu L, Zheng LD, et al. Coordinated upregulation Cell Biol 2013; 45: 1509–1515.
of mitochondrial biogenesis and autophagy in breast cancer 66. Hagenbuchner J, Kuznetsov AV, Obexer P, et al. BIRC5/
cells: the role of dynamin related protein-1 and implication survivin enhances aerobic glycolysis and drug resistance
for breast cancer treatment. Oxid Med Cell Longev 2016; by altered regulation of the mitochondrial fusion/fission
2016: 4085727. machinery. Oncogene 2013; 32: 4748–4757.
58. Huang Q, Zhan L, Cao H, et al. Increased mitochondrial 67. Oskarsson T, Batlle E and Massague J. Metastatic stem
fission promotes autophagy and hepatocellular carcinoma cells: sources, niches, and vital pathways. Cell Stem Cell
cell survival through the ROS-modulated coordinated regu- 2014; 14: 306–321.
lation of the NFKB and TP53 pathways. Autophagy 2016; 68. Kreso A and Dick JE. Evolution of the cancer stem cell
12: 999–1014. model. Cell Stem Cell 2014; 14: 275–291.
59. Zhao J, Zhang J, Yu M, et al. Mitochondrial dynamics 69. Xie Q, Wu Q, Horbinski CM, et al. Mitochondrial control
regulates migration and invasion of breast cancer cells. by DRP1 in brain tumor initiating cells. Nat Neurosci 2015;
Oncogene 2013; 32: 4814–4824. 18: 501–510.
60. Parone PA, Da Cruz S, Tondera D, et al. Preventing mito- 70. Lamb R, Harrison H, Hulit J, et al. Mitochondria as new
chondrial fission impairs mitochondrial function and leads therapeutic targets for eradicating cancer stem cells: quan-
to loss of mitochondrial DNA. PLoS ONE 2008; 3: e3257. titative proteomics and functional validation via MCT1/2
61. Michaloglou C, Vredeveld LC, Mooi WJ, et al. BRAF(E600) inhibition. Oncotarget 2014; 5: 11029–11037.
in benign and malignant human tumours. Oncogene 2008; 71. Dashzeveg N and Yoshida K. Cell death decision by p53 via
27: 877–895. control of the mitochondrial membrane. Cancer Lett 2015;
62. Wan YY, Zhang JF, Yang ZJ, et al. Involvement of Drp1 367: 108–112.
in hypoxia-induced migration of human glioblastoma U251 72. Arismendi-Morillo G. Electron microscopy morphology of the
cells. Oncol Rep 2014; 32: 619–626. mitochondrial network in gliomas and their vascular microen-
63. Choudhary V, Kaddour-Djebbar I, Lakshmikanthan V, et al. vironment. Biochim Biophys Acta 2011; 1807: 602–608.
Novel role of androgens in mitochondrial fission and apop- 73. Chiang YY, Chen SL, Hsiao YT, et al. Nuclear expression
tosis. Mol Cancer Res 2011; 9: 1067–1077. of dynamin-related protein 1 in lung adenocarcinomas. Mod
64. Sastre-Serra J, Nadal-Serrano M, Pons DG, et al.
Pathol 2009; 22: 1139–1150.
Mitochondrial dynamics is affected by 17β-estradiol in 74. Wieder SY, Serasinghe MN, Sung JC, et al. Activation of
the MCF-7 breast cancer cell line. Effects on fusion and the mitochondrial fragmentation protein DRP1 correlates
fission related genes. Int J Biochem Cell Biol 2012; 44: with BRAFV600E melanoma. J Invest Dermatol 2015; 135:
1901–1905. 2544–2547.

You might also like