ArticlePDF AvailableLiterature Review

Autophagy and Hallmarks of Cancer

Authors:

Abstract

Autophagy is a catabolic program that is responsible for the degradation of dysfunctional or unnecessary proteins and organelles to maintain cellular homeostasis. Mechanistically, it involves the formation of double-membrane autophagosomes that sequester cytoplasmic material and deliver it to lysosomes for degradation. Eventually, the material is recycled back to the cytoplasm. Abnormalities of autophagy often lead to human diseases, such as neurodegeneration and cancer. In the case of cancer, increasing evidence has revealed the paradoxical roles of autophagy in both tumor inhibition and tumor promotion. Here, we summarize the context-dependent role of autophagy and its complicated molecular mechanisms in the hallmarks of cancer. Moreover, we discuss how therapeutics targeting autophagy can counter malignant transformation and tumor progression. Overall, the findings of studies discussed here shed new light on exploiting the complicated mechanisms of the autophagic machinery and relevant small-molecule modulators as potential antitumor agents to improve therapeutic outcomes.
0893-9675/18/$35.00 © 2018 by Begell House, Inc. www.begellhouse.com
Critical Reviews™ in Oncogenesis 23(5-6) 247–267 (2018)
247
Autophagy and Hallmarks of Cancer
Tianzhi Huang,a Xiao Song,a Yongyong Yang,a Xuechao Wan,a Angel A. Alvarez,a Namratha
Sastry,a Haizhong Feng,b Bo Hu,a,* & Shi-Yuan Chenga,*
aKen & Ruth Davee Department of Neurology, Lou & Jean Malnati Brain Tumor Institute, Robert H. Lurie
Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL; bState Key
Laboratory of Oncogenes and Related Genes, Renji-Med X Clinical Stem Cell Research Center, Ren Ji Hospital,
School of Medicine, Shanghai Jiao Tong University, Shanghai, China
*Address correspondence to: Shi-Yuan Cheng, PhD, Ken & Ruth Davee Department of Neurology, Lou & Jean Malnati Brain Tumor Institute,
Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, 303 E. Superior, Lurie 6-119, Chicago, IL
60611, E-mail: shiyuan.cheng@northwestern.edu; and to Bo Hu, PhD, Ken & Ruth Davee Department of Neurology, Lou & Jean Malnati Brain
Tumor Institute, Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, 303 E. Superior, Lurie
6-115, Chicago, IL 60611, E-mail: bo.hu@northwestern.edu.
ABSTRACT: Autophagy is a catabolic program that is responsible for the degradation of dysfunctional or unnecessary
proteins and organelles to maintain cellular homeostasis. Mechanistically, it involves the formation of double-membrane
autophagosomes that sequester cytoplasmic material and deliver it to lysosomes for degradation. Eventually, the material
is recycled back to the cytoplasm. Abnormalities of autophagy often lead to human diseases, such as neurodegeneration
and cancer. In the case of cancer, increasing evidence has revealed the paradoxical roles of autophagy in both tumor inhi-
bition and tumor promotion. Here, we summarize the context-dependent role of autophagy and its complicated molecular
mechanisms in the hallmarks of cancer. Moreover, we discuss how therapeutics targeting autophagy can counter malig-
nant transformation and tumor progression. Overall, the ndings of studies discussed here shed new light on exploiting
the complicated mechanisms of the autophagic machinery and relevant small-molecule modulators as potential antitumor
agents to improve therapeutic outcomes.
KEY WORDS: therapy, autophagosomes, ATGs, cancer, macroautophagy
ABBREVIATIONS: ATG, autophagy-related protein; mTORC1, mammalian target of rapamycin complex 1; ULK1, unc-51-like
autophagy-activating kinase 1; TFEB, transcription factor EB; UBL, ubiquitin-like protein; EMT, epithelial-mesenchymal transi-
tion; CSC, cancer stem cell; CQ, chloroquine
I. INTRODUCTION
Macroautophagy (hereafter referred to as autopha-
gy) is a conserved process by which eukaryotic cells
sequester intracellular material including damaged
organelles, redox-active protein aggregates, and for-
eign matter within double-membraned vesicles (also
known as autophagosomes). These vesicles are then
transported to the lysosome for degradation.1,2 The
autophagic network is a multilayer cellular process
that is tightly controlled by a set of autophagy-rel-
evant factors, including (but not limited to) ATG5,
ATG7, and BECN1.3 Under normal physiological
conditions, baseline levels of autophagy mediate a
key homeostatic function, essentially operating as
an intracellular quality control system. Autophagy
can be upregulated in response to a variety of in-
trinsic or extrinsic stimuli: starvation, growth factor
deprivation, hypoxia, pathogens, and many others.4,5
Autophagy is a critical regulator of cellular homeo-
stasis, and autophagic dysfunction is associated with
human diseases, including Alzheimer’s disease, Par-
kinson’s disease, and cancers.6,7 Of note, autophagy
has complex and context-dependent roles in cancer.
In some contexts, autophagy has a preventive role
during cancer development. Moreover, in most con-
texts autophagy promotes tumorigenesis.7,8 In this
review, we summarize the mechanism of autopha-
gy and its paradoxical roles in cancer. Importantly,
we discuss the molecular and cellular mechanisms
whereby the autophagic network interfaces with
multiple hallmarks of cancer. Finally, we illustrate a
Huang et al.
Critical Reviews™ in Oncogenesis
248
series of molecules and agents targeting autophagy
executors for potential cancer treatments.
II. MECHANISMS OF AUTOPHAGY
The autophagic process involves the sequential
formation of the phagophore (also known as the
isolation membrane), autophagosome, and autol-
ysosome, ultimately leading to the degradation
and recycling of autophagic cargo.9 The molecular
machinery of autophagy is highly conserved from
yeasts to mammals and executed by a group of
autophagy-related proteins (ATGs) that associate
with other proteins to form different complexes in
autophagic ux.9
A. Autophagy Initiation and Phagophore
Nucleation
In yeast, autophagy is initiated at the phagophore
assembly site (PAS).10 Contrarily in mammals,
phagophores are nucleated at an endoplasmic retic-
ulum (ER) -emanating membrane domain enriched
for the lipid phosphatidylinositol 3-phosphate
(PI3P), known as the omegasome.11 Signals that
activate the autophagic process typically originate
from various stressors, including energy or nutrient
shortage, hypoxia, oxidative stress, and other cy-
totoxic insults. The best characterized triggers for
autophagy induction include inactivation of mam-
malian target of rapamycin complex 1 (mTORC1)
under amino acid deprivation12,13 and activation
of AMP-activated protein kinase (AMPK), which
senses changes in the AMP:ATP ratio resulting
from energy starvation.14,15 These two kinases regu-
late autophagy initiation by controlling the activa-
tion of the unc-51-like autophagy-activating kinase
1 (ULK1) complex (known as the Atg1 complex
in yeast), which consists of serine/threonine kinase
ULK1, scaffolding subunit FIP200 (FAK family-
interacting protein of 200 kDa), and regulatory
subunits ATG13 and ATG101.16 In the absence of
starvation (hereafter referred to as fed), mTORC1
binds the ULK1 complex and phosphorylates
both ULK117 and ATG13,18 thereby suppressing
the ULK1 complex function. Upon energy deple-
tion, AMPK directly activates the ULK1 complex
by phosphorylation at multiple sites in the central
intrinsically disordered region (IDR).17,19,20 Addi-
tionally, AMPK indirectly induces autophagy by
inhibiting mTORC1 through phosphorylation of
the regulatory-associated protein of mTOR (RAP-
TOR).21
The activated ULK1 complex then triggers
nucleation of the phagophore by activating down-
stream the Class III phosphoinositide 3-kinase
complex I (PI3KC3-C1). PI3KC3-C1 is another
pivotal autophagy-initiating complex, and consists
of phosphoinositide (PI) kinase vacuolar protein
sorting 34 (VPS34), regulatory subunit BECN1
(Atg6 in yeast), scaffold protein VPS15, and ER-
targeting protein ATG14L.16 ULK1 directly phos-
phorylates VPS34 and BECN1,22 thereby activat-
ing the PI3KC3-C1 complex, and in turn generates
local phosphatidylinositol-3-phosphate (PI3P) in
the phagophore.23 This autophagosome-specic
pool of PI3P is essential for phagophore nucle-
ation.24,25 PI3P promotes recruitment of additional
autophagy-specic PI3P effectors, such as WD-
repeat domain PI-interacting protein 2 (WIPI2).
WIPI2 subsequently recruits the ATG12–ATG16L
conjugation complex for phagophore expansion.26
B. Phagophore Elongation and
Autophagosome Completion
From initiation at the PAS or omegasome, the
phagophore elongates into a cup-shaped structure
and begins to engulf cellular material. In the ex-
pansion stage, two ubiquitin-like protein (UBL)
conjugation systems are required for the elongation
of the phagophore membrane: the ATG12–ATG5
UBL and the microtubule-associated light-chain B
(LC3B, the mammalian orthologue of yeast Atg8)-
phosphatidylethanolamine (PE) UBL.27 ATG12 is a
ubiquitin-like protein, which covalently attaches to
substrates via the carboxyl group of its C-terminal
glycine.28 The ATG12–ATG5 conjugation is medi-
ated by the E1-like enzyme ATG7 and the E2-like
enzyme ATG10.29,30 The ATG12–ATG5 conjugate
then forms a supermolecular complex with AT-
G16L1. This complex is recruited to the phagophore
Authophagy and Hallmarks of Cancer 249
Volume 23, Issues 5-6, 2018
by WIPI2,26 where it acts as an E3-like enzyme for
the LC3-PE UBL.31 Nascent pro-LC3 is cleaved by
the cysteine protease ATG4B, thereby exposing a
glycine residue for its conjugation with PE.32,33 The
processed LC3 is then conjugated to membrane-as-
sociated PE by the E1-like enzyme ATG7 and the
E2-like enzyme ATG3 in concert with the ATG12–
ATG5–ATG16L complex, converting it from a dif-
fuse form (LC3-I) to a membrane-anchored, lipidat-
ed form (LC3-II).34,35 LC3 lipidation is required for
phagophore expansion and closure36,37 and is widely
used as an autophagosome marker. The autophagas-
ome maturation involves the clearance of LC3/Atg8
and PI3P from the autophagasome outer membrane
by ATG4 and YMR1, respectively. Following this
dissociation, the autophagosome can fuse with the
lysosome.38
C. Lysosomal Fusion and Degradation of
Cargo
After formation of the autophagosome is complete,
its outer membrane fuses with the lysosome (vacu-
ole in yeast) to form the structure known as the
autolysosome.39 The autophagosome–lysosome fu-
sion involves the function of three protein families:
Rab GTPases, membrane-tethering factors (such as
HOPS and EPG5), and soluble N-ethylmaleimide-
sensitive factor attachment proteins (SNAREs).
Rab GTPases are located at specic membranes,
which then recruit tethering complexes that act as
bridges to tether the opposing lipid bilayers. These
tethering complexes in turn recruit and promote
SNARE proteins to physically drive fusion of the
autophagosome with the lysosome.40 Additionally,
recent studies have revealed the critical roles of
ATG8 family members in the positioning of au-
tophagosomes near lysosomes. Moreover, these
proteins assist in the assembly of the fusion ma-
chinery, which highlights them as possible hubs for
the coordination of the nal fusion stages of au-
tophagy.41 Degradation of the autophagic cargo is
dependent on a series of lysosomal acid hydrolas-
es. Salvaged nutrients are then released back to the
cytoplasm for protein synthesis and maintenance
of cellular homeostasis.
III. EPIGENETIC, TRANSCRIPTIONAL, AND
POST-TRANSCRIPTIONAL REGULATION
OF AUTOPHAGY
A. Epigenetic Regulation of Autophagy
Accumulating evidence indicates that epigenetic
modications are involved in the control of autoph-
agy.42–53 Post-translational modications of histones
and DNA methylation may affect the chromatin
structure, which leads to the dysregulation of a se-
ries of autophagy-related genes.43–45,48,49 Several his-
tone modications, including H3R17 dimethylation
(me2),54 H3K9me2,45,55,56 H3K27 trimethylation
(me3),48 and H4K16 deacetylation,57 are linked to
autophagy in human cancers.
Nutrient starvation activates the AMPK-fork-
head box protein O3a (FOXO3a) axis to suppress
SKP2. SKP2 repression upregulates protein ar-
ginine methyltransferase CARM1 protein lev-
els.54 The stabilization of CARM1 induces global
H3R17me2 levels in nutrient starvation–induced
autophagy.54 Recent studies have indicated that
H3K9me245,56,58 and H3K27me348 act as repressors
of autophagy. Following autophagy induction,
the G9a methyltransferase and G9a-mediated
H3K9me2 are removed from the target gene pro-
moters, leading to upregulation of autophagy
regulators such as LC3B and sequestosome 1
(SQSTM1/p62).45 EZH2-mediated H3K27me3
epigenetically represses autophagy by downregu-
lating mTOR pathway suppressors such as TSC2,
RGS16, GPI, and RHOA.48 Moreover, EZH2 in-
hibitors such as GSK34359 and UNC199960 signif-
icantly induce autophagy in cancer cells. In addi-
tion to methylation, histone acetylation contributes
to autophagy in tumorigenesis. Downregulation of
H4K16ac is associated with autophagy induction
in human cancer cells, which is mediated by the
histone acetyltransferase hMOF.57 Deacetylation
of H4K16 widely affects autophagy-related gene
expression.57 Other histone modications such as
H2BK120 monoubiquitination,51 H3K56 acetyl-
ation,43,61 and H4K20 methylation43,62 are also in-
volved in autophagy regulation.
Huang et al.
Critical Reviews™ in Oncogenesis
250
B. Transcriptional Regulation of
Autophagy
Emerging studies have demonstrated the key role
played by transcription factors in regulating au-
tophagy in cancer cells.54,63–69 STAT3 is the one of
the most important transcriptional regulators of au-
tophagy-related genes. STAT3 represses BECN1
expression by directly binding to its promoter and
recruiting HDAC3.70 STAT3 also regulates auto-
phagy through PIK3R1 regulation. STAT3 suppress-
es autophagy by transcriptionally upregulating the
PIK3R1 gene products p55α and p50α, which inhib-
it p85α-mediated autophagy.71 Recent studies have
demonstrated that STAT3 also acts as a transcrip-
tional activator of BCL2, BCL2L1, and MCL1,72,73
which are key autophagy-related regulators. Interest-
ingly, both nuclear STAT3 and cytoplasmic STAT3
suppress autophagy in cancer cells. Cytoplasmic
STAT3 directly interacts with EIF2AK2 to inhibit its
enzymatic activity, resulting in decreased autophagy
activator eukaryotic translation initiation factor q al-
pha kinase 2A (EIF2A) phosphorylation.74,75 Further-
more, several STAT3 regulating noncoding RNAs
are involved in autophagy regulation. For example,
the long noncoding RNA HAGLROS, a direct target
of STAT3,76,77 activates the mTOR pathway to pro-
mote autophagy suppression.77
Transcription factor EB (TFEB), a member
of the microphthaimia family, has been reported
to be a key regulator of lysosomal biogenesis and
autophagy.78 Nutrient starvation induces TFEB
nuclear translocation. Nuclear-localized TFEB di-
rectly transcriptionally induces WIPI, VPS11, and
VPS18 in starvation-induced autophagy.78 TFEB
nuclear translocation upregulates autophagy-
related regulators, including ATP6V0D1, LAMP1,
CTSB, MAP1LC3B, and UVRAG.79 Of note, emerg-
ing studies have demonstrated that TFEB serves as
a bridge to mediate upstream regulators and au-
tophagy progression in cancer cells. STUB1 regu-
lates autophagy and mitochondrial biogenesis by
modulating TFEB activity.80 In addition, MAP4K3,
a novel autophagy regulator,81 regulates TFEB
nuclear localization and transcriptional activity by
phosphorylating TFEB at serine 3.81 Several other
genes, such as PEG3,82 AKT,83 and BRD4,84 are
also involved in autophagy regulation by inuenc-
ing TFEB transcription activity.
C. Post-Transcriptional Regulation of
Autophagy
MicroRNAs (miRNAs) are small noncoding RNAs
that are 18–25 nucleotides in length. miRNAs serve
as post-transcriptional regulators of protein-coding
genes by binding to the 3ʹ-untranslated region (UTR)
of target messenger RNAs (mRNAs).85 Emerging
studies report that miRNAs play important roles in
the regulation of autophagy.86–90 miR-30a was the
rst miRNA shown to be involved in autophagy reg-
ulation by targeting BECN1.91 A similar observation
in human chronic myeloid leukemia cells showed
that miR-30a inhibits autophagy by targeting both
BECN1 and ATG5.92 miR-101 is also an autophagy
suppressor.93–95 miR-101-mediated inhibition of au-
tophagy partially depends on three novel targets,
STMN1, RAB5A, and ATG4D.96 Similarly, miR-
376b regulates starvation and mTOR inhibition–re-
lated autophagy through inhibition of ATG4C and
BECN1.97 Finally, miR-23b regulates autophagy to
promote radioresistance in pancreatic cancer cells
by targeting ATG12.98
Unlike miRNAs, long noncoding RNAs (ln-
cRNAs) are RNA transcripts longer than 200
nucleotides with no protein-coding potential.99
Increasingly, reports show the importance of ln-
cRNAs in tumorigenesis by regulating protein
levels through transcriptional, post-transcriptional,
and post-translational levels.99 Since 2013, sev-
eral studies have implicated lncRNAs in autophagy.
APF lncRNA binds and inhibits miR-188-3p. In the
absence of APF lncRNA, miR-188-3p functions
to bind and suppress translation of ATG7 mRNA,
an autophagy-promoting gene. Thus, APF lncRNA
works to inhibit miR-188-3p, thereby disinhibiting
ATG7 gene transcription.100 Knockdown of APF
lncRNA signicantly suppresses ATG7 expression
and autophagy. Additionally, lncRNA HULC, which
is highly upregulated in liver cancer, promotes
autophagy via stabilizing SIRT1 in hepatocellular
carcinoma.101 Finally, MEG3, the most well-known
Authophagy and Hallmarks of Cancer 251
Volume 23, Issues 5-6, 2018
lncRNA involved in autophagy regulation,102–106
interacts with ATG3 mRNA and protects it from
degradation.104 Taken together, these ndings sug-
gest that miRNAs and lncRNAs are important post-
transcriptional regulators of autophagy and act by
regulating autophagy-related genes.
