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.
Keywords: therapy, autophagosomes, ATGs, cancer, macroautophagy
I. INTRODUCTION
Macroautophagy (hereafter referred to as autophagy) is a conserved process by which eukaryotic cells sequester intracellular material including damaged organelles, redox-active protein aggregates, and foreign 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-relevant 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 intrinsic or extrinsic stimuli: starvation, growth factor deprivation, hypoxia, pathogens, and many others.4,5 Autophagy is a critical regulator of cellular homeostasis, and autophagic dysfunction is associated with human diseases, including Alzheimer’s disease, Parkinson’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 contexts autophagy promotes tumorigenesis.7,8 In this review, we summarize the mechanism of autophagy 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 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 autolysosome, 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 flux.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 reticulum (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 cytotoxic insults. The best characterized triggers for autophagy induction include inactivation of mammalian 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 regulate autophagy initiation by controlling the activation 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 depletion, AMPK directly activates the ULK1 complex by phosphorylation at multiple sites in the central intrinsically disordered region (IDR).17,19,20 Additionally, 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 downstream 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 phosphorylates VPS34 and BECN1,22 thereby activating the PI3KC3-C1 complex, and in turn generates local phosphatidylinositol-3-phosphate (PI3P) in the phagophore.23 This autophagosome-specific pool of PI3P is essential for phagophore nucleation.24,25 PI3P promotes recruitment of additional autophagy-specific 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 expansion 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 mediated 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 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-associated 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 diffuse form (LC3-I) to a membrane-anchored, lipidated form (LC3-II).34,35 LC3 lipidation is required for phagophore expansion and closure36,37 and is widely used as an autophagosome marker. The autophagasome 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 (vacuole in yeast) to form the structure known as the autolysosome.39 The autophagosome–lysosome fusion 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 specific 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 autophagosomes near lysosomes. Moreover, these proteins assist in the assembly of the fusion machinery, which highlights them as possible hubs for the coordination of the final fusion stages of autophagy.41 Degradation of the autophagic cargo is dependent on a series of lysosomal acid hydrolases. 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 modifications are involved in the control of autophagy.42–53 Post-translational modifications of histones and DNA methylation may affect the chromatin structure, which leads to the dysregulation of a series of autophagy-related genes.43–45,48,49 Several histone modifications, 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 arginine methyltransferase CARM1 protein levels.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 promoters, leading to upregulation of autophagy regulators such as LC3B and sequestosome 1 (SQSTM1/p62).45 EZH2-mediated H3K27me3 epigenetically represses autophagy by downregulating mTOR pathway suppressors such as TSC2, RGS16, GPI, and RHOA.48 Moreover, EZH2 inhibitors such as GSK34359 and UNC199960 significantly induce autophagy in cancer cells. In addition 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 modifications such as H2BK120 monoubiquitination,51 H3K56 acetylation,43,61 and H4K20 methylation43,62 are also involved in autophagy regulation.
B. Transcriptional Regulation of Autophagy
Emerging studies have demonstrated the key role played by transcription factors in regulating autophagy in cancer cells.54,63–69 STAT3 is the one of the most important transcriptional regulators of autophagy-related genes. STAT3 represses BECN1 expression by directly binding to its promoter and recruiting HDAC3.70 STAT3 also regulates autophagy through PIK3R1 regulation. STAT3 suppresses autophagy by transcriptionally upregulating the PIK3R1 gene products p55α and p50α, which inhibit p85α-mediated autophagy.71 Recent studies have demonstrated that STAT3 also acts as a transcriptional activator of BCL2, BCL2L1, and MCL1,72,73 which are key autophagy-related regulators. Interestingly, 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 alpha kinase 2A (EIF2A) phosphorylation.74,75 Furthermore, 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 promote 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 directly 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, emerging studies have demonstrated that TFEB serves as a bridge to mediate upstream regulators and autophagy progression in cancer cells. STUB1 regulates 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 influencing 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 3′-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 first miRNA shown to be involved in autophagy regulation 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 autophagy partially depends on three novel targets, STMN1, RAB5A, and ATG4D.96 Similarly, miR376b regulates starvation and mTOR inhibition–related 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 (lncRNAs) are RNA transcripts longer than 200 nucleotides with no protein-coding potential.99 Increasingly, reports show the importance of lncRNAs in tumorigenesis by regulating protein levels through transcriptional, post-transcriptional, and post-translational levels.99 Since 2013, several 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 significantly 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 lncRNA involved in autophagy regulation,102–106 interacts with ATG3 mRNA and protects it from degradation.104 Taken together, these findings suggest 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 spontaneous lung cancer, liver cancer, and lymphomas.112,113 Allelic deletion of Becn1 has been reported in other tumor types, such as prostate, breast, and ovarian 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 expression of BIF-1 have been observed in several types of cancers.116–118 Moreover, mice with system mosaic deletion of Atg5 or liver-specific Atg7 deficiency also develop liver tumors.119 Together, these findings suggest that autophagy plays a key role in repressing tumorigenesis.