IV. BIPOLAR NATURE OF AUTOPHAGY IN
CANCER
A. Tumor Suppression by Autophagy
Autophagy has been universally demonstrated to
play a tumor-suppressive role at the benign stage,8
and defective autophagy has been connected with
DNA damage and tumorigenesis.7,107
BECN1 is the mammalian orthologue of the
yeast Atg6 gene.108 BECN1 interacts with either
BCL-2 or PI3K Class III (VPS34),109,110 playing
a critical role in the regulation of both autophagy
and cell death.109,111 Adult mice having monoallelic
deletion of Becn1 (BECN1+/−) showed increased
DNA damage and a higher incidence of spontane-
ous lung cancer, liver cancer, and lymphomas.112,113
Allelic deletion of Becn1 has been reported in other
tumor types, such as prostate, breast, and ovar-
ian cancers.108,114 BECN1 is positively regulated
by the ultraviolet radiation resistance–associated
gene (UVRAG) and Bax-interacting factor 1 (BIF-
1). Both of these proteins enhance the interaction
between BECN1 and VPS34, leading to increased
autophagy.115,116 Mutations in UVRAG and low ex-
pression of BIF-1 have been observed in several
types of cancers.116–118 Moreover, mice with system
mosaic deletion of Atg5 or liver-specic Atg7 de-
ciency also develop liver tumors.119 Together, these
ndings suggest that autophagy plays a key role in
repressing tumorigenesis.
Mitophagy is the selective degradation of dam-
aged mitochondria by autophagy. Dysfunctional
mitochondria promote activation of PTEN-induced
putative kinase 1 (PINK1), which further activates
the E3 ligase parkin (encoded by PARK2) to ubiq-
uitinate mitochondrial substrates, resulting in the
selective degradation of damaged mitochondria by
the autophagy machinery.120,121 Mitophagy helps to
maintain mitochondrial quality and reduce oxidative
stress. Park2 has been shown to function as a tumor
suppressor gene.122 Like Becn1 deletion, Park2 de-
letion in mice leads to increased hepatocellular car-
cinoma,123 implying that defective mitophagy and
oxidative stress contribute to tumor tumorigenesis.
p62, a prominent autophagy substrate, is an
adaptor protein that possesses various binding
motifs. It functions by recruiting proteins and as-
sembling them into complexes.124 Nuclear factor
(erythroid-derived 2) -related factor 2 (NRF2) is
activated by p62125 and is responsible for activating
the transcription of antioxidant defense genes.126 In
the absence of cellular stress, kelch-like ECH–as-
sociated protein 1 (KEAP1), a component of the
CUL3–RBX1 E3 ligase complex, binds and in-
hibits NRF2 activity. However, in the presence of
oxidative stress, p62 expression increases. Thus,
p62 competitively binds KEAP1, thereby releas-
ing NRF2. NRF2 then translocates to the nucleus
and activates the expression of antioxidant defense
genes, promoting cell survival and tumorigenesis.125
Autophagy deciency via liver-specic deletion of
Atg7 in mice results in p62 accumulation and NRF2
activation, increasing the expression of NRF2-target
genes. Liver tumors originate from autophagy-de-
cient hepatocytes, which can be partially suppressed
by p62 deletion.119 Deciency in p62 or NRF2 sup-
presses the development of Ras-driven non-small-
cell lung cancer in mouse models.127,128 Furthermore,
activating mutations of NRF2 and inactivation mu-
tations of its negative regulator, KEAP1, are found
in various types of cancers, implying that p62 and
NRF2 function as oncogenes while KEAP1 func-
tions as a tumor suppressor.129–131
B. Tumor Promotion by Autophagy
Although autophagy inhibits early tumor initiation
and growth, the principal effect of autophagy is to
promote tumor growth. In many cases, cancer cells
show increased autophagy dependency than normal
cells. This context-dependent nature of autophagy
likely results from the elevated metabolic and bio-
synthetic demands of dysregulated proliferating
cancer cells.
Huang et al.
Critical Reviews™ in Oncogenesis
252
Basal levels of autophagy are essential for nor-
mal tissue homeostasis.132 Autophagy was initially
shown to support the survival of yeast under star-
vation conditions through maintaining amino acid
levels and activating the expression of genes in re-
sponse to starvation.133,134 Deletion of Atg5 or Atg7
in mouse brains causes polyubiquitinated protein
accumulation and leads to neurodegeneration.135,136
This suggests that clearance of abnormal proteins
by autophagy is crucial for the survival of neurons.
Becn1−/− mice are embryonic lethal.112 Neonatal
Atg5-decient mice survive for a much shorter time
than wild-type mice. Moreover, Atg5-decient neo-
nates display signs of reduced amino acid concen-
tration and ATP levels,137 suggesting that they suffer
from a metabolic crisis. These ndings support an
important and conserved role for autophagy in nor-
mal cell survival in response to metabolic stress.
The major function of autophagy is to collect,
degrade, and recycle intracellular material when
cells are in starvation. Autophagy can supply mi-
tochondrial substrates in the form of amino and
fatty acids, further promoting tumor cell growth.
Impaired autophagy in cancer cells leads to the ac-
cumulation of morphologically and functionally ab-
normal mitochondria.119 Experimentally induced au-
tophagy deciency in tumor cells results in decient
ATP and lack of key tricarboxylic acid (TCA) -cycle
intermediates, and leads to mitochondrial dysfunc-
tion. Mitochondrial dysfunction is mainly exempli-
ed by generation of toxic reactive oxygen species
(ROS) and mitochondrial damage, promoting accu-
mulation of damaged mitochondria resulting from
a failure of clearance by mitophagy.138,139 However,
the precise mechanisms of autophagy’s support of
mitochondrial function are still under investigation.
Autophagy is vigorously stimulated by vari-
ous stressors in cancer cells to support their high
metabolic demands. In conditions in which apop-
tosis is inhibited, autophagic cancer cells enter a
state of quiescence, which allows them to survive
for weeks. When metabolites and nutrients become
readily available again, these cells are restored to
their normal growth conditions.140 Glucose depriva-
tion and hypoxia, typical physiological stresses in
the tumor microenvironment, activate autophagy in
cancer cells to support survival.141,142 Pharmacologi-
cal inhibition of autophagy or deletion of essential
autophagy genes suppresses cancer cell growth in
normal and stress conditions.142,143
A large number of cancer cell lines have a high
basal level of autophagic activities even without
stressors. Several oncogenes (e.g., RAS and BRAF)
that promote cancer cell growth also increase the
basal level of autophagy.144,145 It is hypothesized that
both RAS and BRAF function using similar mecha-
nisms. Activating mutations in RAS and BRAF, al-
though frequent in cancer, are mutually exclusive, as
concurrent mutations arising in both genes are ex-
tremely rare.146 Ras-activated cancer cells are highly
dependent on autophagy to support their survival
under basal and especially stressful conditions.145 K-
RAS translocates into the mitochondria and causes a
disruption of Complex I. K-RAS activation signi-
cantly suppresses cancer cell mitochondrial respira-
tion and impairs acetyl-CoA production. This results
in increased dependency on autophagy to provide the
necessary substrates for acetyl-CoA biosynthesis to
promote TCA cycle activation.145,147 Genetic or phar-
macologic inhibition of autophagy in cancer cells
with K-RAS activation causes increased ROS and
DNA damage and decreased mitochondrial oxida-
tive phosphorylation,139,145 resulting in robust tumor
suppression.139 Human non-small-cell lung cancer
cells with BRAF activation mutations display high
levels of autophagic activity. Inhibition of autophagy
through Atg7 deciency (Atg7−/−) suppresses the pro-
gression of BrafV600E-driven tumors and accumulates
defective mitochondria.144 Overall, this implies that
the dysregulated mitochondrial metabolism caused
by impaired autophagy promotes tumor growth.
V. AUTOPHAGY AND THE HALLMARKS OF
CANCER
A. Autophagy Sustains Proliferation
One of the key characteristics of cancer cells is sus-
tained proliferation.148 Normal cells have precise con-
trols of cell cycle progression to maintain proper cell
growth and function. However, tumor cells obtain
unlimited proliferating potential by bypassing cell
Authophagy and Hallmarks of Cancer 253
Volume 23, Issues 5-6, 2018
cycle check points, due to mutations in genes such as
TP53 and Retinoblastoma (Rb).148 Basal levels of au-
tophagy in normal tissue maintain genome integrity
and prevent tumorigenesis.7,107 Autophagy deciency
by Atg5 or Becn1 deletion (Atg5−/− or Becn1−/−) causes
early death in mice.112,137 Moreover, mice with mo-
saic deletion of Atg5 or monoallelic deletion of Becn1
show a high incidence of liver cancer.112,119 In cancer
cells, the PI3K/AKT pathway is often activated to
promote cell proliferation through PI3K-activating
mutations, increased expression of AKT, EGFR over-
expression, HER-2 amplication, or PTEN loss, fol-
lowed by mTOR activation. Interestingly, mTORC1
is a major negative regulator of autophagy.149–151
BCL2 that is often overexpressed in cancer cells
also inhibits autophagy through inhibitory binding to
BECN1.110,111 TP53 is the most commonly mutated
gene in human cancers and encodes the p53 protein.
Activation of p53 via starvation or DNA damage ac-
tivates autophagy, which may occur via inhibition of
mTOR.152,153 These ndings suggest that autophagy
suppresses cancer cell growth, which is consistent
with its role as tumor suppressor.
However, as discussed previously, the role of
autophagy in cancer is contextual. High levels of
autophagy are also observed in RAS-BRAF-driven
cancer cells, and autophagy is essential for these tu-
mor cells to grow under both normal and starvation
conditions.144,145 Impaired autophagy in these tumors
greatly inhibits tumor cell growth and tumor pro-
gression, revealing the role of autophagy in sustain-
ing tumor cell proliferation. Taken together, these
cases show that the complexity of tumors and the
role of autophagy in regulating tumor cell prolifera-
tion are highly context-dependent.
B. Autophagy Promotes Epithelial–
Mesenchymal Transition
Epithelial–mesenchymal transition (EMT), another
hallmark of cancer, is the process by which epithe-
lial cells lose their adhesive properties and become
more migratory and invasive mesenchymal cells.
Autophagy and EMT both play crucial roles in hu-
man cancer progression.154,155 However, the inter-
play between autophagy and EMT remains unclear.
Here, we focus on elucidating the intricate relation-
ship between them.
Autophagy is an important mechanism by which
cancer cells evade apoptosis and is a prerequisite
of tumor metastasis.156,159 Previous studies have
revealed that autophagy is positively correlated to
cancer cell EMT. Autophagy can activate hepato-
cellular carcinoma (HCC) cell EMT, which can
subsequently promotes cell invasion.160 Autophagy
impairment through chloroquine (CQ) treatment
or Atg3 and Atg7 silencing inhibits EMT in HCC
cells.160 Moreover, knockdown of BECN-1, a key
autophagy activator, remarkably suppresses EMT
in colon cancer cells.161 Interestingly, ULK2, which
phosphorylates the BECN-1 initiation complex,
may also promote EMT in lung cancer cells.162 Sim-
ilarly, cisplatin treatment activates autophagy in na-
sopharyngeal carcinoma, and autophagy inhibition
impairs EMT in this progression.163
Contrary to these ndings, a number of studies
have shown negative crosstalk between autophagy
and EMT in cancer cells, which is likely due to the
bipolar nature of autophagy. Several reports have
shown that anticancer agents, such as the Aurora ki-
nase inhibitors alisertib164,165 and danusertib,166,167 ac-
tivate autophagy but supress EMT in cancer cells. In
ovarian cancer, autophagy inhibition through Atg7
knockdown promotes EMT via activation of the
ROS/Heme Oxygenase-1 (ROS/HO-1) pathway.168
Similarly, knockdown of ATG5, ATG7, or BECN-
1 in glioblastoma induces autophagy impairment,
which leads to promotion of cancer cell by induc-
ing EMT.169 The direct interaction between CDH6
and GABARAP as well as BNIP3 and BNIP3L in
thyroid cancer suppresses autophagy to promote
EMT.170 TWIST, the basic helix-loop-helix tran-
scription factor, is the hallmark of EMT and plays
crucial roles in cancer metastasis.171,172 Recent stud-
ies have indicated that the degradation of TWIST
underlies autophagy-inhibiting EMT in cancer cells.
The death effector domain–containing DNA-bind-
ing protein (DEDD) directly interacts with PI3KC3/
BECN-1 to induce autophagy-mediated lysosomal
degradation of SNAIL and TWIST,173 which conse-
quentially inhibit EMT. Finally, autophagy impair-
ment stabilizes TWIST1 to promote EMT.174
Huang et al.
Critical Reviews™ in Oncogenesis
254
Recent studies indicate that p62 and TGF-β me-
diate crosstalk between autophagy and EMT. Ear-
lier studies revealed that p62, an autophagy adap-
tor protein, may bind to EMT regulators to affect
EMT progression. For example, p62 sustains a level
of HDAC6 to promote EMT in prostate cancer.175
p62 also interacts with TWIST to promote EMT
by inhibiting its degradation.174 Similarly, p62 in-
creases EMT in cancer cells by stabilizing SMAD4
and TWIST,176and it has been shown to increase the
transcriptional activity of NF-κB to promote EMT
by enhancing the nuclear translocation of p65.177
TGF-β signaling is the most important regulator
of EMT in human cancers.178,179 It has been shown
that TGF-β plays a crucial role in crosstalk between
EMT and autophagy by promoting autophagy in
cancers. TGF-β treatment induces autophagy in
hepatocellular carcinoma (HCC)160,180 by induc-
ing autophagy regulators such as BECN-1, ATG5,
and ATG7.180 Interestingly, autophagy impairment
by CQ or ATG5 knockdown suppresses TGF-β2–
induced EMT.181 Additionally, TGF-β2 activates
autophagy via the SMAD and JNK pathways in
glioma cell lines.182 Autophagy inhibition blocks
TGF-β2–induced EMT by inuencing mitochon-
drial trafcking and membrane potential.
C. Role of Autophagy in Tissue Invasion
and Metastasis
Tumor cells have the capability for local invasion
and distant metastasis. The cascade of tumor in-
vasion and metastasis can be divided into a series
of steps: local invasion, intravasation into nearby
blood and lymphatic vessels, transit of cancer cells
through the circulatory system, extravasation, for-
mation of small nodules of cancer cells, and nally
outgrowth of micrometastatic lesions into macro-
scopic tumors. This process requires the transloca-
tion of cancer cells to new microenvironments, in
which metastatic tumor cells must overcome numer-
ous challenges to survive. These include altered nu-
trient supply, immune surveillance, and T-cell–me-
diated killing mechanisms.183,184 Given the key role
of autophagy as an adaptive response to stress, its
involvement in the different stages of the metastatic
cascade has been postulated.159 Indeed, autophagic
ux has been found to be upregulated by the differ-
ent environmental stressors that promote invasion
and metastasis of cancer cells, such as hypoxia and
nutrient deprivation.185 In addition, recent studies
have identied an association between increased
punctate staining for LC3B and metastasis of vari-
ous cancers, including human breast cancer, mela-
noma, and HCC.186–188
Autophagy can either promote or impede cell
invasion and metastasis. At the initial steps of me-
tastasis, autophagy promotes the survival of tumor
cells from hypoxia and metabolic stress by reducing
tumor cell necrosis. Consequently, autophagy re-
duces the inltration of macrophages at the primary
tumor site, which is required for the initiation of
metastasis.189,190 During detachment from the extra-
cellular matrix, autophagy is induced in tumor cells
to overcome anoikis, a type of cell death signal trig-
gered by the absence of anchorage to the extracel-
lular matrix.191,192 A connection between autophagy,
EMT, and invasion has also been seen in HCC and
lung cancer cells, where autophagy is critical for
EMT and invasion of tumor cells.160,162
Upon successful extravasation, autophagy is
proposed to facilitate tumor cell dormancy by pro-
moting quiescence, a process that is also necessary
for the maintenance of cancer stem cells.193 Indeed,
autophagy has been shown to be induced by the tu-
mor suppressor gene aplasia Ras homolog member I
(ARHI) to increase tumor cell dormancy.194 The dor-
mancy of tumor cells has also been suggested to re-
sult in resistance to genotoxic therapies that primar-
ily target proliferating cells.195 If autophagy is truly
required for dormancy, the combination of genotoxic
therapy with autophagy inhibition comes into focus
as a therapeutic option to eliminate dormant tumor
cells and thereby limit metastatic disease.
The studies previously described suggest that
autophagy prevents initiation of invasion and me-
tastasis by preventing tumor necrosis and inamma-
tion. However, once the tumor cells have entered a
new microenvironment, autophagy helps them sur-
vive and maintain dormancy until they successfully
establish distant colonies. Therefore, autophagy
may be regulated differently at the various stages of
Authophagy and Hallmarks of Cancer 255
Volume 23, Issues 5-6, 2018
metastasis in a way that has signicance for the use
of autophagy modulators in cancer therapy.
D. Autophagy Reprograms Tumor
Metabolism
It has been established that tumor cells have to
change their metabolic pathways to meet the height-
ened metabolic requirements necessary for tumor
survival and unconstrained proliferation.196 To ac-
complish these goals, tumor cells have been shown to
use various sources of energy. Indeed, some tumors
employ increased aerobic glycolysis rather than oxi-
dative phosphorylation to generate the biosynthetic
intermediates required for proliferation (termed the
Warburg effect).197 This is an acquired property of
tumors caused by the impaired mitochondrial me-
tabolism that also helps tumor cell survival in hy-
poxic microenvironments caused by dysfunctional
vasculature. In the absence of pyruvate, other sub-
strates are required to undergo the tricarboxylic
acid (TCA) cycle for ATP synthesis, and autophagy
can provide these substrates by recycling intracel-
lular macromolecules.198 Autophagy can lead to the
degradation of various substrates, thereby providing
metabolites for numerous metabolic pathways.199
For instance, autophagy can provide sugars and nu-
cleosides for glycolysis by degrading carbohydrates
and DNA, respectively. It can also provide metabo-
lites for the TCA cycle by degrading proteins and
lipids. Therefore, autophagy plays an important role
in reprogramming metabolic pathways to promote
tumor cell survival.
In addition to cancer metabolism, autophagy
also functions in the metabolic crosstalk between
tumor cells and other stromal components, such as
pancreatic stellate cells.200 Autophagy is required for
stellate cells to secrete metabolic substrates, such as
the nonessential amino acid alanine, to support pan-
creatic cancer mitochondrial metabolism.201
E. Implications of Autophagy in Cancer
Stem Cells
Cancer stem cells (CSCs) are dened as a small
subset of cancer cells within a tumor that can self-
renew and generate heterogeneous lineages of can-
cer cells that make up the tumor.202,203 CSCs are in-
trinsically resistant to conventional chemotherapy
and radiation treatment, and are postulated to con-
tribute to treatment failure and tumor recurrence.204
Therefore, targeting CSCs represents a useful strat-
egy to improve the effectiveness of therapeutic in-
terventions.
The involvement of autophagy in the physiol-
ogy of CSCs is complicated and is not yet fully
elucidated. Accumulating evidence suggests that
autophagy plays critical roles in the maintenance
and function of various normal stem cells. Given
the similarities between normal stem cells and
CSCs, it is expected that autophagy may be cru-
cial in the maintenance and function of CSCs. The
expression of the CSC marker CD133 is positively
correlated with the expression of autophagy-related
proteins ATG5, ATG12, and LC3. Autophagy in-
duction markedly enhances the radiation resistance
of CD133+ glioma stem cells (GSCs).205 In CD44+/
CD24−/low breast CSCs (BCSCs), ATG4 regulates
BCSC populations by promoting their self-renewal
in vitro and tumor growth in vivo.206,207 Along
similar lines, CQ-mediated autophagy inhibition
depleted the CD44+/CD24–/low BCSC population in
triple-negative breast cancer in both preclinical and
clinical settings.208 Moreover, inhibition of autopha-
gy by ATG4B knockdown altered GSC phenotypes.
Inhibition of ATG4B using a specic antagonist,
NSC185058, sensitized GSCs to gamma-irradiation
and reduced their capability to form glioma tumor
spheres. These ndings illustrate that autophagy is a
crucial regulator of GSCs.143 A better understanding
of the molecular mechanisms governing autophagic
responses in various CSCs may prove critical for
the development of novel antineoplastic therapy
aiming at tumor eradication.
F. Autophagy and Cancer Cell
Resistance to Therapy
Most anticancer therapies, including radiation ther-
apy, chemotherapy, and targeted therapies, invoke
autophagy in tumor cells209–211 which predominantly
functions as a cytoprotective mechanism against
Huang et al.
Critical Reviews™ in Oncogenesis
256
therapy-induced stress responses.212,213 Preclinical
data from immunodecient host animal models in-
dicate that pharmacological suppression of autopha-
gy with inhibitors such as 3-methyladenine, CQ, or
hydroxychloroquine (HCQ) can augment cytotox-
icity in combination with various anticancer treat-
ments. These treatments include conventional che-
motherapeutics (e.g., cisplatin,214 5-uorouracil,215
temozolomide,216 and epirubicin217), radiation ther-
apy,218,219 targeted agents (e.g., getinib22), and anti-
angiogenic agents (e.g., bevacizumab221). Moreover,
genetic silencing of autophagy regulatory genes,
such as ATG5, ATG7, BECN-1, and LC3, leads to an
enhanced sensitivity of tumor cells to chemothera-
py, 214 ionizing radiation,222 and antiangiogenic ther-
apy.142,223 The mechanism of autophagy-mediated
cancer therapy resistance includes removal of geno-
toxic ROS,224,225 blockade of apoptosis,226,227 and
maintenance of the CSC pool.204,228 Such preclinical
evidence supports the idea of targeting autophagy as
a promising therapeutic strategy to overcome cancer
drug resistance. Multiple ongoing clinical trials are
deciphering the combination effect of CQ or HCQ
with various therapies.213,229
Autophagy is also involved in the induction
of robust antitumor immune responses, which
play a pivotal role in eliminating tumor cells after
cytotoxic chemotherapies.230 Like tumor cells, the
immune system derives benets from the cytopro-
tective effects of autophagy, and therapy-induced
autophagy in tumor cells can cause immunogenic
cell death, leading to efcient recognition by the
immune system.231,232 Thus, autophagy-targeted
therapy may generate undesirable effects that
weaken the host immune system against malignant
cells upon treatment. This assertion is evidenced
by studies of immunocompetent hosts showing
that defective autophagy can limit (rather than
increase) the sensitivity of tumors to therapies
that activate anticancer immune responses.222,223
Currently, available clinical data on CQ or HCQ
combinational therapy is disappointing, indicating
that the immune-promoting function of autophagy
seems to dominate over its tumor cytoprotective
effect in response of anticancer therapy.223 Future
efforts should be dedicated to conrming the func-
tion of autophagy activation in malignant cells
upon multiple anticancer regimens in the presence
of a functional immune system.
VI. TARGETING AUTOPHAGY FOR CANCER
THERAPEUTICS
Cytoprotective autophagy is an important response
to treatment with chemotherapeutic agents and ra-
diation.234 In most cases, autophagy supports the
survival of cancer cells in anticancer therapy; how-
ever, under certain conditions it fosters cell death.235
In any case, strategies aimed at the modulation of
autophagy bear the potential to improve the efcacy
of chemotherapy and radiation therapy. The optimal
strategy seems to depend on tumor type, stage, ge-
netic context, and specic treatment.236 Additional-
ly, autophagy manipulation might sensitize resistant
cancer types to the cytotoxic effects of treatment.
Furthermore, a combination of autophagy modula-
tors and conventional treatments may sensitize can-
cer cells to cancer therapies.237
Sensitization of tumor cells to therapies is one
of the most researched topics in the autophagy eld.
There is mounting preclinical evidence that target-
ing autophagy can enhance the benecial effects of
many cancer therapies. In many cancer types, inhibi-
tion of the nucleation step of autophagosome forma-
tion with type III PI3K inhibitors [e.g., 3-methylad-
enine (3-MA) or LY294002] enhances the efcacy
of chemotherapy or radiation. For example, inhibi-
tion of autophagy using 3-MA enhanced cytotoxic-
ity of radiotherapy in human esophageal squamous
carcinoma cells.238 Similarly, treatment of 3-MA
enhanced the efcacy of the Gli inhibitor GANT-61
and increased apoptotic cell death.239 Many preclini-
cal studies indicate that lysosomotropic agents (e.g.,
CQ and HCQ) prevent lysosome acidication and
block autolysosome formation, thereby augment-
ing the effect of chemotherapies and radiotherapy
on various types of cancers.237,240 For example, in
non-small-cell lung cancer, bevacizumab in com-
bination with CQ increases the efcacy of cancer
treatment.241 However, it should be noted that CQ
and HCQ are not specic autophagy inhibitors and
they may impact biological processes other than
Authophagy and Hallmarks of Cancer 257
Volume 23, Issues 5-6, 2018
autophagy. These may include lysosomal membrane
permeabilization, normalization of tumor vascula-
ture, and subsequent activation of the mitochondrial
pathway of apoptosis.242,243 Also, CQ is reported to
eliminate CSCs through autophagy-independent
pathways, including the deregulation of Janus ki-
nase 2 (JAK2)208 and the inhibition of CXCR4 and
Hedgehog signaling.244 An important limitation of
HCQ is its potency, as high doses (up to 1,200 mg
daily) generate only modest autophagy inhibition
in vivo.237 Moreover, HCQ fails to block autophagy
ux in tumor environments due to a decrease in drug
uptake by cells. Therefore, more potent and selec-
tive autophagy inhibitors are urgently needed.
A specic ATG4B inhibitor, NSC185058, has
been developed and shown to effectively inhibit
ATG4B activity and autophagy without affecting
the activities of mTOR and PI3K.245 ATG4B is a
cysteine protease that is essential for LC3 lipida-
tion and recycling.246 Inhibition of autophagy with
NSC185058 had a negative impact on the develop-
ment of Saos-2 osteosarcoma tumors in vivo.245 Ad-
ditionally, NSC185058 in combination with radio-
therapy markedly slows tumor growth and provides
survival benets in mice with intracranial glioblas-
toma xenografts.143 ATG4 proteases are emerging as
potential pharmaceutical targets for the treatment of
aggressive cancers, such as osteosarcoma and glio-
blastoma.
While signicant progress has been made with
the discovery of autophagy inhibitors in the last
decade, we must keep in mind that many of these
pharmacologic agents do not exclusively target the
autophagy pathways. Therefore, the development of
more potent and specic drugs targeting autophagy
at the preclinical stage (e.g., ATG4B inhibitors) is
highly warranted. This can help maximize the poten-
tial for autophagy manipulation in treating cancers.
VII. CONCLUSIONS
A plethora of evidence indicates the importance of
autophagy in tumor development and progression,
with both tumor-suppressive and tumor-promoting
roles. Whether autophagy modulation should be
attempted in tumor therapy remains controversial.
Nevertheless, various research groups are focusing
on therapeutically targeting autophagy in cancer.
Furthermore, many current clinical trials are using
CQ and HCQ for autophagy inhibition in combina-
tion with other therapies for cancer treatment. How-
ever, existing drugs, such as CQ, HCQ, and 3-MA
can mediate multiple effects. Thus, it is not entirely
clear that autophagy inhibition per se is solely re-
sponsible for observed therapeutic benets.247 It is
therefore important to develop more potent and se-
lective autophagy inhibitors to improve our under-
standing of autophagy and expand our therapeutic
options for multiple diseases.
CKNOWLEDGMENTSA
This work was supported by a National Institutes
of Health (NIH) grant (NS095634) to S.-Y.C.; a
Brain Cancer Research Award from the James S.
McDonnell Foundation to B.H.; NIH grants (L32
MD010147 and T32 CA070085) to A.A.A. and
(F31 CA232630) to N.S.; a Fishel Predoctoral Fel-
lowship Award from the Robert H. Lurie Compre-
hensive Cancer Center at Northwestern University
to N.S.; support from the Lou and Jean Malnati
Brain Tumor Institute at Northwestern University to
S.-Y.C. and B.H.; National Natural Science Foun-
dation of China grants (81372704, 81572467) to H.
Feng; Program for Professor of Special Appoint-
ment (Eastern Scholar) at Shanghai Institutions of
Higher Learning (2014024), Shanghai Municipal
Education Commission—Gaofeng Clinical Medi-
cine Grant Support (20161310), New Hundred Tal-
ent Program (Outstanding Academic Leader) at the
Shanghai Municipal Health Bureau (2017BR021),
and the State Key Laboratory of Oncogenes and Re-
lated Genes in China (91-17-25) to H. Feng.
EFERENCESR
1. Lamb CA, Yoshimori T, Tooze SA. The autophagosome:
origins unknown, biogenesis complex. Nat Rev Mol Cell
Biol. 2013;14(12):759–74.
2. Menzies FM, Fleming A, Rubinsztein DC. Compromised
autophagy and neurodegenerative diseases. Nat Rev Neu-
rosci. 2015;16(6):345–57.
Huang et al.
Critical Reviews™ in Oncogenesis
258
3. Vakifahmetoglu-Norberg H, Xia HG, Yuan J. Phar-
macologic agents targeting autophagy. J Clin Invest.
2015;125(1):5–13.
4. Galluzzi L, Pietrocola F, Levine B, Kroemer G. Metabol-
ic control of autophagy. Cell. 2014;159(6):1263–76.
5. Kaur J, Debnath J. Autophagy at the crossroads of
catabolism and anabolism. Nat Rev Mol Cell Biol.
2015;16(8):461–72.
6. Nixon RA. The role of autophagy in neurodegenerative
disease. Nat Med. 2013;19(8):983–97.
7. White E. Deconvoluting the context-dependent role for
autophagy in cancer. Nat Rev Cancer. 2012;12(6):401–10.
8. White E. The role for autophagy in cancer. J Clin Invest.
2015;125(1):42–6.
9. Klionsky DJ, Baehrecke EH, Brumell JH, Chu CT, Codo-
gno P, Cuervo AM, Debnath J, Deretic V, Elazar Z, Eske-
linen EL, Finkbeiner S, Fueyo-Margareto J, Gewirtz D,
Jaattela M, Kroemer G, Levine B, Melia TJ, Mizushima
N, Rubinsztein DC, Simonsen A, Thorburn A, Thumm M,
Tooze SA. A comprehensive glossary of autophagy-relat-
ed molecules and processes (2nd edition). Autophagy.
2011;7(11):1273–94.
10. Suzuki K, Kirisako T, Kamada Y, Mizushima N, Noda T,
Ohsumi Y. The pre-autophagosomal structure organized
by concerted functions of APG genes is essential for au-
tophagosome formation. EMBO J. 2001;20(21):5971–81.
11. Axe EL, Walker SA, Manifava M, Chandra P, Roderick
HL, Habermann A, Grifths G, Ktistakis NT. Autopha-
gosome formation from membrane compartments en-
riched in phosphatidylinositol 3-phosphate and dynami-
cally connected to the endoplasmic reticulum. J Cell Biol.
2008;182(4):685–701.
12. Gonzalez A, Hall MN. Nutrient sensing and TOR sig-
naling in yeast and mammals. EMBO J. 2017;36(4):
397–408.
13. Saxton RA, Sabatini DM. mTOR Signaling in growth,
metabolism, and disease. Cell. 2017;169(2):361–71.
14. Alers S, Lofer AS, Wesselborg S, Stork B. Role of
AMPK-mTOR-Ulk1/2 in the regulation of autopha-
gy: cross talk, shortcuts, and feedbacks. Mol Cell Biol.
2012;32(1):2–11.
15. Mihaylova MM, Shaw RJ. The AMPK signalling path-
way coordinates cell growth, autophagy and metabolism.
Nat Cell Biol. 2011;13(9):1016–23.
16. Hurley JH, Young LN. Mechanisms of autophagy initia-
tion. Annu Rev Biochem. 2017;86:225–44.
17. Kim J, Kundu M, Viollet B, Guan KL. AMPK and mTOR
regulate autophagy through direct phosphorylation of
Ulk1. Nat Cell Biol. 2011;13(2):132–41.
18. Kamada Y, Yoshino K, Kondo C, Kawamata T, Oshiro N,
Yonezawa K, Ohsumi Y. Tor directly controls the Atg1
kinase complex to regulate autophagy. Mol Cell Biol.
2010;30(4):1049–58.
19. Egan D, Kim J, Shaw RJ, Guan KL. The autophagy
initiating kinase ULK1 is regulated via opposing phos-
phorylation by AMPK and mTOR. Autophagy. 2011;7(6)
:643–4.
20. Egan DF, Shackelford DB, Mihaylova MM, Gelino S,
Kohnz RA, Mair W, Vasquez DS, Joshi A, Gwinn DM,
Taylor R, Asara JM, Fitzpatrick J, Dillin A, Viollet B,
Kundu M, Hansen M, Shaw RJ. Phosphorylation of ULK1
(hATG1) by AMP-activated protein kinase connects en-
ergy sensing to mitophagy. Science. 2011;331(6016):
456–61.
21. Gwinn DM, Shackelford DB, Egan DF, Mihaylova MM,
Mery A, Vasquez DS, Turk BE, Shaw RJ. AMPK phos-
phorylation of raptor mediates a metabolic checkpoint.
Mol Cell. 2008;30(2):214–26.
22. Russell RC, Tian Y, Yuan H, Park HW, Chang YY, Kim
J, Kim H, Neufeld TP, Dillin A, Guan KL. ULK1 induces
autophagy by phosphorylating Beclin-1 and activating
VPS34 lipid kinase. Nat Cell Biol. 2013;15(7):741–50.
23. Backer JM. The intricate regulation and complex func-
tions of the Class III phosphoinositide 3-kinase Vps34.
Biochem J. 2016;473(15):2251–71.
24. Vergne I, Roberts E, Elmaoued RA, Tosch V, Delgado
MA, Proikas-Cezanne T, Laporte J, Deretic V. Control of
autophagy initiation by phosphoinositide 3-phosphatase
Jumpy. EMBO J. 2009;28(15):2244–58.
25. Taguchi-Atarashi N, Hamasaki M, Matsunaga K, Omori
H, Ktistakis NT, Yoshimori T, Noda T. Modulation of lo-
cal PtdIns3P levels by the PI phosphatase MTMR3 regu-
lates constitutive autophagy. Trafc. 2010;11(4):468–78.
26. Dooley HC, Razi M, Polson HE, Girardin SE, Wilson MI,
Tooze SA. WIPI2 links LC3 conjugation with PI3P, au-
tophagosome formation, and pathogen clearance by re-
cruiting Atg12-5-16L1. Mol Cell. 2014;55(2):238–52.
27. Geng J, Klionsky DJ. The Atg8 and Atg12 ubiquitin-like
conjugation systems in macroautophagy. Protein modi-
cations: beyond the usual suspects’ review series. EMBO
Rep. 2008;9(9):859–64.
28. Bento CF, Renna M, Ghislat G, Puri C, Ashkenazi A,
Vicinanza M, Menzies FM, Rubinsztein DC. Mamma-
lian autophagy: how does it work? Annu Rev Biochem.
2016;85:685–713.
29. Nakatogawa H. Two ubiquitin-like conjugation systems
that mediate membrane formation during autophagy. Es-
says Biochem. 2013;55:39–50.
30. Mizushima N, Noda T, Yoshimori T, Tanaka Y, Ishii T,
George MD, Klionsky DJ, Ohsumi M, Ohsumi Y. A pro-
tein conjugation system essential for autophagy. Nature.
1998;395(6700):395–8.
31. Sakoh-Nakatogawa M, Matoba K, Asai E, Kirisako H,
Ishii J, Noda NN, Inagaki F, Nakatogawa H, Ohsumi
Y. Atg12-Atg5 conjugate enhances E2 activity of Atg3
by rearranging its catalytic site. Nat Struct Mol Biol.
2013;20(4):433–9.
32. Slobodkin MR, Elazar Z. The Atg8 family: multifunc-
Authophagy and Hallmarks of Cancer 259
Volume 23, Issues 5-6, 2018
tional ubiquitin-like key regulators of autophagy. Essays
Biochem. 2013;55:51–64.
33. Li M, Hou Y, Wang J, Chen X, Shao ZM, Yin XM. Ki-
netics comparisons of mammalian Atg4 homologues indi-
cate selective preferences toward diverse Atg8 substrates.
J Biol Chem. 2011;286(9):7327–38.
34. Tanida I, Ueno T, Kominami E. LC3 conjugation sys-
tem in mammalian autophagy. Int J Biochem Cell Biol.
2004;36(12):2503–18.
35. Schaaf MB, Keulers TG, Vooijs MA, Rouschop KM.
LC3/GABARAP family proteins: autophagy-(un)related
functions. FASEB J. 2016;30(12):3961–78.
36. Fujita N, Hayashi-Nishino M, Fukumoto H, Omori H,
Yamamoto A, Noda T, Yoshimori T. An Atg4B mutant
hampers the lipidation of LC3 paralogues and causes
defects in autophagosome closure. Mol Biol Cell.