Mitophagy is the selective degradation of damaged 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 ubiquitinate 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 deletion in mice leads to increased hepatocellular carcinoma,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 assembling 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–associated protein 1 (KEAP1), a component of the CUL3–RBX1 E3 ligase complex, binds and inhibits NRF2 activity. However, in the presence of oxidative stress, p62 expression increases. Thus, p62 competitively binds KEAP1, thereby releasing NRF2. NRF2 then translocates to the nucleus and activates the expression of antioxidant defense genes, promoting cell survival and tumorigenesis.125 Autophagy deficiency via liver-specific deletion of Atg7 in mice results in p62 accumulation and NRF2 activation, increasing the expression of NRF2-target genes. Liver tumors originate from autophagy-deficient hepatocytes, which can be partially suppressed by p62 deletion.119 Deficiency in p62 or NRF2 suppresses the development of Ras-driven non-small-cell lung cancer in mouse models.127,128 Furthermore, activating mutations of NRF2 and inactivation mutations of its negative regulator, KEAP1, are found in various types of cancers, implying that p62 and NRF2 function as oncogenes while KEAP1 functions 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 biosynthetic demands of dysregulated proliferating cancer cells.
Basal levels of autophagy are essential for normal tissue homeostasis.132 Autophagy was initially shown to support the survival of yeast under starvation conditions through maintaining amino acid levels and activating the expression of genes in response 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-deficient mice survive for a much shorter time than wild-type mice. Moreover, Atg5-deficient neonates display signs of reduced amino acid concentration and ATP levels,137 suggesting that they suffer from a metabolic crisis. These findings support an important and conserved role for autophagy in normal 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 mitochondrial substrates in the form of amino and fatty acids, further promoting tumor cell growth. Impaired autophagy in cancer cells leads to the accumulation of morphologically and functionally abnormal mitochondria.119 Experimentally induced autophagy deficiency in tumor cells results in deficient ATP and lack of key tricarboxylic acid (TCA) -cycle intermediates, and leads to mitochondrial dysfunction. Mitochondrial dysfunction is mainly exemplified by generation of toxic reactive oxygen species (ROS) and mitochondrial damage, promoting accumulation 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 various stressors in cancer cells to support their high metabolic demands. In conditions in which apoptosis 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 deprivation and hypoxia, typical physiological stresses in the tumor microenvironment, activate autophagy in cancer cells to support survival.141,142 Pharmacological 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 mechanisms. Activating mutations in RAS and BRAF, although frequent in cancer, are mutually exclusive, as concurrent mutations arising in both genes are extremely 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 significantly suppresses cancer cell mitochondrial respiration 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 pharmacologic inhibition of autophagy in cancer cells with K-RAS activation causes increased ROS and DNA damage and decreased mitochondrial oxidative 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 deficiency (Atg7−/−) suppresses the progression 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 sustained proliferation.148 Normal cells have precise controls of cell cycle progression to maintain proper cell growth and function. However, tumor cells obtain unlimited proliferating potential by bypassing cell cycle check points, due to mutations in genes such as TP53 and Retinoblastoma (Rb).148 Basal levels of autophagy in normal tissue maintain genome integrity and prevent tumorigenesis.7,107 Autophagy deficiency by Atg5 or Becn1 deletion (Atg5−/− or Becn1−/−) causes early death in mice.112,137 Moreover, mice with mosaic 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 overexpression, HER-2 amplification, or PTEN loss, followed 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 activates autophagy, which may occur via inhibition of mTOR.152,153 These findings 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 tumor cells to grow under both normal and starvation conditions.144,145 Impaired autophagy in these tumors greatly inhibits tumor cell growth and tumor progression, revealing the role of autophagy in sustaining tumor cell proliferation. Taken together, these cases show that the complexity of tumors and the role of autophagy in regulating tumor cell proliferation are highly context-dependent.
B. Autophagy Promotes Epithelial–Mesenchymal Transition
Epithelial–mesenchymal transition (EMT), another hallmark of cancer, is the process by which epithelial cells lose their adhesive properties and become more migratory and invasive mesenchymal cells. Autophagy and EMT both play crucial roles in human cancer progression.154,155 However, the interplay between autophagy and EMT remains unclear. Here, we focus on elucidating the intricate relationship 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 hepatocellular 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 Similarly, cisplatin treatment activates autophagy in nasopharyngeal carcinoma, and autophagy inhibition impairs EMT in this progression.163
Contrary to these findings, 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 kinase inhibitors alisertib164,165 and danusertib,166,167 activate 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 inducing 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 transcription factor, is the hallmark of EMT and plays crucial roles in cancer metastasis.171,172 Recent studies have indicated that the degradation of TWIST underlies autophagy-inhibiting EMT in cancer cells. The death effector domain–containing DNA-binding protein (DEDD) directly interacts with PI3KC3/BECN-1 to induce autophagy-mediated lysosomal degradation of SNAIL and TWIST,173 which consequentially inhibit EMT. Finally, autophagy impairment stabilizes TWIST1 to promote EMT.174
Recent studies indicate that p62 and TGF-β mediate crosstalk between autophagy and EMT. Earlier studies revealed that p62, an autophagy adaptor 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 increases 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 inducing 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 influencing mitochondrial trafficking 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 invasion 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, formation of small nodules of cancer cells, and finally outgrowth of micrometastatic lesions into macroscopic tumors. This process requires the translocation of cancer cells to new microenvironments, in which metastatic tumor cells must overcome numerous challenges to survive. These include altered nutrient supply, immune surveillance, and T-cell–mediated 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 flux has been found to be upregulated by the different environmental stressors that promote invasion and metastasis of cancer cells, such as hypoxia and nutrient deprivation.185 In addition, recent studies have identified an association between increased punctate staining for LC3B and metastasis of various cancers, including human breast cancer, melanoma, and HCC.186–188
Autophagy can either promote or impede cell invasion and metastasis. At the initial steps of metastasis, autophagy promotes the survival of tumor cells from hypoxia and metabolic stress by reducing tumor cell necrosis. Consequently, autophagy reduces the infiltration of macrophages at the primary tumor site, which is required for the initiation of metastasis.189,190 During detachment from the extracellular matrix, autophagy is induced in tumor cells to overcome anoikis, a type of cell death signal triggered by the absence of anchorage to the extracellular 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 promoting 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 tumor suppressor gene aplasia Ras homolog member I (ARHI) to increase tumor cell dormancy.194 The dormancy of tumor cells has also been suggested to result in resistance to genotoxic therapies that primarily 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 metastasis by preventing tumor necrosis and inflammation. However, once the tumor cells have entered a new microenvironment, autophagy helps them survive and maintain dormancy until they successfully establish distant colonies. Therefore, autophagy may be regulated differently at the various stages of metastasis in a way that has significance 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 heightened metabolic requirements necessary for tumor survival and unconstrained proliferation.196 To accomplish these goals, tumor cells have been shown to use various sources of energy. Indeed, some tumors employ increased aerobic glycolysis rather than oxidative 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 metabolism that also helps tumor cell survival in hypoxic microenvironments caused by dysfunctional vasculature. In the absence of pyruvate, other substrates are required to undergo the tricarboxylic acid (TCA) cycle for ATP synthesis, and autophagy can provide these substrates by recycling intracellular 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 nucleosides for glycolysis by degrading carbohydrates and DNA, respectively. It can also provide metabolites 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 pancreatic cancer mitochondrial metabolism.201
E. Implications of Autophagy in Cancer Stem Cells
Cancer stem cells (CSCs) are defined as a small subset of cancer cells within a tumor that can self-renew and generate heterogeneous lineages of cancer cells that make up the tumor.202,203 CSCs are intrinsically resistant to conventional chemotherapy and radiation treatment, and are postulated to contribute to treatment failure and tumor recurrence.204 Therefore, targeting CSCs represents a useful strategy to improve the effectiveness of therapeutic interventions.