2008;19(11):4651–9.
37. Weidberg H, Shpilka T, Shvets E, Abada A, Shimron F,
Elazar Z. LC3 and GATE-16 N termini mediate mem-
brane fusion processes required for autophagosome bio-
genesis. Dev Cell. 2011;20(4):444–54.
38. Reggiori F, Ungermann C. Autophagosome maturation
and fusion. J Mol Biol. 2017;429(4):486–96.
39. Ganley IG. Autophagosome maturation and lysosomal
fusion. Essays Biochem. 2013;55:65–78.
40. Nakamura S, Yoshimori T. New insights into autophago-
some-lysosome fusion. J Cell Sci. 2017;130(7):1209–16.
41. Kriegenburg F, Ungermann C, Reggiori F. Coordination
of autophagosome-lysosome fusion by Atg8 family mem-
bers. Curr Biol. 2018;28(8):R512–8.
42. Morani F, Titone R, Pagano L, Galetto A, Alabiso O,
Aimaretti G, Isidoro C. Autophagy and thyroid carcino-
genesis: genetic and epigenetic links. Endocr Relat Can-
cer. 2014;21(1):R13–29.
43. Fullgrabe J, Klionsky DJ, Joseph B. The return of the nu-
cleus: transcriptional and epigenetic control of autophagy.
Nat Rev Mol Cell Biol. 2014;15(1):65–74.
44. Boehrer S, Lainey E, Kroemer G. Coordinated epigen-
etic regulation of autophagy and apoptosis. Cell Cycle.
2011;10(17):2832–3.
45. Artal-Martinez de Narvajas A, Gomez TS, Zhang JS,
Mann AO, Taoda Y, Gorman JA, Herreros-Villanueva M,
Gress TM, Ellenrieder V, Bujanda L, Kim DH, Kozikows-
ki AP, Koenig A, Billadeau DD. Epigenetic regulation of
autophagy by the methyltransferase G9a. Mol Cell Biol.
2013;33(20):3983–93.
46. Xu Z, Klionsky DJ. The AMPK-SKP2-CARM1 axis links
nutrient sensing to transcriptional and epigenetic regula-
tion of autophagy. Ann Transl Med. 2016;4(Suppl 1):S7.
47. Wu BJ, Zhao LX, Zhu CC, Chen YL, Wei MY, Bao SQ,
Sun SC, Li XH. Altered apoptosis/autophagy and epigen-
etic modications cause the impaired postimplantation
octaploid embryonic development in mice. Cell Cycle.
2017;16(1):82–90.
48. Wei FZ, Cao Z, Wang X, Wang H, Cai MY, Li T, Hattori
N, Wang D, Du Y, Song B, Cao LL, Shen C, Wang L,
Wang H, Yang Y, Xie D, Wang F, Ushijima T, Zhao Y, Zhu
WG. Epigenetic regulation of autophagy by the methyl-
transferase EZH2 through an MTOR-dependent pathway.
Autophagy. 2015;11(12):2309–22.
49. Sui X, Zhu J, Zhou J, Wang X, Li D, Han W, Fang Y, Pan
H. Epigenetic modications as regulatory elements of au-
tophagy in cancer. Cancer Lett. 2015;360(2):106–13.
50. Shin HR, Kim H, Kim KI, Baek SH. Epigenetic and
transcriptional regulation of autophagy. Autophagy.
2016;12(11):2248–9.
51. Chen S, Jing Y, Kang X, Yang L, Wang DL, Zhang W,
Zhang L, Chen P, Chang JF, Yang XM, Sun FL. Histone
H2B monoubiquitination is a critical epigenetic switch
for the regulation of autophagy. Nucleic Acids Res.
2017;45(3):1144–58.
52. Byun S, Kim YC, Zhang Y, Kong B, Guo G, Sadoshima
J, Ma J, Kemper B, Kemper JK. A postprandial FGF19-
SHP-LSD1 regulatory axis mediates epigenetic repression
of hepatic autophagy. EMBO J. 2017;36(12):1755–69.
53. Baek SH, Kim KI. Epigenetic Control of autoph-
agy: nuclear events gain more attention. Mol Cell.
2017;65(5):781–5.
54. Shin HJ, Kim H, Oh S, Lee JG, Kee M, Ko HJ, Kweon
MN, Won KJ, Baek SH. AMPK-SKP2-CARM1 signal-
ling cascade in transcriptional regulation of autophagy.
Nature. 2016;534(7608):553–7.
55. An PNT, Shimaji K, Tanaka R, Yoshida H, Kimura H,
Fukusaki E, Yamaguchi M. Epigenetic regulation of star-
vation-induced autophagy in Drosophila by histone meth-
yltransferase G9a. Sci Rep. 2017;7(1):7343.
56. Ren A, Qiu Y, Cui H, Fu G. Inhibition of H3K9 methyl-
transferase G9a induces autophagy and apoptosis in oral
squamous cell carcinoma. Biochem Biophys Res Com-
mun. 2015;459(1):10–7.
57. Fullgrabe J, Lynch-Day MA, Heldring N, Li W, Struijk
RB, Ma Q, Hermanson O, Rosenfeld MG, Klionsky
DJ, Joseph B. The histone H4 lysine 16 acetyltransfer-
ase hMOF regulates the outcome of autophagy. Nature.
2013;500(7463):468–71.
58. Ke XX, Zhang D, Zhu S, Xia Q, Xiang Z, Cui H. Inhibi-
tion of H3K9 methyltransferase G9a repressed cell pro-
liferation and induced autophagy in neuroblastoma cells.
PLoS One. 2014;9(9):e106962.
59. Liu TP, Lo HL, Wei LS, Hsiao HH, Yang PM. S-Ade-
nosyl-L-methionine-competitive inhibitors of the his-
tone methyltransferase EZH2 induce autophagy and en-
hance drug sensitivity in cancer cells. Anticancer Drugs.
2015;26(2):139–47.
60. Hsieh YY, Lo HL, Yang PM. EZH2 inhibitors transcrip-
tionally upregulate cytotoxic autophagy and cytoprotec-
tive unfolded protein response in human colorectal cancer
cells. Am J Cancer Res. 2016;6(8):1661–80.
Huang et al.
Critical Reviews™ in Oncogenesis
260
61. Chen H, Fan M, Pfeffer LM, Laribee RN. The histone
H3 lysine 56 acetylation pathway is regulated by tar-
get of rapamycin (TOR) signaling and functions direct-
ly in ribosomal RNA biogenesis. Nucleic Acids Res.
2012;40(14):6534–46.
62. Kourmouli N, Jeppesen P, Mahadevhaiah S, Burgoyne
P, Wu R, Gilbert DM, Bongiorni S, Prantera G, Fanti L,
Pimpinelli S, Shi W, Fundele R, Singh PB. Heterochro-
matin and tri-methylated lysine 20 of histone H4 in ani-
mals. J Cell Sci. 2004;117(Pt 12):2491–501.
63. Goiran T, Duplan E, Rouland L, El Manaa W, Lauritzen
I, Dunys J, You H, Checler F, Alves da Costa C. Nuclear
p53-mediated repression of autophagy involves PINK1
transcriptional down-regulation. Cell Death Differ.
2018;25(5):873–84.
64. Sakamaki JI, Long JS, New M, Van Acker T, Tooze SA,
Ryan KM. Emerging roles of transcriptional programs in
autophagy regulation. Transcription. 2018;9(2):131–6.
65. Sakamaki JI, Ryan KM. Transcriptional regulation of au-
tophagy and lysosomal function by bromodomain protein
BRD4. Autophagy. 2017;13(11):2006–7.
66. Pan H, Yan Y, Liu C, Finkel T. The role of ZKSCAN3
in the transcriptional regulation of autophagy. Autophagy.
2017;13(7):1235–8.
67. Moreau K, Ghislat G, Hochfeld W, Renna M, Zavodsz-
ky E, Runwal G, Puri C, Lee S, Siddiqi F, Menzies FM,
Ravikumar B, Rubinsztein DC. Transcriptional regula-
tion of Annexin A2 promotes starvation-induced autoph-
agy. Nat Commun. 2015;6:8045.
68. Hsieh MJ, Hsieh YH, Lin CW, Chen MK, Yang SF, Chiou
HL. Transcriptional regulation of Mcl-1 plays an impor-
tant role of cellular protective effector of vincristine-trig-
gered autophagy in oral cancer cells. Expert Opin Ther
Targets. 2015;19(4):455–70.
69. Desgeorges MM, Freyssenet D, Chanon S, Castells J,
Pugniere P, Bechet D, Peinnequin A, Devillard X, Defour
A. Post-transcriptional regulation of autophagy in C2C12
myotubes following starvation and nutrient restoration.
Int J Biochem Cell Biol. 2014;54:208–16.
70. Miao LJ, Huang FX, Sun ZT, Zhang RX, Huang SF,
Wang J. Stat3 inhibits Beclin 1 expression through re-
cruitment of HDAC3 in nonsmall cell lung cancer cells.
Tumour Biol. 2014;35(7):7097–103.
71. Pensa S, Lloyd-Lewis B, Sargeant TJ, Resemann HK,
Kahn CR, Watson CJ. Signal transducer and activator
of transcription 3 and the phosphatidylinositol 3-kinase
regulatory subunits p55alpha and p50alpha regulate au-
tophagy in vivo. FEBS J. 2014;281(20):4557–67.
72. Tai WT, Shiau CW, Chen HL, Liu CY, Lin CS, Cheng AL,
Chen PJ, Chen KF. Mcl-1-dependent activation of Beclin
1 mediates autophagic cell death induced by sorafenib
and SC-59 in hepatocellular carcinoma cells. Cell Death
Dis. 2013;4:e485.
73. Feng Y, Ke C, Tang Q, Dong H, Zheng X, Lin W, Ke J,
Huang J, Yeung SC, Zhang H. Metformin promotes au-
tophagy and apoptosis in esophageal squamous cell carci-
noma by downregulating Stat3 signaling. Cell Death Dis.
2014;5:e1088.
74. Niso-Santano M, Shen S, Adjemian S, Malik SA, Marino
G, Lachkar S, Senovilla L, Kepp O, Galluzzi L, Maiuri
MC, Kroemer G. Direct interaction between STAT3 and
EIF2AK2 controls fatty acid-induced autophagy. Autoph-
agy. 2013;9(3):415–7.
75. Shen S, Niso-Santano M, Adjemian S, Takehara T, Ma-
lik SA, Minoux H, Souquere S, Marino G, Lachkar S,
Senovilla L, Galluzzi L, Kepp O, Pierron G, Maiuri MC,
Hikita H, Kroemer R, Kroemer G. Cytoplasmic STAT3
represses autophagy by inhibiting PKR activity. Mol Cell.
2012;48(5):667–80.
76. Liu M, Han T, Shi S, Chen E. Long noncoding RNA
HAGLROS regulates cell apoptosis and autophagy in
lipopolysaccharides-induced WI-38 cells via modulating
miR-100/NF-kappaB axis. Biochem Biophys Res Com-
mun. 2018;500(3):589–96.
77. Chen JF, Wu P, Xia R, Yang J, Huo XY, Gu DY, Tang
CJ, De W, Yang F. STAT3-induced lncRNA HAGLROS
overexpression contributes to the malignant progression
of gastric cancer cells via mTOR signal-mediated inhibi-
tion of autophagy. Mol Cancer. 2018;17(1):6.
78. Sardiello M, Palmieri M, di Ronza A, Medina DL, Valen-
za M, Gennarino VA, Di Malta C, Donaudy F, Embrione
V, Polishchuk RS, Ban S, Parenti G, Cattaneo E, Bal-
labio A. A gene network regulating lysosomal biogenesis
and function. Science. 2009;325(5939):473–7.
79. Zeng W, Xiao T, Cai A, Cai W, Liu H, Liu J, Li J, Tan M,
Xie L, Liu Y, Yang X, Long Y. Inhibiting ROS-TFEB-
dependent autophagy enhances salidroside-induced apop-
tosis in human chondrosarcoma cells. Cell Physiol Bio-
chem. 2017;43(4):1487–502.
80. Sha Y, Rao L, Settembre C, Ballabio A, Eissa NT. STUB1
regulates TFEB-induced autophagy-lysosome pathway.
EMBO J. 2017;36(17):2544–52.
81. Hsu CL, Lee EX, Gordon KL, Paz EA, Shen WC, Ohnishi
K, Meisenhelder J, Hunter T, La Spada AR. MAP4K3 me-
diates amino acid-dependent regulation of autophagy via
phosphorylation of TFEB. Nat Commun. 2018;9(1):942.
82. Neill T, Sharpe C, Owens RT, Iozzo RV. Decorin-evoked
paternally expressed gene 3 (PEG3) is an upstream
regulator of the transcription factor EB (TFEB) in en-
dothelial cell autophagy. J Biol Chem. 2017;292(39):
16211–20.
83. Palmieri M, Pal R, Sardiello M. AKT modulates the
autophagy-lysosome pathway via TFEB. Cell Cycle.
2017;16(13):1237–8.
84. Sakamaki JI, Wilkinson S, Hahn M, Tasdemir N, O’Prey
J, Clark W, Hedley A, Nixon C, Long JS, New M, Van
Acker T, Tooze SA, Lowe SW, Dikic I, Ryan KM. Bro-
modomain protein BRD4 is a transcriptional repres-
Authophagy and Hallmarks of Cancer 261
Volume 23, Issues 5-6, 2018
sor of autophagy and lysosomal function. Mol Cell.
2017;66(4):517–32.
85. Uribesalgo I, Ballare C, Di Croce L. Polycomb regulates
NF-kappaB signaling in cancer through miRNA. Cancer
Cell. 2012;21(1):5–7.
86. Ren Y, Chen Y, Liang X, Lu Y, Pan W, Yang M. MiR-
NA-638 promotes autophagy and malignant phenotypes
of cancer cells via directly suppressing DACT3. Cancer
Lett. 2017;390:126–36.
87. Zhang P, Zhang H. Autophagy modulates miRNA-me-
diated gene silencing and selectively degrades AIN-1/
GW182 in C. elegans. EMBO Rep. 2013;14(6):568–76.
88. Gibbings D, Mostowy S, Voinnet O. Autophagy se-
lectively regulates miRNA homeostasis. Autophagy.
2013;9(5):781–3.
89. Ucar A, Gupta SK, Fiedler J, Erikci E, Kardasinski M,
Batkai S, Dangwal S, Kumarswamy R, Bang C, Holz-
mann A, Remke J, Caprio M, Jentzsch C, Engelhardt S,
Geisendorf S, Glas C, Hofmann TG, Nessling M, Richter
K, Schiffer M, Carrier L, Napp LC, Bauersachs J, Chow-
dhury K, Thum T. The miRNA-212/132 family regulates
both cardiac hypertrophy and cardiomyocyte autophagy.
Nat Commun. 2012;3:1078.
90. Kovaleva V, Mora R, Park YJ, Plass C, Chiramel AI,
Bartenschlager R, Dohner H, Stilgenbauer S, Pscherer
A, Lichter P, Seiffert M. miRNA-130a targets ATG2B
and DICER1 to inhibit autophagy and trigger killing
of chronic lymphocytic leukemia cells. Cancer Res.
2012;72(7):1763–72.
91. Zhu H, Wu H, Liu X, Li B, Chen Y, Ren X, Liu CG,
Yang JM. Regulation of autophagy by a beclin 1-tar-
geted microRNA, miR-30a, in cancer cells. Autophagy.
2009;5(6):816–23.
92. Yu Y, Yang L, Zhao M, Zhu S, Kang R, Vernon P, Tang
D, Cao L. Targeting microRNA-30a-mediated autophagy
enhances imatinib activity against human chronic my-
eloid leukemia cells. Leukemia. 2012;26(8):1752–60.
93. Fu Z, Luo W, Wang J, Peng T, Sun G, Shi J, Li Z, Zhang
B. Malat1 activates autophagy and promotes cell prolif-
eration by sponging miR-101 and upregulating STMN1,
RAB5A and ATG4D expression in glioma. Biochem Bio-
phys Res Commun. 2017;492(3):480–6.
94. Guo J, Huang X, Wang H, Yang H. Celastrol induces
autophagy by targeting AR/miR-101 in prostate cancer
cells. PLoS One. 2015;10(10):e0140745.
95. Xu Y, An Y, Wang Y, Zhang C, Zhang H, Huang C, Jiang
H, Wang X, Li X. miR-101 inhibits autophagy and en-
hances cisplatin-induced apoptosis in hepatocellular car-
cinoma cells. Oncol Rep. 2013;29(5):2019–24.
96. Frankel LB, Wen J, Lees M, Hoyer-Hansen M, Farkas
T, Krogh A, Jaattela M, Lund AH. microRNA-101 is a
potent inhibitor of autophagy. EMBO J. 2011;30(22):
4628–41.
97. Korkmaz G, le Sage C, Tekirdag KA, Agami R, Gozuacik
D. miR-376b controls starvation and mTOR inhibition-
related autophagy by targeting ATG4C and BECN1. Au-
tophagy. 2012;8(2):165–76.
98. Wang P, Zhang J, Zhang L, Zhu Z, Fan J, Chen L,
Zhuang L, Luo J, Chen H, Liu L, Chen Z, Meng Z. Mi-
croRNA 23b regulates autophagy associated with radio-
resistance of pancreatic cancer cells. Gastroenterology.
2013;145(5):1133–43.
99. Schmitt AM, Chang HY. Long noncoding RNAs in can-
cer pathways. Cancer Cell. 2016;29(4):452–63.
100. Wang K, Liu CY, Zhou LY, Wang JX, Wang M, Zhao
B, Zhao WK, Xu SJ, Fan LH, Zhang XJ, Feng C, Wang
CQ, Zhao YF, Li PF. APF lncRNA regulates autophagy
and myocardial infarction by targeting miR-188-3p. Nat
Commun. 2015;6:6779.
101. Xiong H, Ni Z, He J, Jiang S, Li X, He J, Gong W, Zheng
L, Chen S, Li B, Zhang N, Lyu X, Huang G, Chen B,
Zhang Y, He F. LncRNA HULC triggers autophagy via
stabilizing Sirt1 and attenuates the chemosensitivity of
HCC cells. Oncogene. 2017;36(25):3528–40.
102. Xia H, Qu XL, Liu LY, Qian DH, Jing HY. LncRNA
MEG3 promotes the sensitivity of vincristine by inhib-
iting autophagy in lung cancer chemotherapy. Eur Rev
Med Pharmacol Sci. 2018;22(4):1020–7.