The involvement of autophagy in the physiology 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 crucial 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 induction 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 autophagy by ATG4B knockdown altered GSC phenotypes. Inhibition of ATG4B using a specific antagonist, NSC185058, sensitized GSCs to gamma-irradiation and reduced their capability to form glioma tumor spheres. These findings 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 therapy, chemotherapy, and targeted therapies, invoke autophagy in tumor cells209–211 which predominantly functions as a cytoprotective mechanism against therapy-induced stress responses.212,213 Preclinical data from immunodeficient host animal models indicate that pharmacological suppression of autophagy with inhibitors such as 3-methyladenine, CQ, or hydroxychloroquine (HCQ) can augment cytotoxicity in combination with various anticancer treatments. These treatments include conventional chemotherapeutics (e.g., cisplatin,214 5-fluorouracil,215 temozolomide,216 and epirubicin217), radiation therapy,218,219 targeted agents (e.g., gefitinib22), and antiangiogenic 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 chemotherapy,214 ionizing radiation,222 and antiangiogenic therapy.142,223 The mechanism of autophagy-mediated cancer therapy resistance includes removal of genotoxic 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 benefits from the cytoprotective effects of autophagy, and therapy-induced autophagy in tumor cells can cause immunogenic cell death, leading to efficient 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 confirming the function 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 radiation.234 In most cases, autophagy supports the survival of cancer cells in anticancer therapy; however, under certain conditions it fosters cell death.235 In any case, strategies aimed at the modulation of autophagy bear the potential to improve the efficacy of chemotherapy and radiation therapy. The optimal strategy seems to depend on tumor type, stage, genetic context, and specific treatment.236 Additionally, autophagy manipulation might sensitize resistant cancer types to the cytotoxic effects of treatment. Furthermore, a combination of autophagy modulators and conventional treatments may sensitize cancer cells to cancer therapies.237
Sensitization of tumor cells to therapies is one of the most researched topics in the autophagy field. There is mounting preclinical evidence that targeting autophagy can enhance the beneficial effects of many cancer therapies. In many cancer types, inhibition of the nucleation step of autophagosome formation with type III PI3K inhibitors [e.g., 3-methyladenine (3-MA) or LY294002] enhances the efficacy of chemotherapy or radiation. For example, inhibition of autophagy using 3-MA enhanced cytotoxicity of radiotherapy in human esophageal squamous carcinoma cells.238 Similarly, treatment of 3-MA enhanced the efficacy of the Gli inhibitor GANT-61 and increased apoptotic cell death.239 Many preclinical studies indicate that lysosomotropic agents (e.g., CQ and HCQ) prevent lysosome acidification and block autolysosome formation, thereby augmenting the effect of chemotherapies and radiotherapy on various types of cancers.237,240 For example, in non-small-cell lung cancer, bevacizumab in combination with CQ increases the efficacy of cancer treatment.241 However, it should be noted that CQ and HCQ are not specific autophagy inhibitors and they may impact biological processes other than autophagy. These may include lysosomal membrane permeabilization, normalization of tumor vasculature, 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 kinase 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 flux in tumor environments due to a decrease in drug uptake by cells. Therefore, more potent and selective autophagy inhibitors are urgently needed.
A specific 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 lipidation and recycling.246 Inhibition of autophagy with NSC185058 had a negative impact on the development of Saos-2 osteosarcoma tumors in vivo.245 Additionally, NSC185058 in combination with radiotherapy markedly slows tumor growth and provides survival benefits in mice with intracranial glioblastoma xenografts.143 ATG4 proteases are emerging as potential pharmaceutical targets for the treatment of aggressive cancers, such as osteosarcoma and glioblastoma.
While significant 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 specific drugs targeting autophagy at the preclinical stage (e.g., ATG4B inhibitors) is highly warranted. This can help maximize the potential 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 combination with other therapies for cancer treatment. However, 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 responsible for observed therapeutic benefits.247 It is therefore important to develop more potent and selective autophagy inhibitors to improve our understanding of autophagy and expand our therapeutic options for multiple diseases.
ACKNOWLEDGMENT
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 Fellowship Award from the Robert H. Lurie Comprehensive 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 Foundation of China grants (81372704, 81572467) to H. Feng; Program for Professor of Special Appointment (Eastern Scholar) at Shanghai Institutions of Higher Learning (2014024), Shanghai Municipal Education Commission—Gaofeng Clinical Medicine Grant Support (20161310), New Hundred Talent Program (Outstanding Academic Leader) at the Shanghai Municipal Health Bureau (2017BR021), and the State Key Laboratory of Oncogenes and Related Genes in China (91–17-25) to H. Feng.
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 transition
- CSC
cancer stem cell
- CQ
chloroquine
REFERENCES
- 1.Lamb CA, Yoshimori T, Tooze SA. The autophagosome: origins unknown, biogenesis complex. Nat Rev Mol Cell Biol 2013;14(12):759–74. [DOI] [PubMed] [Google Scholar]
- 2.Menzies FM, Fleming A, Rubinsztein DC. Compromised autophagy and neurodegenerative diseases. Nat Rev Neurosci 2015;16(6):345–57. [DOI] [PubMed] [Google Scholar]
- 3.Vakifahmetoglu-Norberg H, Xia HG, Yuan J. Pharmacologic agents targeting autophagy. J Clin Invest. 2015;125(1):5–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Galluzzi L, Pietrocola F, Levine B, Kroemer G. Metabolic control of autophagy. Cell. 2014;159(6):1263–76. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Kaur J, Debnath J. Autophagy at the crossroads of catabolism and anabolism. Nat Rev Mol Cell Biol 2015;16(8):461–72. [DOI] [PubMed] [Google Scholar]
- 6.Nixon RA. The role of autophagy in neurodegenerative disease. Nat Med. 2013;19(8):983–97. [DOI] [PubMed] [Google Scholar]
- 7.White E Deconvoluting the context-dependent role for autophagy in cancer. Nat Rev Cancer. 2012;12(6):401–10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.White E The role for autophagy in cancer. J Clin Invest. 2015;125(1):42–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Klionsky DJ, Baehrecke EH, Brumell JH, Chu CT, Codogno P, Cuervo AM, Debnath J, Deretic V, Elazar Z, Eskelinen 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-related molecules and processes (2nd edition). Autophagy. 2011;7(11):1273–94. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 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 autophagosome formation. EMBO J. 2001;20(21):5971–81. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Axe EL, Walker SA, Manifava M, Chandra P, Roderick HL, Habermann A, Griffiths G, Ktistakis NT. Autophagosome formation from membrane compartments enriched in phosphatidylinositol 3-phosphate and dynamically connected to the endoplasmic reticulum. J Cell Biol 2008;182(4):685–701. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Gonzalez A, Hall MN. Nutrient sensing and TOR signaling in yeast and mammals. EMBO J. 2017;36(4): 397–408. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Saxton RA, Sabatini DM. mTOR Signaling in growth, metabolism, and disease. Cell. 2017;169(2):361–71. [DOI] [PubMed] [Google Scholar]
- 14.Alers S, Loffler AS, Wesselborg S, Stork B. Role of AMPK-mTOR-Ulk1/2 in the regulation of autophagy: cross talk, shortcuts, and feedbacks. Mol Cell Biol. 2012;32(1):2–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Mihaylova MM, Shaw RJ. The AMPK signalling pathway coordinates cell growth, autophagy and metabolism. Nat Cell Biol. 2011;13(9):1016–23. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Hurley JH, Young LN. Mechanisms of autophagy initiation. Annu Rev Biochem. 2017;86:225–44. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 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. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 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. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Egan D, Kim J, Shaw RJ, Guan KL. The autophagy initiating kinase ULK1 is regulated via opposing phosphorylation by AMPK and mTOR. Autophagy. 2011;7(6) :643–4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 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 energy sensing to mitophagy. Science. 2011;331(6016): 456–61. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Gwinn DM, Shackelford DB, Egan DF, Mihaylova MM, Mery A, Vasquez DS, Turk BE, Shaw RJ. AMPK phosphorylation of raptor mediates a metabolic checkpoint. Mol Cell. 2008;30(2):214–26. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 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. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Backer JM. The intricate regulation and complex functions of the Class III phosphoinositide 3-kinase Vps34. Biochem J. 2016;473(15):2251–71. [DOI] [PubMed] [Google Scholar]
- 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. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Taguchi-Atarashi N, Hamasaki M, Matsunaga K, Omori H, Ktistakis NT, Yoshimori T, Noda T. Modulation of local PtdIns3P levels by the PI phosphatase MTMR3 regulates constitutive autophagy. Traffic. 2010;11(4):468–78. [DOI] [PubMed] [Google Scholar]
- 26.Dooley HC, Razi M, Polson HE, Girardin SE, Wilson MI, Tooze SA. WIPI2 links LC3 conjugation with PI3P, autophagosome formation, and pathogen clearance by recruiting Atg12–5-16L1. Mol Cell. 2014;55(2):238–52. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Geng J, Klionsky DJ. The Atg8 and Atg12 ubiquitin-like conjugation systems in macroautophagy. Protein modifications: beyond the usual suspects’ review series. EMBO Rep. 2008;9(9):859–64. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Bento CF, Renna M, Ghislat G, Puri C, Ashkenazi A, Vicinanza M, Menzies FM, Rubinsztein DC. Mammalian autophagy: how does it work? Annu Rev Biochem. 2016;85:685–713. [DOI] [PubMed] [Google Scholar]