103. Ma B, Gao Z, Lou J, Zhang H, Yuan Z, Wu Q, Li X,
Zhang B. Long noncoding RNA MEG3 contributes to
cisplatin-induced apoptosis via inhibition of autophagy in
human glioma cells. Mol Med Rep. 2017;16(3):2946–52.
104. Xiu YL, Sun KX, Chen X, Chen S, Zhao Y, Guo QG,
Zong ZH. Upregulation of the lncRNA Meg3 induces au-
tophagy to inhibit tumorigenesis and progression of epi-
thelial ovarian carcinoma by regulating activity of ATG3.
Oncotarget. 2017;8(19):31714–25.
105. Pawar K, Hanisch C, Palma Vera SE, Einspanier R,
Sharbati S. Down regulated lncRNA MEG3 eliminates
mycobacteria in macrophages via autophagy. Sci Rep.
2016;6:19416.
106. Ying L, Huang Y, Chen H, Wang Y, Xia L, Chen Y, Liu Y,
Qiu F. Downregulated MEG3 activates autophagy and in-
creases cell proliferation in bladder cancer. Mol Biosyst.
2013;9(3):407–11.
107. Kung CP, Budina A, Balaburski G, Bergenstock MK,
Murphy M. Autophagy in tumor suppression and can-
cer therapy. Crit Rev Eukaryot Gene Expr. 2011;21(1):
71–100.
108. Liang XH, Jackson S, Seaman M, Brown K, Kempkes
B, Hibshoosh H, Levine B. Induction of autophagy
and inhibition of tumorigenesis by beclin 1. Nature.
1999;402(6762):672–6.
109. Furuya N, Yu F, Byeld M, Pattingre S, Levine B. The
evolutionarily conserved domain of Beclin 1 is required
for Vps34 binding, autophagy and tumor suppressor func-
tion. Autophagy. 2005;1(1):46–52.
110. Liang XH, Kleeman LK, Jiang HH, Gordon G, Goldman
Huang et al.
Critical Reviews™ in Oncogenesis
262
JE, Berry G, Herman B, Levine B. Protection against fatal
Sindbis virus encephalitis by beclin, a novel Bcl-2-inter-
acting protein. J Virol. 1998;72(11):8586–96.
111. Pattingre S, Tassa A, Qu X, Garuti R, Liang XH, Mizushi-
ma N, Packer M, Schneider MD, Levine B. Bcl-2 anti-
apoptotic proteins inhibit Beclin 1-dependent autophagy.
Cell. 2005;122(6):927–39.
112. Yue Z, Jin S, Yang C, Levine AJ, Heintz N. Beclin 1, an
autophagy gene essential for early embryonic develop-
ment, is a haploinsufcient tumor suppressor. Proc Natl
Acad Sci U S A. 2003;100(25):15077–82.
113. Qu X, Yu J, Bhagat G, Furuya N, Hibshoosh H, Troxel
A, Rosen J, Eskelinen EL, Mizushima N, Ohsumi Y, Cat-
toretti G, Levine B. Promotion of tumorigenesis by het-
erozygous disruption of the beclin 1 autophagy gene. J
Clin Invest. 2003;112(12):1809–20.
114. Edinger AL, Thompson CB. Defective autophagy leads to
cancer. Cancer Cell. 2003;4(6):422–4.
115. Takahashi Y, Coppola D, Matsushita N, Cualing HD, Sun
M, Sato Y, Liang C, Jung JU, Cheng JQ, Mule JJ, Pledg-
er WJ, Wang HG. Bif-1 interacts with Beclin 1 through
UVRAG and regulates autophagy and tumorigenesis. Nat
Cell Biol. 2007;9(10):1142–51.
116. Liang C, Feng P, Ku B, Dotan I, Canaani D, Oh BH,
Jung JU. Autophagic and tumour suppressor activity of
a novel Beclin1-binding protein UVRAG. Nat Cell Biol.
2006;8(7):688–99.
117. Balakrishnan A, von Neuhoff N, Rudolph C, Kamphues
K, Schraders M, Groenen P, van Krieken JH, Callet-Bau-
chu E, Schlegelberger B, Steinemann D. Quantitative mi-
crosatellite analysis to delineate the commonly deleted
region 1p22.3 in mantle cell lymphomas. Genes Chromo
Cancer. 2006;45(10):883–92.
118. Lee JW, Jeong EG, Soung YH, Nam SW, Lee JY, Yoo NJ,
Lee SH. Decreased expression of tumour suppressor Bax-
interacting factor-1 (Bif-1), a Bax activator, in gastric car-
cinomas. Pathology. 2006;38(4):312–5.
119. Takamura A, Komatsu M, Hara T, Sakamoto A, Kishi
C, Waguri S, Eishi Y, Hino O, Tanaka K, Mizushima N.
Autophagy-decient mice develop multiple liver tumors.
Genes Dev. 2011;25(8):795–800.
120. Narendra D, Tanaka A, Suen DF, Youle RJ. Parkin is
recruited selectively to impaired mitochondria and pro-
motes their autophagy. J Cell Biol. 2008;183(5):795–803.
121. Youle RJ, Narendra DP. Mechanisms of mitophagy. Nat
Rev Mol Cell Biol. 2011;12(1):9–14.
122. Veeriah S, Morris LG, Solit D, Chan TA. The familial
Parkinson disease gene PARK2 is a multisite tumor sup-
pressor on chromosome 6q25.2-27 that regulates cyclin
E. Cell Cycle. 2010;9(8):1451–2.
123. Poulogiannis G, McIntyre RE, Dimitriadi M, Apps JR,
Wilson CH, Ichimura K, Luo F, Cantley LC, Wyllie AH,
Adams DJ, Arends MJ. PARK2 deletions occur frequent-
ly in sporadic colorectal cancer and accelerate adenoma
development in Apc mutant mice. Proc Natl Acad Sci U S
A. 2010;107(34):15145–50.
124. Moscat J, Diaz-Meco MT. p62: a versatile multitasker
takes on cancer. Trends Biochem Sci. 2012;37(6):230–6.
125. Komatsu M, Kurokawa H, Waguri S, Taguchi K, Ko-
bayashi A, Ichimura Y, Sou YS, Ueno I, Sakamoto A,
Tong KI, Kim M, Nishito Y, Iemura S, Natsume T, Ueno
T, Kominami E, Motohashi H, Tanaka K, Yamamoto M.
The selective autophagy substrate p62 activates the stress
responsive transcription factor Nrf2 through inactivation
of Keap1. Nat Cell Biol. 2010;12(3):213–23.
126. Nioi P, McMahon M, Itoh K, Yamamoto M, Hayes JD.
Identication of a novel Nrf2-regulated antioxidant re-
sponse element (ARE) in the mouse NAD(P)H:quinone
oxidoreductase 1 gene: reassessment of the ARE consen-
sus sequence. Biochem J. 2003;374(Pt 2):337–48.
127. Duran A, Linares JF, Galvez AS, Wikenheiser K, Flores
JM, Diaz-Meco MT, Moscat J. The signaling adaptor p62
is an important NF-kappa B mediator in tumorigenesis.
Cancer Cell. 2008;13(4):343–54.
128. DeNicola GM, Karreth FA, Humpton TJ, Gopinathan A,
Wei C, Frese K, Mangal D, Yu KH, Yeo CJ, Calhoun ES,
Scrimieri F, Winter JM, Hruban RH, Iacobuzio-Donahue
C, Kern SE, Blair IA, Tuveson DA. Oncogene-induced
Nrf2 transcription promotes ROS detoxication and tu-
morigenesis. Nature. 2011;475(7354):106–9.
129. Shibata T, Ohta T, Tong KI, Kokubu A, Odogawa R, Tsuta
K, Asamura H, Yamamoto M, Hirohashi S. Cancer related
mutations in NRF2 impair its recognition by Keap1-Cul3
E3 ligase and promote malignancy. Proc Natl Acad Sci U
S A. 2008;105(36):13568–73.
130. Hayes JD, McMahon M. NRF2 and KEAP1 mutations:
permanent activation of an adaptive response in cancer.
Trends Biochem Sci. 2009;34(4):176–88.
131. Konstantinopoulos PA, Spentzos D, Fountzilas E, Fran-
coeur N, Sanisetty S, Grammatikos AP, Hecht JL, Can-
nistra SA. Keap1 mutations and Nrf2 pathway activation
in epithelial ovarian cancer. Cancer Res. 2011;71(15):
5081–9.
132. Mathew R, Karantza-Wadsworth V, White E. Role of au-
tophagy in cancer. Nat Rev Cancer. 2007;7(12):961–7.
133. Suzuki SW, Onodera J, Ohsumi Y. Starvation in-
duced cell death in autophagy-defective yeast mutants
is caused by mitochondria dysfunction. PLoS One.
2011;6(2):e17412.
134. Onodera J, Ohsumi Y. Autophagy is required for main-
tenance of amino acid levels and protein synthesis un-
der nitrogen starvation. J Biol Chem. 2005;280(36):
31582–6.
135. Hara T, Nakamura K, Matsui M, Yamamoto A, Nakahara
Y, Suzuki-Migishima R, Yokoyama M, Mishima K, Saito
I, Okano H, Mizushima N. Suppression of basal autoph-
agy in neural cells causes neurodegenerative disease in
mice. Nature. 2006;441(7095):885–9.
Authophagy and Hallmarks of Cancer 263
Volume 23, Issues 5-6, 2018
136. Komatsu M, Waguri S, Chiba T, Murata S, Iwata J, Tanida
I, Ueno T, Koike M, Uchiyama Y, Kominami E, Tanaka
K. Loss of autophagy in the central nervous system causes
neurodegeneration in mice. Nature. 2006;441(7095):
880–4.
137. Kuma A, Hatano M, Matsui M, Yamamoto A, Nakaya H,
Yoshimori T, Ohsumi Y, Tokuhisa T, Mizushima N. The
role of autophagy during the early neonatal starvation pe-
riod. Nature. 2004;432(7020):1032–6.
138. Rabinowitz JD, White E. Autophagy and metabolism.
Science. 2010;330(6009):1344–8.
139. Yang S, Wang X, Contino G, Liesa M, Sahin E, Ying H,
Bause A, Li Y, Stommel JM, Dell’antonio G, Mautner J,
Tonon G, Haigis M, Shirihai OS, Doglioni C, Bardeesy
N, Kimmelman AC. Pancreatic cancers require autopha-
gy for tumor growth. Genes Dev. 2011;25(7):717–29.
140. Lum JJ, Bauer DE, Kong M, Harris MH, Li C, Lindsten
T, Thompson CB. Growth factor regulation of autoph-
agy and cell survival in the absence of apoptosis. Cell.
2005;120(2):237–48.
141. Degenhardt K, Mathew R, Beaudoin B, Bray K, Ander-
son D, Chen G, Mukherjee C, Shi Y, Gelinas C, Fan Y,
Nelson DA, Jin S, White E. Autophagy promotes tumor
cell survival and restricts necrosis, inammation, and tu-
morigenesis. Cancer Cell. 2006;10(1):51–64.
142. Hu YL, DeLay M, Jahangiri A, Molinaro AM, Rose
SD, Carbonell WS, Aghi MK. Hypoxia-induced au-
tophagy promotes tumor cell survival and adaptation to
antiangiogenic treatment in glioblastoma. Cancer Res.
2012;72(7):1773–83.
143. Huang T, Kim CK, Alvarez AA, Pangeni RP, Wan X,
Song X, Shi T, Yang Y, Sastry N, Horbinski CM, Lu S,
Stupp R, Kessler JA, Nishikawa R, Nakano I, Sulman EP,
Lu X, James CD, Yin XM, Hu B, Cheng SY. MST4 Phos-
phorylation of ATG4B regulates autophagic activity, tu-
morigenicity, and radioresistance in glioblastoma. Cancer
cell. 2017;32(6):840–55.
144. Strohecker AM, White E. Autophagy promotes
BrafV600E-driven lung tumorigenesis by preserving mi-
tochondrial metabolism. Autophagy. 2014;10(2):384–5.
145. Guo JY, Chen HY, Mathew R, Fan J, Strohecker AM,
Karsli-Uzunbas G, Kamphorst JJ, Chen G, Lemons JM,
Karantza V, Coller HA, Dipaola RS, Gelinas C, Rabi-
nowitz JD, White E. Activated Ras requires autophagy to
maintain oxidative metabolism and tumorigenesis. Genes
Dev. 2011;25(5):460–70.
146. Larki P, Gharib E, Yaghoob Taleghani M, Khorshidi F,
Nazemalhosseini-Mojarad E, Asadzadeh Aghdaei H. Co-
existence of KRAS and BRAF mutations in colorectal
cancer: a case report supporting the concept of tumoral
heterogeneity. Cell J. 2017;19(Suppl 1):113–7.
147. Hu YM, Lu WQ, Chen G, Wang P, Chen Z, Zhou Y, Oga-
sawara M, Trachootham D, Feng L, Pelicano H, Chiao PJ,
Keating MJ, Garcia-Manero G, Huang P. K-ras(G12V)
transformation leads to mitochondrial dysfunction and a
metabolic switch from oxidative phosphorylation to gly-
colysis. Cell Res. 2012;22(2):399–412.
148. Hanahan D, Weinberg RA. Hallmarks of cancer: the next
generation. Cell. 2011;144(5):646–74.
149. Schmelzle T, Hall MN. TOR, a central controller of cell
growth. Cell. 2000;103(2):253–62.
150. Noda T, Ohsumi Y. Tor, a phosphatidylinositol kinase
homologue, controls autophagy in yeast. J Biol Chem.
1998;273(7):3963–6.
151. Heras-Sandoval D, Perez-Rojas JM, Hernandez-Damian
J, Pedraza-Chaverri J. The role of PI3K/AKT/mTOR
pathway in the modulation of autophagy and the clear-
ance of protein aggregates in neurodegeneration. Cell
Signal. 2014;26(12):2694–701.
152. Feng Z, Zhang H, Levine AJ, Jin S. The coordinate regu-
lation of the p53 and mTOR pathways in cells. Proc Natl
Acad Sci U S A. 2005;102(23):8204–9.
153. Balaburski GM, Hontz RD, Murphy ME. p53 and ARF:
unexpected players in autophagy. Trends Cell Biol.
2010;20(6):363–9.
154. Zhong Z, Sanchez-Lopez E, Karin M. Autophagy, inam-
mation, and immunity: a troika governing cancer and its
treatment. Cell. 2016;166(2):288–98.
155. Fischer KR, Durrans A, Lee S, Sheng J, Li F, Wong
ST, Choi H, El Rayes T, Ryu S, Troeger J, Schwabe
RF, Vahdat LT, Altorki NK, Mittal V, Gao D. Epitheli-
al-to-mesenchymal transition is not required for lung
metastasis but contributes to chemoresistance. Nature.
2015;527(7579):472–6.
156. Peng YF, Shi YH, Ding ZB, Ke AW, Gu CY, Hui B, Zhou
J, Qiu SJ, Dai Z, Fan J. Autophagy inhibition suppresses
pulmonary metastasis of HCC in mice via impairing anoi-
kis resistance and colonization of HCC cells. Autophagy.
2013;9(12):2056–68.
157. Chen S, Han Q, Wang X, Yang M, Zhang Z, Li P, Chen A,
Hu C, Li S. IBP-mediated suppression of autophagy pro-
motes growth and metastasis of breast cancer cells via ac-
tivating mTORC2/Akt/FOXO3a signaling pathway. Cell
Death Dis. 2013;4:e842.
158. Okura R, Nakamura M. Overexpression of autopha-
gy-related beclin-1 in cutaneous squamous cell car-
cinoma with lymph-node metastasis. Eur J Dermatol.
2011;21(6):1002–3.
159. Kenic CM, Thorburn A, Debnath J. Autophagy and me-
tastasis: another double-edged sword. Curr Opin Cell
Biol. 2010;22(2):241–5.
160. Li J, Yang B, Zhou Q, Wu Y, Shang D, Guo Y, Song
Z, Zheng Q, Xiong J. Autophagy promotes hepato-
cellular carcinoma cell invasion through activation of
epithelial-mesenchymal transition. Carcinogenesis.
2013;34(6):1343–51.
161. Shen H, Yin L, Deng G, Guo C, Han Y, Li Y, Cai C, Fu Y,
Liu S, Zeng S. Knockdown of Beclin-1 impairs epithelial-
Huang et al.
Critical Reviews™ in Oncogenesis
264
mesenchymal transition of colon cancer cells. J Cell Bio-
chem. 2018. DOI:10.1002/jcb.26912.
162. Kim YH, Baek SH, Kim EK, Ha JM, Jin SY, Lee HS,
Ha HK, Song SH, Kim SJ, Shin HK, Yong J, Kim DH,
Kim CD, Bae SS. Uncoordinated 51-like kinase 2 sig-
naling pathway regulates epithelial-mesenchymal transi-
tion in A549 lung cancer cells. FEBS Lett. 2016;590(9):
1365–74.
163. Su Z, Li G, Liu C, Ren S, Deng T, Zhang S, Tian Y, Liu
Y, Qiu Y. Autophagy inhibition impairs the epithelial-
mesenchymal transition and enhances cisplatin sensitiv-
ity in nasopharyngeal carcinoma. Oncol Lett. 2017;13(6):
4147–54.
164. Wang F, Li H, Yan XG, Zhou ZW, Yi ZG, He ZX, Pan ST,
Yang YX, Wang ZZ, Zhang X, Yang T, Qiu JX, Zhou SF.
Alisertib induces cell cycle arrest and autophagy and sup-
presses epithelial-to-mesenchymal transition involving
PI3K/Akt/mTOR and sirtuin 1-mediated signaling path-
ways in human pancreatic cancer cells. Drug Des Devel
Ther. 2015;9:575–601.
165. Ding YH, Zhou ZW, Ha CF, Zhang XY, Pan ST, He ZX,
Edelman JL, Wang D, Yang YX, Zhang X, Duan W, Yang
T, Qiu JX, Zhou SF. Alisertib, an Aurora kinase A inhibi-
tor, induces apoptosis and autophagy but inhibits epitheli-
al to mesenchymal transition in human epithelial ovarian
cancer cells. Drug Des Devel Ther. 2015;9:425–64.
166. Zi D, Zhou ZW, Yang YJ, Huang L, Zhou ZL, He SM,
He ZX, Zhou SF. Danusertib induces apoptosis, cell cy-
cle arrest, and autophagy but inhibits epithelial to mes-
enchymal transition involving PI3K/Akt/mTOR Signal-
ing pathway in human ovarian cancer cells. Int J Mol Sci.
2015;16(11):27228–51.
167. Li JP, Yang YX, Liu QL, Zhou ZW, Pan ST, He ZX, Zhang
X, Yang T, Pan SY, Duan W, He SM, Chen XW, Qiu JX,
Zhou SF. The pan-inhibitor of Aurora kinases danusertib
induces apoptosis and autophagy and suppresses epithe-
lial-to-mesenchymal transition in human breast cancer
cells. Drug Des Devel Ther. 2015;9:1027–62.
168. Zhao Z, Zhao J, Xue J, Zhao X, Liu P. Autophagy inhibi-
tion promotes epithelial-mesenchymal transition through
ROS/HO-1 pathway in ovarian cancer cells. Am J Cancer
Res. 2016;6(10):2162–77.
169. Catalano M, D’Alessandro G, Lepore F, Corazzari M,
Caldarola S, Valacca C, Faienza F, Esposito V, Limatola
C, Cecconi F, Di Bartolomeo S. Autophagy induction im-
pairs migration and invasion by reversing EMT in glio-
blastoma cells. Mol Oncol. 2015;9(8):1612–25.
170. Gugnoni M, Sancisi V, Gandol G, Manzotti G, Raga-
zzi M, Giordano D, Tamagnini I, Tigano M, Frasoldati
A, Piana S, Ciarrocchi A. Cadherin-6 promotes EMT and
cancer metastasis by restraining autophagy. Oncogene.
2017;36(5):667–77.
171. Lo HW, Hsu SC, Xia W, Cao X, Shih JY, Wei Y, Abbru-
zzese JL, Hortobagyi GN, Hung MC. Epidermal growth
factor receptor cooperates with signal transducer and ac-
tivator of transcription 3 to induce epithelial-mesenchy-
mal transition in cancer cells via up-regulation of TWIST
gene expression. Cancer Res. 2007;67(19):9066–76.
172. Yang F, Sun L, Li Q, Han X, Lei L, Zhang H, Shang Y.
SET8 promotes epithelial-mesenchymal transition and
confers TWIST dual transcriptional activities. EMBO J.
2012;31(1):110–23.
173. Lv Q, Wang W, Xue J, Hua F, Mu R, Lin H, Yan J, Lv
X, Chen X, Hu ZW. DEDD interacts with PI3KC3 to
activate autophagy and attenuate epithelial-mesenchy-
mal transition in human breast cancer. Cancer Res.
2012;72(13):3238–50.
174. Qiang L, He YY. Autophagy deciency stabilizes
TWIST1 to promote epithelial-mesenchymal transition.
Autophagy. 2014;10(10):1864–5.
175. Jiang X, Huang Y, Liang X, Jiang F, He Y, Li T, Xu G,
Zhao H, Yang W, Jiang G, Su Z, Jiang L, Liu L. Meta-
static prostate cancer-associated P62 inhibits autophagy
ux and promotes epithelial to mesenchymal transition
by sustaining the level of HDAC6. Prostate. 2018;78(6):
426–34.
176. Bertrand M, Petit V, Jain A, Amsellem R, Johansen T,
Larue L, Codogno P, Beau I. SQSTM1/p62 regulates
the expression of junctional proteins through epithelial-
mesenchymal transition factors. Cell Cycle. 2015;14(3):
364–74.
177. Yang Q, Zhang MX, Zou X, Liu YP, You R, Yu T, Jiang
R, Zhang YN, Cao JY, Hong MH, Liu Q, Guo L, Kang
TB, Zhu XF, Chen MY. A prognostic bio-model based on
SQSTM1 and N-stage identies nasopharyngeal carcino-
ma patients at high risk of metastasis for additional induc-
tion chemotherapy. Clin Cancer Res. 2018;24(3):648–58.
178. Wang D, Shi W, Tang Y, Liu Y, He K, Hu Y, Li J, Yang Y,
Song J. Prefoldin 1 promotes EMT and lung cancer pro-
gression by suppressing cyclin A expression. Oncogene.
2017;36(7):885–98.
179. Pang MF, Georgoudaki AM, Lambut L, Johansson J, Ta-
bor V, Hagikura K, Jin Y, Jansson M, Alexander JS, Nel-
son CM, Jakobsson L, Betsholtz C, Sund M, Karlsson
MC, Fuxe J. TGF-beta1-induced EMT promotes targeted
migration of breast cancer cells through the lymphatic
system by the activation of CCR7/CCL21-mediated che-
motaxis. Oncogene. 2016;35(6):748–60.
180. Kiyono K, Suzuki HI, Matsuyama H, Morishita Y, Komu-
ro A, Kano MR, Sugimoto K, Miyazono K. Autophagy is
activated by TGF-beta and potentiates TGF-beta-mediat-
ed growth inhibition in human hepatocellular carcinoma
cells. Cancer Res. 2009;69(23):8844–52.
181. Dash S, Sarashetti PM, Rajashekar B, Chowdhury R,
Mukherjee S. TGF-beta2-induced EMT is dampened by
inhibition of autophagy and TNF-alpha treatment. Onco-
target. 2018;9(5):6433–49.
182. Zhang C, Zhang X, Xu R, Huang B, Chen AJ, Li C, Wang
Authophagy and Hallmarks of Cancer 265
Volume 23, Issues 5-6, 2018
J, Li XG. TGF-beta2 initiates autophagy via Smad and
non-Smad pathway to promote glioma cells’ invasion. J
Exp Clin Cancer Res. 2017;36(1):162.
183. McGowan PM, Kirstein JM, Chambers AF. Micrometa-
static disease and metastatic outgrowth: clinical issues
and experimental approaches. Future Oncol. 2009;5(7):
1083–98.
184. Chaffer CL, Weinberg RA. A perspective on cancer cell
metastasis. Science. 2011;331(6024):1559–64.
185. Kroemer G, Marino G, Levine B. Autophagy and the in-
tegrated stress response. Mol Cell. 2010;40(2):280–93.
186. Lazova R, Camp RL, Klump V, Siddiqui SF, Amaravadi
RK, Pawelek JM. Punctate LC3B expression is a com-
mon feature of solid tumors and associated with prolif-
eration, metastasis, and poor outcome. Clin Cancer Res.
2012;18(2):370–9.
187. Zhao H, Yang M, Zhao J, Wang J, Zhang Y, Zhang Q.
High expression of LC3B is associated with progression
and poor outcome in triple-negative breast cancer. Med
Oncol. 2013;30(1):475.
188. Han C, Sun B, Wang W, Cai W, Lou D, Sun Y, Zhao X.
Overexpression of microtubule-associated protein-1 light
chain 3 is associated with melanoma metastasis and vascu-
logenic mimicry. Tohoku J Exp Med. 2011;223(4):243–51.
189. DeNardo DG, Barreto JB, Andreu P, Vasquez L, Tawk
D, Kolhatkar N, Coussens LM. CD4(+) T cells regulate
pulmonary metastasis of mammary carcinomas by en-
hancing protumor properties of macrophages. Cancer
Cell. 2009;16(2):91–102.
190. Bingle L, Brown NJ, Lewis CE. The role of tumour-associ-
ated macrophages in tumour progression: implications for
new anticancer therapies. J Pathol. 2002;196(3):254–65.
191. Guo W, Giancotti FG. Integrin signalling during tumour
progression. Nat Rev Mol Cell Biol. 2004;5(10):816–26.
192. Guadamillas MC, Cerezo A, Del Pozo MA. Overcoming
anoikis—pathways to anchorage-independent growth in
cancer. J Cell Sci. 2011;124(Pt 19):3189–97.
193. Sosa MS, Bragado P, Aguirre-Ghiso JA. Mechanisms of
disseminated cancer cell dormancy: an awakening eld.
Nat Rev Cancer. 2014;14(9):611–22.
194. Lu Z, Luo RZ, Lu Y, Zhang X, Yu Q, Khare S, Kondo
S, Kondo Y, Yu Y, Mills GB, Liao WS, Bast RC Jr. The
tumor suppressor gene ARHI regulates autophagy and tu-
mor dormancy in human ovarian cancer cells. J Clin In-
vest. 2008;118(12):3917–29.
195. Mowers EE, Shari MN, Macleod KF. Autophagy in can-
cer metastasis. Oncogene. 2017;36(12):1619–30.
196. Vander Heiden MG, DeBerardinis RJ. Understanding the
intersections between metabolism and cancer biology.
Cell. 2017;168(4):657–69.
197. Vander Heiden MG, Cantley LC, Thompson CB. Under-
standing the Warburg effect: the metabolic requirements
of cell proliferation. Science. 2009;324(5930):1029–33.
198. Singh SS, Vats S, Chia AY, Tan TZ, Deng S, Ong MS,
Arfuso F, Yap CT, Goh BC, Sethi G, Huang RY, Shen
HM, Manjithaya R, Kumar AP. Dual role of autophagy in
hallmarks of cancer. Oncogene. 2018;37(9):1142–58.
199. Kimmelman AC, White E. Autophagy and tumor metabo-
lism. Cell Metab. 2017;25(5):1037–43.
200. Janji B, Berchem G, Chouaib S. Targeting autophagy in
the tumor microenvironment: new challenges and oppor-
tunities for regulating tumor immunity. Front Immunol.
2018;9:887.
201. Sousa CM, Biancur DE, Wang X, Halbrook CJ, Sherman
MH, Zhang L, Kremer D, Hwang RF, Witkiewicz AK,
Ying H, Asara JM, Evans RM, Cantley LC, Lyssiotis CA,
Kimmelman AC. Pancreatic stellate cells support tumour
metabolism through autophagic alanine secretion. Nature.
2016;536(7617):479–83.
202. Huang T, Alvarez A, Hu B, Cheng SY. Noncoding
RNAs in cancer and cancer stem cells. Chin J Cancer.
2013;32(11):582–93.
203. Lathia JD, Mack SC, Mulkearns-Hubert EE, Valentim
CL, Rich JN. Cancer stem cells in glioblastoma. Genes
Dev. 2015;29(12):1203–17.
204. Ojha R, Bhattacharyya S, Singh SK. Autophagy in can-
cer stem cells: a potential link between chemoresis-
tance, recurrence, and metastasis. Biores Open Access.
2015;4(1):97–108.
205. Lomonaco SL, Finniss S, Xiang C, Decarvalho A, Uman-
sky F, Kalkanis SN, Mikkelsen T, Brodie C. The induc-
tion of autophagy by gamma-radiation contributes to
the radioresistance of glioma stem cells. Int J Cancer.
2009;125(3):717–22.
206. Cu S, Vazquez-Martin A, Oliveras-Ferraros C, Martin-
Castillo B, Vellon L, Menendez JA. Autophagy positively
regulates the CD44(+) CD24(-/low) breast cancer stem-
like phenotype. Cell Cycle. 2011;10(22):3871–85.
207. Wolf J, Dewi DL, Fredebohm J, Muller-Decker K, Flech-
tenmacher C, Hoheisel JD, Boettcher M. A mammo-
sphere formation RNAi screen reveals that ATG4A pro-
motes a breast cancer stem-like phenotype. Breast Cancer
Res. 2013;15(6):R109.
208. Choi DS, Blanco E, Kim YS, Rodriguez AA, Zhao H,
Huang TH, Chen CL, Jin G, Landis MD, Burey LA, Qian
W, Granados SM, Dave B, Wong HH, Ferrari M, Wong
ST, Chang JC. Chloroquine eliminates cancer stem cells
through deregulation of Jak2 and DNMT1. Stem Cells.
2014;32(9):2309–23.
209. Paglin S, Hollister T, Delohery T, Hackett N, McMa-
hill M, Sphicas E, Domingo D, Yahalom J. A novel re-
sponse of cancer cells to radiation involves autophagy
and formation of acidic vesicles. Cancer Res. 2001;61(2):
439–44.
210. Zou Z, Yuan Z, Zhang Q, Long Z, Chen J, Tang Z, Zhu
Y, Chen S, Xu J, Yan M, Wang J, Liu Q. Aurora kinase A
inhibition-induced autophagy triggers drug resistance in
breast cancer cells. Autophagy. 2012;8(12):1798–810.
Huang et al.
Critical Reviews™ in Oncogenesis
266
211. Gewirtz DA. The autophagic response to radiation: rele-
vance for radiation sensitization in cancer therapy. Radiat
Res. 2014;182(4):363–7.
212. Sui X, Chen R, Wang Z, Huang Z, Kong N, Zhang M,
Han W, Lou F, Yang J, Zhang Q, Wang X, He C, Pan H.
Autophagy and chemotherapy resistance: a promising
therapeutic target for cancer treatment. Cell Death Dis.
2013;4:e838.
213. Das CK, Mandal M, Kogel D. Pro-survival autophagy
and cancer cell resistance to therapy. Cancer Meta Rev.
2018. DOI: 10.1007/s10555-018-9727-z.
214. Zhao XG, Sun RJ, Yang XY, Liu DY, Lei DP, Jin T, Pan
XL. Chloroquine-enhanced efcacy of cisplatin in the
treatment of hypopharyngeal carcinoma in xenograft
mice. PLoS One. 2015;10(4):e0126147.
215. Li J, Hou N, Faried A, Tsutsumi S, Kuwano H. Inhibi-
tion of autophagy augments 5-uorouracil chemotherapy
in human colon cancer In vitro and in vivo model. Eur J
Cancer. 2010;46(10):1900–9.
216. Golden EB, Cho HY, Jahanian A, Hofman FM, Louie SG,
Schonthal AH, Chen TC. Chloroquine enhances temo-
zolomide cytotoxicity in malignant gliomas by blocking
autophagy. Neurosurg Focus. 2014;37(6):E12.
217. Chittaranjan S, Bortnik S, Dragowska WH, Xu J, Abey-
sundara N, Leung A, Go NE, DeVorkin L, Weppler
SA, Gelmon K, Yapp DT, Bally MB, Gorski SM. Au-
tophagy inhibition augments the anticancer effects of
epirubicin treatment in anthracycline-sensitive and -re-
sistant triple-negative breast cancer. Clin Cancer Res.
2014;20(12):3159–73.
218. Tseng HC, Liu WS, Tyan YS, Chiang HC, Kuo WH,
Chou FP. Sensitizing effect of 3-methyladenine on radi-
ation-induced cytotoxicity in radio-resistant HepG2 cells
In vitro and in tumor xenografts. Chem Biol Interact.
2011;192(3):201–8.
219. Chen Y, Li X, Guo L, Wu X, He C, Zhang S, Xiao Y, Yang
Y, Hao D. Combining radiation with autophagy inhibi-
tion enhances suppression of tumor growth and angio-
genesis in esophageal cancer. Mol Med Rep. 2015;12(2):
1645–52.
220. Dragowska WH, Weppler SA, Wang JC, Wong LY, Kapa-
nen AI, Rawji JS, Warburton C, Qadir MA, Donohue E,
Roberge M, Gorski SM, Gelmon KA, Bally MB. Induc-
tion of autophagy is an early response to getinib and a
potential therapeutic target in breast cancer. PLoS One.
2013;8(10):e76503.
221. Selvakumaran M, Amaravadi RK, Vasilevskaya IA,
O’Dwyer PJ. Autophagy inhibition sensitizes colon can-
cer cells to antiangiogenic and cytotoxic therapy. Clin
Cancer Res. 2013;19(11):2995–3007.
222. Ko A, Kanehisa A, Martins I, Senovilla L, Chargari C,
Dugue D, Marino G, Kepp O, Michaud M, Perfettini JL,
Kroemer G, Deutsch E. Autophagy inhibition radiosen-
sitizes In vitro, yet reduces radioresponses in vivo due
to decient immunogenic signalling. Cell Death Differ.
2014;21(1):92–9.
223. Mohan N, Chakrabarti M, Banik NL, Ray SK. Combi-
nation of LC3 shRNA plasmid transfection and genistein
treatment inhibited autophagy and increased apoptosis in
malignant neuroblastoma in cell culture and animal mod-
els. PLoS One. 2013;8(10):e78958.
224. Ding ZB, Hui B, Shi YH, Zhou J, Peng YF, Gu CY, Yang
H, Shi GM, Ke AW, Wang XY, Song K, Dai Z, Shen
YH, Fan J. Autophagy activation in hepatocellular car-
cinoma contributes to the tolerance of oxaliplatin via
reactive oxygen species modulation. Clin Cancer Res.
2011;17(19):6229–38.
225. Chen X, Wang P, Guo F, Wang X, Wang J, Xu J, Yuan D,
Zhang J, Shao C. Autophagy enhanced the radioresistance
of non-small cell lung cancer by regulating ROS level
under hypoxia condition. Int J Radiat Biol. 2017;93(8):
764–70.
226. Firat E, Weyerbrock A, Gaedicke S, Grosu AL, Nieder-
mann G. Chloroquine or chloroquine-PI3K/Akt pathway
inhibitor combinations strongly promote gamma-irradia-
tion-induced cell death in primary stem-like glioma cells.
PLoS One. 2012;7(10):e47357.
227. Sun WL, Chen J, Wang YP, Zheng H. Autophagy protects
breast cancer cells from epirubicin-induced apoptosis and
facilitates epirubicin-resistance development. Autophagy.
2011;7(9):1035–44.
228. Lei Y, Zhang D, Yu J, Dong H, Zhang J, Yang S. Targeting
autophagy in cancer stem cells as an anticancer therapy.
Cancer Lett. 2017;393:33–9.
229. Levy JMM, Towers CG, Thorburn A. Targeting autopha-
gy in cancer. Nat Rev Cancer. 2017;17(9):528–42.
230. Galluzzi L, Buque A, Kepp O, Zitvogel L, Kroemer G. Im-
munological effects of conventional chemotherapy and tar-
geted anticancer agents. Cancer Cell. 2015;28(6):690–714.
231. Galluzzi L, Pietrocola F, Bravo-San Pedro JM, Amara-
vadi RK, Baehrecke EH, Cecconi F, Codogno P, Debnath
J, Gewirtz DA, Karantza V, Kimmelman A, Kumar S,
Levine B, Maiuri MC, Martin SJ, Penninger J, Piacentini
M, Rubinsztein DC, Simon HU, Simonsen A, Thorburn
AM, Velasco G, Ryan KM, Kroemer G. Autophagy in
malignant transformation and cancer progression. EMBO
J. 2015;34(7):856–80.
232. Michaud M, Martins I, Sukkurwala AQ, Adjemian S, Ma
Y, Pellegatti P, Shen S, Kepp O, Scoazec M, Mignot G,
Rello-Varona S, Tailler M, Menger L, Vacchelli E, Gal-
luzzi L, Ghiringhelli F, di Virgilio F, Zitvogel L, Kroemer
G. Autophagy-dependent anticancer immune responses
induced by chemotherapeutic agents in mice. Science.