- 29.Nakatogawa H Two ubiquitin-like conjugation systems that mediate membrane formation during autophagy. Essays Biochem. 2013;55:39–50. [DOI] [PubMed] [Google Scholar]
- 30.Mizushima N, Noda T, Yoshimori T, Tanaka Y, Ishii T, George MD, Klionsky DJ, Ohsumi M, Ohsumi Y. A protein conjugation system essential for autophagy. Nature. 1998;395(6700):395–8. [DOI] [PubMed] [Google Scholar]
- 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. [DOI] [PubMed] [Google Scholar]
- 32.Slobodkin MR, Elazar Z. The Atg8 family: multifunctional ubiquitin-like key regulators of autophagy. Essays Biochem. 2013;55:51–64. [DOI] [PubMed] [Google Scholar]
- 33.Li M, Hou Y, Wang J, Chen X, Shao ZM, Yin XM. Kinetics comparisons of mammalian Atg4 homologues indicate selective preferences toward diverse Atg8 substrates. J Biol Chem. 2011;286(9):7327–38. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Tanida I, Ueno T, Kominami E. LC3 conjugation system in mammalian autophagy. Int J Biochem Cell Biol. 2004;36(12):2503–18. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Schaaf MB, Keulers TG, Vooijs MA, Rouschop KM. LC3/GABARAP family proteins: autophagy-(un)related functions. FASEB J. 2016;30(12):3961–78. [DOI] [PubMed] [Google Scholar]
- 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. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Weidberg H, Shpilka T, Shvets E, Abada A, Shimron F, Elazar Z. LC3 and GATE-16 N termini mediate membrane fusion processes required for autophagosome biogenesis. Dev Cell. 2011;20(4):444–54. [DOI] [PubMed] [Google Scholar]
- 38.Reggiori F, Ungermann C. Autophagosome maturation and fusion. J Mol Biol. 2017;429(4):486–96. [DOI] [PubMed] [Google Scholar]
- 39.Ganley IG. Autophagosome maturation and lysosomal fusion. Essays Biochem. 2013;55:65–78. [DOI] [PubMed] [Google Scholar]
- 40.Nakamura S, Yoshimori T. New insights into autophagosome-lysosome fusion. J Cell Sci. 2017;130(7):1209–16. [DOI] [PubMed] [Google Scholar]
- 41.Kriegenburg F, Ungermann C, Reggiori F. Coordination of autophagosome-lysosome fusion by Atg8 family members. Curr Biol. 2018;28(8):R512–8. [DOI] [PubMed] [Google Scholar]
- 42.Morani F, Titone R, Pagano L, Galetto A, Alabiso O, Aimaretti G, Isidoro C. Autophagy and thyroid carcinogenesis: genetic and epigenetic links. Endocr Relat Cancer. 2014;21(1):R13–29. [DOI] [PubMed] [Google Scholar]
- 43.Fullgrabe J, Klionsky DJ, Joseph B. The return of the nucleus: transcriptional and epigenetic control of autophagy. Nat Rev Mol Cell Biol. 2014;15(1):65–74. [DOI] [PubMed] [Google Scholar]
- 44.Boehrer S, Lainey E, Kroemer G. Coordinated epigenetic regulation of autophagy and apoptosis. Cell Cycle. 2011;10(17):2832–3. [PubMed] [Google Scholar]
- 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, Kozikowski AP, Koenig A, Billadeau DD. Epigenetic regulation of autophagy by the methyltransferase G9a. Mol Cell Biol. 2013;33(20):3983–93. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Xu Z, Klionsky DJ. The AMPK-SKP2-CARM1 axis links nutrient sensing to transcriptional and epigenetic regulation of autophagy. Ann Transl Med. 2016;4(Suppl 1):S7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Wu BJ, Zhao LX, Zhu CC, Chen YL, Wei MY, Bao SQ, Sun SC, Li XH. Altered apoptosis/autophagy and epigenetic modifications cause the impaired postimplantation octaploid embryonic development in mice. Cell Cycle. 2017;16(1):82–90. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 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 methyltransferase EZH2 through an MTOR-dependent pathway. Autophagy. 2015;11(12):2309–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Sui X, Zhu J, Zhou J, Wang X, Li D, Han W, Fang Y, Pan H. Epigenetic modifications as regulatory elements of autophagy in cancer. Cancer Lett. 2015;360(2):106–13. [DOI] [PubMed] [Google Scholar]
- 50.Shin HR, Kim H, Kim KI, Baek SH. Epigenetic and transcriptional regulation of autophagy. Autophagy. 2016;12(11):2248–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 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. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 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. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Baek SH, Kim KI. Epigenetic Control of autophagy: nuclear events gain more attention. Mol Cell. 2017;65(5):781–5. [DOI] [PubMed] [Google Scholar]
- 54.Shin HJ, Kim H, Oh S, Lee JG, Kee M, Ko HJ, Kweon MN, Won KJ, Baek SH. AMPK-SKP2-CARM1 signalling cascade in transcriptional regulation of autophagy. Nature. 2016;534(7608):553–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.An PNT, Shimaji K, Tanaka R, Yoshida H, Kimura H, Fukusaki E, Yamaguchi M. Epigenetic regulation of starvation-induced autophagy in Drosophila by histone methyltransferase G9a. Sci Rep. 2017;7(1):7343. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Ren A, Qiu Y, Cui H, Fu G. Inhibition of H3K9 methyltransferase G9a induces autophagy and apoptosis in oral squamous cell carcinoma. Biochem Biophys Res Commun. 2015;459(1):10–7. [DOI] [PubMed] [Google Scholar]