2011;334(6062):1573–7.
233. Galluzzi L, Bravo-San Pedro JM, Demaria S, Formenti
SC, Kroemer G. Activating autophagy to potentiate im-
munogenic chemotherapy and radiation therapy. Nat Rev
Clin Oncol. 2017;14(4):247–58.
Authophagy and Hallmarks of Cancer 267
Volume 23, Issues 5-6, 2018
234. Fulda S. Targeting autophagy for the treatment of cancer.
Biol Chem. 2018.
235. Fulda S. Autophagy in cancer therapy. Front Oncol.
2017;7:128.
236. Kimmelman AC. The dynamic nature of autophagy in
cancer. Genes Dev. 2011;25(19):1999–2010.
237. Chude CI, Amaravadi RK. Targeting autophagy in can-
cer: update on clinical trials and novel inhibitors. Int J
Mol Sci. 2017;18(6):160.e1–160.e13.
238. Chen YS, Song HX, Lu Y, Li X, Chen T, Zhang Y, Xue
JX, Liu H, Kan B, Yang G, Fu T. Autophagy inhibi-
tion contributes to radiation sensitization of esophageal
squamous carcinoma cells. Dis Esophagus. 2011;24(6):
437–43.
239. Wang J, Gu S, Huang J, Chen S, Zhang Z, Xu M. Inhibi-
tion of autophagy potentiates the efcacy of Gli inhibi-
tor GANT-61 in MYCN-amplied neuroblastoma cells.
BMC Cancer. 2014;14:768.
240. Shi TT, Yu XX, Yan LJ, Xiao HT. Research progress of
hydroxychloroquine and autophagy inhibitors on cancer.
Cancer Chemother Pharmacol. 2017;79(2):287–94.
241. Shen H, Yin L, Deng G, Guo C, Han Y, Li Y, Cai C, Fu Y,
Liu S, Zeng S. Knockdown of Beclin-1 impairs epithelial-
mesenchymal transition of colon cancer cells. J Cell Bio-
chem. 2018;119(8):7022–31.
242. Maycotte P, Aryal S, Cummings CT, Thorburn J, Mor-
gan MJ, Thorburn A. Chloroquine sensitizes breast can-
cer cells to chemotherapy independent of autophagy. Au-
tophagy. 2012;8(2):200–12.
243. Maes H, Kuchnio A, Peric A, Moens S, Nys K, De Bock
K, Quaegebeur A, Schoors S, Georgiadou M, Wouters J,
Vinckier S, Vankelecom H, Garmyn M, Vion AC, Radtke
F, Boulanger C, Gerhardt H, Dejana E, Dewerchin M,
Ghesquiere B, Annaert W, Agostinis P, Carmeliet P. Tu-
mor vessel normalization by chloroquine independent of
autophagy. Cancer Cell. 2014;26(2):190–206.
244. Balic A, Sorensen MD, Trabulo SM, Sainz B Jr., Ciof
M, Vieira CR, Miranda-Lorenzo I, Hidalgo M, Kleeff J,
Erkan M, Heeschen C. Chloroquine targets pancreatic
cancer stem cells via inhibition of CXCR4 and hedgehog
signaling. Mol Cancer Ther. 2014;13(7):1758–71.
245. Akin D, Wang SK, Habibzadegah-Tari P, Law B, Ostrov
D, Li M, Yin XM, Kim JS, Horenstein N, Dunn WA Jr.
A novel ATG4B antagonist inhibits autophagy and has
a negative impact on osteosarcoma tumors. Autophagy.
2014;10(11):2021–35.
246. Zhang L, Li J, Ouyang L, Liu B, Cheng Y. Unraveling
the roles of Atg4 proteases from autophagy modulation to
targeted cancer therapy. Cancer Lett. 2016;373(1):19–26.
247. Rybstein MD, Bravo-San Pedro JM, Kroemer G, Galluzzi
L. The autophagic network and cancer. Nat Cell Biol.
2018;20(3):243–51.
... In 2022, Hanahan added another four additional emerging hallmarks and enabling characteristics of cancer: unlocking phenotypic plasticity, non-mutational epigenetic reprogramming, polymorphic microbiomes and senescent cells [110]. Moreover, it is now established that the components of the tumor microenvironment (TME) contribute to different cancer hallmarks [111], while the molecular mechanisms involved in autophagy also have a role in the hallmarks of cancer [112]. Studying the research effort involved in elucidating each of these hallmarks and their relevance for BC, Saha et al. (2021) placed invasion and metastasis as the central hallmark, suggesting that this hallmark is the most highly explored, followed by sustaining proliferative signaling in the primary tumor, inducing angiogenesis, resisting apoptosis, enabling replicative immortality, evading growth suppressors, genomic instability, reprogramming energy metabolism, evading immune destruction and tumor-promoting inflammation [113]. ...
... Autophagy is known as a self-catalytic and self-protective program that is responsible for the degradation and recycling of abnormal or unneeded cellular proteins or other macromolecules and damaged organelles to maintain proteostasis and cellular homeostasis, and provide energy, so that activated autophagy enables cancer cells to survive and rapidly adapt and evade most therapies [112,158]. However, activated autophagy can also promote cancer cell death through the excessive degradation of cellular components [159]. ...
... However, activated autophagy can also promote cancer cell death through the excessive degradation of cellular components [159]. Generally, autophagy involves the formation of autophagosomes that sequester cytoplasmic material and deliver it to lysosomal compartments for degradation by lysosomal hydrolases [112]. ...
Article
Full-text available
We are living in an era of advanced nanoscience and nanotechnology. Numerous nanomaterials, culminating in nanorobots, have demonstrated ingenious applications in biomedicine, including breast cancer (BC) nano-theranostics. To solve the complicated problem of BC heterogeneity, non-targeted drug distribution, invasive diagnostics or surgery, resistance to classic onco-therapies and real-time monitoring of tumors, nanorobots are designed to perform multiple tasks at a small scale, even at the organelles or molecular level. Over the last few years, most nanorobots have been bioengineered as biomimetic and biocompatible nano(bio)structures, resembling different organisms and cells, such as urchin, spider, octopus, fish, spermatozoon, flagellar bacterium or helicoidal cyanobacterium. In this review, readers will be able to deepen their knowledge of the structure, behavior and role of several types of nanorobots, among other nanomaterials, in BC theranostics. We summarized here the characteristics of many functionalized nanodevices designed to counteract the main neoplastic hallmark features of BC, from sustaining proliferation and evading anti-growth signaling and resisting programmed cell death to inducing angiogenesis, activating invasion and metastasis, preventing genomic instability, avoiding immune destruction and deregulating autophagy. Most of these nanorobots function as targeted and self-propelled smart nano-carriers or nano-drug delivery systems (nano-DDSs), enhancing the efficiency and safety of chemo-, radio- or photodynamic therapy, or the current imagistic techniques used in BC diagnosis. Most of these nanorobots have been tested in vitro, using various BC cell lines, as well as in vivo, mainly based on mice models. We are still waiting for nanorobots that are low-cost, as well as for a wider transition of these favorable effects from laboratory to clinical practice.
... Autophagy is one of the main processes of degradation and recycling of intracellular components, maintaining homeostasis, and allowing cell adaptation and survival in adverse physiological conditions such as starvation or hypoxia 9 . In cancer, the role of autophagy has been widely explored, and it is recognized that in the early stages prevents malignant transformation. ...
... In cancer, the role of autophagy has been widely explored, and it is recognized that in the early stages prevents malignant transformation. Still, in an established tumor, it is necessary for proliferation, acquisition of metastatic potential and increased tumor malignancy 8,9,32,33 . Since proliferation and survival of some cancer cells driven by RAS/MAPK oncogenic mutations were identified as addicted to the autophagic process, targeting autophagy has been implemented as a potential therapeutic strategy 10,22,34 . ...
Article
Full-text available
Leptin is an adipokine secreted by adipose tissue, which promotes tumor progression by activating canonical signaling pathways such as MAPK/ERK. Recent studies have shown that leptin induces autophagy, and this process is involved in leptin-induced characteristics of malignancy. Autophagy is an intracellular degradation process associated with different hallmarks of cancer, such as cell survival, migration, and metabolic reprogramming. However, its relationship with metabolic reprogramming has not been clearly described. The purpose of this study was to determine the role of leptin-induced autophagy in cancer cell metabolism and its association with cellular proliferation and migration in breast cancer cells. We used ER⁺/PR⁺ and triple-negative breast cancer cell lines treated with leptin, autophagy inhibition, or mitochondrial metabolism inhibitors. Our results show that leptin induces autophagy, increases proliferation, mitochondrial ATP production and mitochondrial function in ER⁺/PR⁺ cells. Importantly, autophagy was required to maintain metabolic changes and cell proliferation driven by leptin. In triple-negative cells, leptin did not induce autophagy or cell proliferation but increased glycolytic and mitochondrial ATP production, mitochondrial function, and cell migration. In triple negative cells, autophagy was required to support metabolic changes and cell migration, and autophagy inhibition decreased cellular migration similar to mitochondrial inhibitors. In conclusion, leptin-induced autophagy supports mitochondrial metabolism in breast cancer cells as well as glycolysis in triple negative cells. Importantly, leptin-induced mitochondrial metabolism promoted cancer cell migration.
... [60] However, other researchers also pointed out that autophagy plays a tumor suppression role. Cancer initiation is suppressed by autophagy, [61,62] and autophagy was reported to inhibit breast cancer metastasis by degrading the autophagy cargo receptor. [63] The core machinery of autophagy is highly conserved by a group of ATGs that associate with other proteins in autophagic flux. ...
Article
Full-text available
Oral squamous cell carcinoma (OSCC) is a prevalent cancer worldwide, and the development of anti-OSCC materials is urgent. The tumor microenvironment has been identified as a significant characteristic of cancer, with the pHe value in OSCC ranging from 6.56 to 6.97. Given the acidic nature of OSCC, the creation of pH-sensitive antitumor materials has become a prominent area of research. A pH-sensitive tertiary amine monomer, dodecylmethylaminoethyl methacrylate (DMAEM), has been previously synthesized. This study aims to evaluate the impact of DMAEM on OSCC. The results demonstrated that DMAEM inhibited the proliferation, migration, and invasion of OSCC cells. Furthermore, it promoted apoptosis and autophagy of OSCC cells, with its anti-OSCC effect being strengthened in the acidic environment. In a subcutaneous transplantation tumor model, DMAEM inhibited the growth of OSCC and expression of Ki-67. The analysis of 16S rDNA sequencing data revealed that DMAEM had no significant impact on the Alpha/Beta diversity of the gastrointestinal tract microbiota in mice and had minimal effect on its composition. Overall, this study suggests that DMAEM exhibits pH-responsive behavior in the acidic tumor microenvironment, effectively inhibiting OSCC without disturbing the gastrointestinal microbiota. These findings highlight the potential of DMAEM for clinical applications in OSCC treatment.
... Aberrant autophagy has been implicated in various diseases including glioblastoma [9]. At the benign stage, autophagy has been shown to perform a tumor-suppressive role, while faulty autophagy has been linked to DNA damage and cancer [10,11]. Autophagy performs multiple functions in tumor formation and progression. ...
Article
Full-text available
Background Glioblastoma (GBM) is the most common brain tumor with the worst prognosis. Temozolomide is the only first-line drug for GBM. Unfortunately, the resistance issue is a classic problem. Therefore, it is essential to develop new drugs to treat GBM. As an oncogene, Skp2 is involved in the pathogenesis of various cancers including GBM. In this study, we investigated the anticancer effect of AAA237 on human glioblastoma cells and its underlying mechanism. Methods CCK-8 assay was conducted to evaluate IC50 values of AAA237 at 48, and 72 h, respectively. The Cellular Thermal Shift Assay (CETSA) was employed to ascertain the status of Skp2 as an intrinsic target of AAA237 inside the cellular milieu. The EdU-DNA synthesis test, Soft-Agar assay and Matrigel assay were performed to check the suppressive effects of AAA237 on cell growth. To identify the migration and invasion ability of GBM cells, transwell assay was conducted. RT-qPCR and Western Blot were employed to verify the level of BNIP3. The mRFP-GFP-LC3 indicator system was utilized to assess alterations in autophagy flux and investigate the impact of AAA237 on the dynamic fusion process between autophagosomes and lysosomes. To investigate the effect of compound AAA237 on tumor growth in vivo, LN229 cells were injected into the brains of mice in an orthotopic model. Results AAA237 could inhibit the growth of GBM cells in vitro. AAA237 could bind to Skp2 and inhibit Skp2 expression and the degradation of p21 and p27. In a dose-dependent manner, AAA237 demonstrated the ability to inhibit colony formation, migration, and invasion of GBM cells. AAA237 treatment could upregulate BNIP3 as the hub gene and therefore induce BNIP3-dependent autophagy through the mTOR pathway whereas 3-MA can somewhat reverse this process. In vivo, the administration of AAA237 effectively suppressed the development of glioma tumors with no side effects. Conclusion Compound AAA237, a novel Skp2 inhibitor, inhibited colony formation, migration and invasion of GBM cells in a dose-dependent manner and time-dependent manner through upregulating BNIP3 as the hub gene and induced BNIP3-dependent autophagy through the mTOR pathway therefore it might be a viable therapeutic drug for the management of GBM. Graphical Abstract
... 25 O processo é o cerne de processo de degradação catabólica, em que macromoléculas citoplasmáticas, proteínas agregadas, organelas danificadas ou tecidos e células em que são degradadas no ambiente extracelular, são fagocitadas, entregues aos lisossomas intracelular, digeridos por hidrolases lisossomais, tendo-se como resultado a geração de nucleotídeos, aminoácidos, ácidos graxos, açúcares e ATP. 26 Essa autodigestão celular sustenta o metabolismo celular de célula tumoral e a sua sobrevivência durante a inanição e o estresse. 27,28 A motilidade celular é baseada em alterações dinâmicas na forma envolvendo ciclos contínuos de formação de pseudópodes com polimerização de citoesqueleto intracelular e formação de filamentos de actina, que, quando contrátil, estimula a migração por estímulo. A entrada de Ca 2+ é necessária para a ativação deste processo, com a criação de ambiente de eletrovoltagem dependente e favorecimento de contrações celulares e por consequência movimentações celulares. ...
Preprint
Full-text available
Introdução: O câncer de pele não melanoma (CPNM) compeende grupo de neoplasias com alta incidência na população mundial. É dividido em carcinoma basocelular (CBC) e de células escamosas (CEC). Por ser de grande prevalência, entender o processo de oncogênese e a relação com íons, proteínas e receptores celulares no CPMN pode contribuir para que novas terapêuticas sejam avaliadas. Objetivo: Entender o processo da oncogênese dos tumores de pele não melanomas e sua relação com a imunolocalização do IP3R. Método: Revisão integrativa da literatura com síntese de evidências. A base de dados foi o PUBMED; a estratégia de busca: “carcinoma espinocelular, AND/OR carcinoma basocelular, AND/OR IP3R, AND/OR imunoistoquímica”. Foram considerados para revisão os trabalhos publicados entre 2018 e 2023. Foram incluídos 40 trabalhos, integralmente lidos e resumidos. Resultados: CPNMs são os tumores malignos mais comuns em todo o mundo, sendo 75-80% o CBC, e até 25% o CEC. As interações moleculares de forma geral, envolvem grande participação de moléculas supressoras tumorais, assim como de procto-oncogenes. Além disso, canais iônicos voltagem dependente controlam o fluxo citosólico de íons, dentre eles o cálcio. O IP3R (receptor do fosfatidil inositol-3) permite a saída de cálcio do retículo endoplasmático para que seja utilizado pela célula para atividades fisiológias como proliferação, angiogênese, motilidade e capacidade de invasão. Conclusão: O IP3R, pelas características de expressão imunoistoquímica, parece estar relacionado também, à fisiopatologia do CPNM.
... 25 O processo é o cerne de processo de degradação catabólica, em que macromoléculas citoplasmáticas, proteínas agregadas, organelas danificadas ou tecidos e células em que são degradadas no ambiente extracelular, são fagocitadas, entregues aos lisossomas intracelular, digeridos por hidrolases lisossomais, tendo-se como resultado a geração de nucleotídeos, aminoácidos, ácidos graxos, açúcares e ATP. 26 Essa autodigestão celular sustenta o metabolismo celular de célula tumoral e a sua sobrevivência durante a inanição e o estresse. 27,28 A motilidade celular é baseada em alterações dinâmicas na forma envolvendo ciclos contínuos de formação de pseudópodes com polimerização de citoesqueleto intracelular e formação de filamentos de actina, que, quando contrátil, estimula a migração por estímulo. A entrada de Ca2+ é necessária para a ativação deste processo, com a criação de ambiente de eletrovoltagem dependente e favorecimento de contrações celulares e por consequência movimentações celulares. ...
Article
Full-text available
Introdução: O câncer de pele não melanoma compeende grupo de neoplasias com alta incidência na população mundial. É dividido em carcinoma basocelular e de células escamosas. Por ser de grande prevalência, entender o processo de oncogênese e a relação com íons, proteínas e receptores celulares no câncer de pele não melanoma pode contribuir para que novas terapêuticas sejam avaliadas. Objetivo: Entender o processo da oncogênese dos tumores de pele não melanomas e sua relação com a imunolocalização do IP3R. Métodos: Revisão integrativa da literatura com síntese de evidências. A base de dados foi o PubMed; a estratégia de busca: “carcinoma espinocelular, AND/ OR carcinoma basocelular, AND/OR IP3R, AND/OR imunoistoquímica”. Foram considerados para revisão os trabalhos publicados entre 2018 e 2023. Foram incluídos 40 trabalhos, integralmente lidos e resumidos. Resultados: Câncer de pele não melanoma são os tumores malignos mais comuns em todo o mundo, sendo 75-80% o carcinoma basocelular, e até 25% o de células escamosas. As interações moleculares de forma geral, envolvem grande participação de moléculas supressoras tumorais, assim como de procto-oncogenes. Além disso, canais iônicos voltagem dependente controlam o fluxo citosólico de íons, dentre eles o cálcio. O IP3R (receptor do fosfatidil inositol-3) permite a saída de cálcio do retículo endoplasmático para que seja utilizado pela célula para atividades fisiológias como proliferação, angiogênese, motilidade e capacidade de invasão. Conclusão: O IP3R, pelas características de expressão imunoistoquímica, parece estar relacionado também, à fisiopatologia do câncer de pele não melanoma.