- 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 acetyltransferase hMOF regulates the outcome of autophagy. Nature. 2013;500(7463):468–71. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Ke XX, Zhang D, Zhu S, Xia Q, Xiang Z, Cui H. Inhibition of H3K9 methyltransferase G9a repressed cell proliferation and induced autophagy in neuroblastoma cells. PLoS One. 2014;9(9):e106962. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Liu TP, Lo HL, Wei LS, Hsiao HH, Yang PM. S-Adenosyl-L-methionine-competitive inhibitors of the histone methyltransferase EZH2 induce autophagy and enhance drug sensitivity in cancer cells. Anticancer Drugs. 2015;26(2):139–47. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Hsieh YY, Lo HL, Yang PM. EZH2 inhibitors transcriptionally upregulate cytotoxic autophagy and cytoprotective unfolded protein response in human colorectal cancer cells. Am J Cancer Res. 2016;6(8):1661–80. [PMC free article] [PubMed] [Google Scholar]
- 61.Chen H, Fan M, Pfeffer LM, Laribee RN. The histone H3 lysine 56 acetylation pathway is regulated by target of rapamycin (TOR) signaling and functions directly in ribosomal RNA biogenesis. Nucleic Acids Res. 2012;40(14):6534–46. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 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. Heterochromatin and tri-methylated lysine 20 of histone H4 in animals. J Cell Sci. 2004;117(Pt 12):2491–501. [DOI] [PubMed] [Google Scholar]
- 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. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 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. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Sakamaki JI, Ryan KM. Transcriptional regulation of autophagy and lysosomal function by bromodomain protein BRD4. Autophagy. 2017;13(11):2006–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 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. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Moreau K, Ghislat G, Hochfeld W, Renna M, Zavodszky E, Runwal G, Puri C, Lee S, Siddiqi F, Menzies FM, Ravikumar B, Rubinsztein DC. Transcriptional regulation of Annexin A2 promotes starvation-induced autophagy. Nat Commun. 2015;6:8045. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Hsieh MJ, Hsieh YH, Lin CW, Chen MK, Yang SF, Chiou HL. Transcriptional regulation of Mcl-1 plays an important role of cellular protective effector of vincristine-triggered autophagy in oral cancer cells. Expert Opin Ther Targets. 2015;19(4):455–70. [DOI] [PubMed] [Google Scholar]
- 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. [DOI] [PubMed] [Google Scholar]
- 70.Miao LJ, Huang FX, Sun ZT, Zhang RX, Huang SF, Wang J. Stat3 inhibits Beclin 1 expression through recruitment of HDAC3 in nonsmall cell lung cancer cells. Tumour Biol. 2014;35(7):7097–103. [DOI] [PubMed] [Google Scholar]
- 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 autophagy in vivo. FEBS J. 2014;281(20):4557–67. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 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. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Feng Y, Ke C, Tang Q, Dong H, Zheng X, Lin W, Ke J, Huang J, Yeung SC, Zhang H. Metformin promotes autophagy and apoptosis in esophageal squamous cell carcinoma by downregulating Stat3 signaling. Cell Death Dis. 2014;5:e1088. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 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. Autophagy. 2013;9(3):415–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Shen S, Niso-Santano M, Adjemian S, Takehara T, Malik 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. [DOI] [PubMed] [Google Scholar]
- 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 Commun. 2018;500(3):589–96. [DOI] [PubMed] [Google Scholar]
- 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 inhibition of autophagy. Mol Cancer. 2018;17(1):6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Sardiello M, Palmieri M, di Ronza A, Medina DL, Valenza M, Gennarino VA, Di Malta C, Donaudy F, Embrione V, Polishchuk RS, Banfi S, Parenti G, Cattaneo E, Ballabio A. A gene network regulating lysosomal biogenesis and function. Science. 2009;325(5939):473–7. [DOI] [PubMed] [Google Scholar]
- 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 apoptosis in human chondrosarcoma cells. Cell Physiol Biochem. 2017;43(4):1487–502. [DOI] [PubMed] [Google Scholar]
- 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. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Hsu CL, Lee EX, Gordon KL, Paz EA, Shen WC, Ohnishi K, Meisenhelder J, Hunter T, La Spada AR. MAP4K3 mediates amino acid-dependent regulation of autophagy via phosphorylation of TFEB. Nat Commun. 2018;9(1):942. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 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 endothelial cell autophagy. J Biol Chem. 2017;292(39): 16211–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Palmieri M, Pal R, Sardiello M. AKT modulates the autophagy-lysosome pathway via TFEB. Cell Cycle. 2017;16(13):1237–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 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. Bromodomain protein BRD4 is a transcriptional repressor of autophagy and lysosomal function. Mol Cell. 2017;66(4):517–32. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Uribesalgo I, Ballare C, Di Croce L. Polycomb regulates NF-kappaB signaling in cancer through miRNA. Cancer Cell. 2012;21(1):5–7. [DOI] [PubMed] [Google Scholar]