... Starvation, chemotherapeutic drugs, DNA damage and other cellular stresses can induce autophagy, which plays a critical role in cell survival or death [17,20]. The general consensus is that autophagy suppresses tumor progression in the initial stages but supports cancer cell survival in the later stages [21,22]. Zhu et al. reported that overexpression of MIR106A-5p accelerated the malignant transformation of NPC cells by inhibiting BTG3-mediated autophagy [23]. ...
Article
Full-text available
Nasopharyngeal carcinoma (NPC) is a commonly diagnosed malignancy in southern China and southeast Asia. Previous studies have identified galactosamine-(N-acetyl)-6-sulfatase (GALNS) as a potential biomarker for multiple cancers. However, it is unknown whether GALNS plays a role in NPC development, and the underlying mechanisms remain unclear. In this study, we found that GALNS is overexpressed in NPC cell lines and tissues compared to the normal nasopharyngeal counterparts. Knocking down GALNS expression in the NPC cells significantly decreased their proliferation in vitro, and inhibited xenograft growth in a mouse model. Mechanistically, the anti-proliferative effect of GALNS silencing was the result of autophagy induction via the inhibition of PI3K–AKT–mTOR signaling pathway. Taken together, GALNS drives the progression of NPC via PI3K–AKT–mTOR signaling-mediated autophagy, and is therefore a promising therapeutic target. Supplementary Information The online version contains supplementary material available at 10.1007/s12672-023-00782-4.
Article
Full-text available
Autophagy, a highly regulated cellular process, assumes a dual role in the context of cancer. On the one hand, it functions as a crucial homeostatic pathway, responsible for degrading malfunctioning molecules and organelles, thereby maintaining cellular health. On the other hand, its involvement in cancer development and regression is multifaceted, contingent upon a myriad of factors. This review meticulously examines the intricacies of autophagy, from its molecular machinery orchestrated by Autophagy-Related Genes (ATG) initially discovered in yeast to the various modes of autophagy operative within cells. Beyond its foundational role in cellular maintenance, autophagy reveals context-specific functions in processes like angiogenesis and inflammation. Our analysis delves into how autophagy-related factors directly impact inflammation, underscoring their profound implications for cancer dynamics. Additionally, we extend our inquiry to explore autophagy’s associations with cardiovascular conditions, neurodegenerative disorders, and autoimmune diseases, illuminating the broader medical relevance of this process. Furthermore, this review elucidates how autophagy contributes to sustaining hallmark cancer features, including stem cell maintenance, proliferation, angiogenesis, metastasis, and metabolic reprogramming. Autophagy emerges as a pivotal process that necessitates careful consideration in cancer treatment strategies. To this end, we investigate innovative approaches, ranging from enzyme-based therapies to MTOR inhibitors, lysosomal blockers, and nanoparticle-enabled interventions, all aimed at optimizing cancer treatment outcomes by targeting autophagy pathways. In summary, this comprehensive review provides a nuanced perspective on the intricate and context-dependent role of autophagy in cancer biology. Our exploration not only deepens our understanding of this fundamental process but also highlights its potential as a therapeutic target. By unraveling the complex interplay between autophagy and cancer, we pave the way for more precise and effective cancer treatments, promising better outcomes for patients.
Article
Diabetic kidney disease (DKD) is a devastating complication of diabetes mellitus (DM) and is the most prevalent chronic kidney disease (CKD). Poricoic acid A (PAA), a component isolated from Traditional Chinese Medicine (TCM) Poria cocos , has hypoglycaemic and anti‐fibrosis effects. However, the role of PAA in DKD remains largely unclear. To mimics an in vitro model of DKD, the mouse podocyte MPC5 cells were treated with high glucose (25 mM; HG) for 24 h. CCK‐8 and flow cytometry assays were conducted for assessing MPC5 cell viability and apoptosis. Meanwhile, streptozotocin (STZ) was used to induce experimental DKD in mice by intraperitoneal injection. PAA notably inhibited the apoptosis and inflammation, reduced the generation of ROS, and elevated the MMP level in HG‐treated MPC5 cells. Moreover, PAA obviously reduced blood glucose and urine protein levels, inhibited renal fibrosis in DKD mice. Meanwhile, PAA markedly increased LC3 and ATG5 levels and declined p62 and FUNDC1 levels in HG‐treated MPC5 cells and in the kidney tissues of DKD mice, leading to the activation of cell mitophagy. Furthermore, the downregulation of FUNDC1 also inhibited apoptosis, inflammation, and promoted mitophagy in HG‐treated MPC5 cells. As expected, the knockdown of FUNDC1 further enhanced the protective role of PAA in MPC5 cells following HG treatment, indicating that induction of mitophagy could attenuate podocyte injury. Collectively, PAA could exert beneficial effects on podocyte injury in DKD by promoting mitophagy via downregulating FUNDC1. These findings suggested that PAA may have great potential in alleviating kidney injury in DKD.
Preprint
Full-text available
Background Autophagy-related protein plays a pivotal role in cancer development, progression, and prognosis. Among these proteins, PIK3C3 holds significant importance as it is involved in canonical autophagy, endocytosis, and vesicle trafficking, thereby exerting influential effects on various types of cancer progression. However, the diverse biological significance of PIK3C3 in pan-cancer has not been systematically and comprehensively studied. Methods: Data from The Cancer Genome Atlas (TCGA) and Genotype-Tissue Expression (GTEx) were utilized, and bioinformatics approaches were also employed to explore the potential mechanisms of PIK3C3 in diverse cancers. Results: PIK3C3 exhibited upregulation in several tumors and showed prognostic associations. Low expression of PIK3C3 predicted poorer overall survival (OS) in kidney renal clear cell carcinoma patients, while high expression of PIK3C3 predicted poorer OS in adrenocortical carcinoma, bladder urothelial carcinoma, brain lower grade glioma, and liver hepatocellular carcinoma. Additionally, PIK3C3 expression significantly correlated with immune infiltrating cells and tumor mutational burden, microsatellite instability and neoantigens in several cancer types. Furthermore, knockdown of PIK3C3 in colorectal cancer cells lines significantly suppressed cell proliferation and metastasis. Conclusion: PIK3C3 can be used as an auxiliary indicator for early tumor diagnosis and a prognostic marker for many types of tumors.
Article
Full-text available
Activation of autophagy significantly affects cancer cell behaviors, such as proliferation, differentiation, and invasiveness. Epithelial‐to‐mesenchymal transition (EMT) as an initial step of malignant transformation of cancer cells was linked to the activation of autophagy, but the detailed molecular mechanisms are still unknown. The present study investigates the effects of Beclin‐1, a key molecule involved in activation of autophagy, on EMT of colon cancer cells. The normal colon epithelia cell line of CCD‐18Co and six colon cancer cell lines with different expression levels of Beclin‐1 were used in this study. The activation of autophagy and EMT markers of cancer cells were monitored by Western blotting and quantitative real‐time PCR assay in the presence or absence of rapamycin (autophagy activator) and 3‐MA (autophagy inhibitor). The expression of Beclin‐1 in selected cell lines was modulated using small interfering RNA, and consequentially EMT markers, and cancer cell behaviors including migration and invasion, were also explored. Activation or inhibition of autophagy in colon cancer cells had positive or negative impacts on the expression of EMT markers and malignant behaviors such as cell migration and invasion. Knockdown of beclin‐1 by siRNA apparently inhibited the activation of autophagy induced by rapamycin, consequentially resulted in suppression of EMT and attenuation of invasiveness of colon cancer cells. The results in this study demonstrated an association between activation of autophagy and EMT in colon cancer cells. The results showed suppression of Beclin‐1 expression significantly reduced EMT and invasive behaviors in colon cancer cells.
Article
Full-text available
Cancer cells evolve in the tumor microenvironment, which is now well established as an integral part of the tumor and a determinant player in cancer cell adaptation and resistance to anti-cancer therapies. Despite the remarkable and fairly rapid progress over the past two decades regarding our understanding of the role of the tumor microenvironment in cancer development, its precise contribution to cancer resistance is still fragmented. This is mainly related to the complexity of the “tumor ecosystem” and the diversity of the stromal cell types that constitute the tumor microenvironment. Emerging data indicate that several factors, such as hypoxic stress, activate a plethora of resistance mechanisms, including autophagy, in tumor cells. Hypoxia-induced autophagy in the tumor microenvironment also activates several tumor escape mechanisms, which effectively counteract anti-tumor immune responses mediated by natural killer and cytotoxic T lymphocytes. Therefore, strategies aiming at targeting autophagy in cancer cells in combination with other therapeutic strategies have inspired significant interest to overcome immunological tolerance and promote tumor regression. However, a number of obstacles still hamper the application of autophagy inhibitors in clinics. First, the lack of selectivity of the current pharmacological inhibitors of autophagy makes difficult to draw a clear statement about its effective contribution in cancer. Second, autophagy has been also described as an important mechanism in tumor cells involved in presentation of antigens to T cells. Third, there is a circumstantial evidence that autophagy activation in some innate immune cells may support the maturation of these cells, and it is required for their anti-tumor activity. In this review, we will address these aspects and discuss our current knowledge on the benefits and the drawbacks of targeting autophagy in the context of anti-tumor immunity. We believe that it is important to resolve these issues to predict the use of autophagy inhibitors in combination with immunotherapies in clinical settings.
Article
Full-text available
Pneumonia is a lower respiratory disease caused by pathogens or other factors. This study aimed to explore the roles and mechanism of long noncoding RNA HAGLROS in lipopolysaccharides (LPS)-induced inflammatory injury in pneumonia. The HAGLROS expression in serum of patients with acute stage pneumonia was detected. To induce pulmonary injury, WI-38 human lung fibroblasts were stimulated with lipopolysaccharides (LPS). The HAGLROS expressions in LPS-treated WI-38 cells and the effects of HAGLROS knockdown on the viability, apoptosis, and autophagy of LPS-induced cells were detected. Moreover, the regulatory relationship between HAGLROS and miR-100 was explored as well as the functional targets of miR-100 were identified. Furthermore, the regulatory relationship between miR-100 and PI3K/AKT/NF-κB pathway was elucidated. LncRNA HAGLROS was higher expressed in serum of patients with acute stage pneumonia compared with that in serum of healthy control. LPS caused WI-38 cell injury and increased HAGLROS levels. Downregulation of HAGLROS alleviated LPS-induced cell injury via increasing cell viability, and inhibiting apoptosis and autophagy. Moreover, there was a negative correlation between HAGLROS and miR-100, and the effects of HAGLROS downregulation on LPS-induced apoptosis and autophagy in WI-38 cells were by regulation of miR-100. Furthermore, NFΚB3 was verified as a functional target of miR-100 and effects of miR-100 inhibition on LPS-induced WI-38 cell injury were alleviated by knockdown of NFΚB3. Besides, Knockdown of HAGLROS inhibited the activation of PI3K/AKT/NF-κB pathway. Our findings reveal that downregulation of HAGLROS may alleviate LPS-induced inflammatory injury in WI-38 cells via modulating miR-100/NF-κB axis. HAGLROS/miR-100/NF-κB axis may provide a new strategy for treating acute stage of pneumonia.
Article
Full-text available
Resistance to therapy is one of the prime causes for treatment failure in cancer and recurrent disease. In recent years, autophagy has emerged as an important cell survival mechanism in response to different stress conditions that are associated with cancer treatment and aging. Autophagy is an evolutionary conserved catabolic process through which damaged cellular contents are degraded after uptake into autophagosomes that subsequently fuse with lysosomes for cargo degradation, thereby alleviating stress. In addition, autophagy serves to maintain cellular homeostasis by enriching nutrient pools. Although autophagy can act as a double-edged sword at the interface of cell survival and cell death, increasing evidence suggest that in the context of cancer therapy-induced stress responses, it predominantly functions as a cell survival mechanism. Here, we provide an up-to-date overview on our current knowledge of the role of pro-survival autophagy in cancer therapy at the preclinical and clinical stages and delineate the molecular mechanisms of autophagy regulation in response to therapy-related stress conditions. A better understanding of the interplay of cancer therapy and autophagy may allow to unveil new targets and avenues for an improved treatment of therapy-resistant tumors in the foreseeable future.
Article
Full-text available
Autophagy is the major cellular pathway by which macromolecules are degraded, and amino acid depletion powerfully activates autophagy. MAP4K3, or germinal-center kinase-like kinase, is required for robust cell growth in response to amino acids, but the basis for MAP4K3 regulation of cellular metabolic disposition remains unknown. Here we identify MAP4K3 as an amino acid-dependent regulator of autophagy through its phosphorylation of transcription factor EB (TFEB), a transcriptional activator of autophagy, and through amino acid starvation-dependent lysosomal localization of MAP4K3. We document that MAP4K3 physically interacts with TFEB and MAP4K3 inhibition is sufficient for TFEB nuclear localization, target gene transactivation, and autophagy, even when mTORC1 is activated. Moreover, MAP4K3 serine 3 phosphorylation of TFEB is required for TFEB interaction with mTORC1-Rag GTPase-Ragulator complex and TFEB cytosolic sequestration. Our results uncover a role for MAP4K3 in the control of autophagy and reveal MAP4K3 as a central node in nutrient-sensing regulation.
Article
Macroautophagy is a conserved intracellular lysosomal degradative pathway, vital for the maintenance of cellular homeostasis. It is characterized by double-membrane vesicles called autophagosomes, which sequester the cytoplasmic material destined for lysosomal turnover. In a final step, autophagosomes fuse with lysosomes to release their cargo into the acidic and hydrolytic lumen of these organelles. In recent years, numerous new insights into this fusion event have been gained. Notably, many proteins implicated in autophagosome-lysosome fusion interact with members of the Atg8 protein family. Moreover, Atg8 proteins are described to have intrinsic membrane tethering and fusogenic properties themselves. Here, we summarize the current knowledge about the members of this intriguing protein family, which highlights them as possible hubs for the coordination of the final fusion stages of autophagy.
Article
Objective: Lung cancer is one of the most common malignancies worldwide, the morbidity and mortality of which have been on rising in recent years. Moreover, lncRNAs have been implicated in the development of various cancers, as well as cancer treatment and prognosis. In this study, long non-coding RNA (lncRNA) MEG3, an identified tumor suppressor, was explored for its role in the chemotherapy of lung cancer. Materials and methods: All cases were divided into (I+II) group and (III+IV) group according to different stages of tumor node metastasis (TNM), and were divided into sensitive group and insensitive group according to chemotherapy sensitivity. A549 and H292 cells were selected as the resistant cell and non-resistant lung cancer cells. Quantitative reverse transcriptase-polymerase chain reaction (qRT-PCR) was performed to detect the expression of MEG3. After transfection with overexpression plasmid pcDNA-MEG3 or/and different concentrations of vincristine, cell viability and proliferation were measured by cell counting kit-8 (CCK-8) assay and plate cloning assay, respectively. Western blotting was used to analyze the expressions of autophagy-related proteins. Results: In vivo, lncRNA MEG3 was significantly lower in III+IV group and insensitive group than that in I+II group and sensitive group. In vitro, MEG3 expression in resistant cells was significantly lower than that in non-resistant cells. Overexpression of MEG3 significant inhibited the viability and proliferation of both resistant and non-resistant lung cancer cells. Western blot results showed that autophagy level was higher in resistant cells than that in non-resistant cells, while overexpression of MEG3 significantly reduced the expression of autophagy-related proteins. CCK-8 results also indicated that the cell viability was negatively correlated with the dose of vincristine, while the viability of drug-resistant cells was higher than that of non-drug resistant cells after the treatment of vincristine. The vitality of both cells decreased in a concentration-dependent manner after combined treatment with vincristine and MEG3. Conclusions: Our data indicated that lncRNA MEG3 showed a low expression in chemotherapy-sensitive lung cancer tissues, and overexpression of lncRNA MEG3 attenuated autophagy level, thus increasing the sensitivity of vincristine in chemotherapy of lung cancer.
Article
Macroautophagy (herein termed autophagy) is evolutionarily highly conserved across eukaryotic cells and represents an intracellular catabolic process that targets damaged macromolecules and organelles for degradation. Autophagy is dysregulated in various human diseases including cancer. In addition, many drugs currently used for the treatment of cancer can engage autophagy, which typically promotes cancer cell survival by mitigating cellular stress. However, under certain circumstances activation of autophagy upon anticancer drug treatment can also trigger a lethal type of autophagy termed autophagic cell death (ACD). This may pave new avenues for exploiting the autophagic circuitry in oncology. This review presents the concept and some examples of anticancer drug-induced ACD.
Article
Mammalian cells harness autophagy to eliminate physiological byproducts of metabolism and cope with microenvironmental perturbations. Moreover, autophagy connects cellular adaptation with extracellular circuitries that impinge on immunity and metabolism. As it links transformed and non-transformed components of the tumour microenvironment, such an autophagic network is important for cancer initiation, progression and response to therapy. Here, we discuss the mechanisms whereby the autophagic network interfaces with multiple aspects of malignant disease.
Article
Background: P62 (also named sequestosome-1, SQSTM1) is involved in autophagy regulation through multiple pathways. It interacts with autophagosomes-associated LC3-II and ubiquitinated protein aggregates to engulf the aggregates in autophagosomes, interacts with HDAC6 to inhibit its deacetylase activity to maintain the levels of acetylated α-tubulin and stabilities of microtubules to enhance autophagosome trafficking, and regulates autophagy initiation and cell survival. We performed immunohistochemistry staining of P62 in prostate tissues from prostate cancer patients and found that levels of P62 in patients with prostate adenocarcinomas (PCA) are significantly higher than those in patients with benign prostate hyperplasia (BPH). High levels of P62 predict high tumor grade and high intensity of metastasis. Methods: We created prostate cancer cell lines stably overexpressing P62 and then suppress the expression of P62 in the cell line stably overexpressing P62 with CRISPR technology. Cell proliferation assay with crystal violet, cell migration assay, cell invasion assay, Western blot analysis, and confocal fluorescent microscopy were conducted to test the impact of altered levels of P62 on the growth, migration, invasion, epithelial-to-mesenchymal transition, autophagy flux, HDAC6 activity, and microtubular acetylation of cancer cells. Results: P62 increased the levels of HDAC6 and reduced the acetylation of α-tubulin and the stability of microtubules. Consequently, high levels of P62 caused a promotion of epithelial-to-mesenchymal transition in addition to an impairment of autophagy flux, and further led to an enhancement of proliferation, migration, and invasion of prostate cancer cells. Conclusion: P62 promotes metastasis of PCA by sustaining the level of P62 to inhibit autophagy and promote epithelial-to-mesenchymal transition.