- 86.Ren Y, Chen Y, Liang X, Lu Y, Pan W, Yang M. MiRNA-638 promotes autophagy and malignant phenotypes of cancer cells via directly suppressing DACT3. Cancer Lett. 2017;390:126–36. [DOI] [PubMed] [Google Scholar]
- 87.Zhang P, Zhang H. Autophagy modulates miRNA-mediated gene silencing and selectively degrades AIN-1/GW182 in C. elegans. EMBO Rep. 2013;14(6):568–76. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Gibbings D, Mostowy S, Voinnet O. Autophagy selectively regulates miRNA homeostasis. Autophagy. 2013;9(5):781–3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Ucar A, Gupta SK, Fiedler J, Erikci E, Kardasinski M, Batkai S, Dangwal S, Kumarswamy R, Bang C, Holzmann 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, Chowdhury K, Thum T. The miRNA-212/132 family regulates both cardiac hypertrophy and cardiomyocyte autophagy. Nat Commun. 2012;3:1078. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 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. [DOI] [PubMed] [Google Scholar]
- 91.Zhu H, Wu H, Liu X, Li B, Chen Y, Ren X, Liu CG, Yang JM. Regulation of autophagy by a beclin 1-targeted microRNA, miR-30a, in cancer cells. Autophagy. 2009;5(6):816–23. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 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 myeloid leukemia cells. Leukemia. 2012;26(8):1752–60. [DOI] [PubMed] [Google Scholar]
- 93.Fu Z, Luo W, Wang J, Peng T, Sun G, Shi J, Li Z, Zhang B. Malat1 activates autophagy and promotes cell proliferation by sponging miR-101 and upregulating STMN1, RAB5A and ATG4D expression in glioma. Biochem Biophys Res Commun. 2017;492(3):480–6. [DOI] [PubMed] [Google Scholar]
- 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. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Xu Y, An Y, Wang Y, Zhang C, Zhang H, Huang C, Jiang H, Wang X, Li X. miR-101 inhibits autophagy and enhances cisplatin-induced apoptosis in hepatocellular carcinoma cells. Oncol Rep. 2013;29(5):2019–24. [DOI] [PubMed] [Google Scholar]
- 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. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 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. Autophagy. 2012;8(2):165–76. [DOI] [PubMed] [Google Scholar]
- 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. MicroRNA 23b regulates autophagy associated with radioresistance of pancreatic cancer cells. Gastroenterology. 2013;145(5):1133–43. [DOI] [PubMed] [Google Scholar]
- 99.Schmitt AM, Chang HY. Long noncoding RNAs in cancer pathways. Cancer Cell. 2016;29(4):452–63. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 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. [DOI] [PubMed] [Google Scholar]
- 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. [DOI] [PubMed] [Google Scholar]
- 102.Xia H, Qu XL, Liu LY, Qian DH, Jing HY. LncRNA MEG3 promotes the sensitivity of vincristine by inhibiting autophagy in lung cancer chemotherapy. Eur Rev Med Pharmacol Sci. 2018;22(4):1020–7. [DOI] [PubMed] [Google Scholar]
- 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. [DOI] [PubMed] [Google Scholar]
- 104.Xiu YL, Sun KX, Chen X, Chen S, Zhao Y, Guo QG, Zong ZH. Upregulation of the lncRNA Meg3 induces autophagy to inhibit tumorigenesis and progression of epithelial ovarian carcinoma by regulating activity of ATG3. Oncotarget. 2017;8(19):31714–25. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 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. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Ying L, Huang Y, Chen H, Wang Y, Xia L, Chen Y, Liu Y, Qiu F. Downregulated MEG3 activates autophagy and increases cell proliferation in bladder cancer. Mol Biosyst. 2013;9(3):407–11. [DOI] [PubMed] [Google Scholar]
- 107.Kung CP, Budina A, Balaburski G, Bergenstock MK, Murphy M. Autophagy in tumor suppression and cancer therapy. Crit Rev Eukaryot Gene Expr. 2011;21(1): 71–100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 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. [DOI] [PubMed] [Google Scholar]
- 109.Furuya N, Yu F, Byfield M, Pattingre S, Levine B. The evolutionarily conserved domain of Beclin 1 is required for Vps34 binding, autophagy and tumor suppressor function. Autophagy. 2005;1(1):46–52. [DOI] [PubMed] [Google Scholar]
- 110.Liang XH, Kleeman LK, Jiang HH, Gordon G, Goldman JE, Berry G, Herman B, Levine B. Protection against fatal Sindbis virus encephalitis by beclin, a novel Bcl-2-interacting protein. J Virol. 1998;72(11):8586–96. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Pattingre S, Tassa A, Qu X, Garuti R, Liang XH, Mizushima N, Packer M, Schneider MD, Levine B. Bcl-2 antiapoptotic proteins inhibit Beclin 1-dependent autophagy. Cell. 2005;122(6):927–39. [DOI] [PubMed] [Google Scholar]
- 112.Yue Z, Jin S, Yang C, Levine AJ, Heintz N. Beclin 1, an autophagy gene essential for early embryonic development, is a haploinsufficient tumor suppressor. Proc Natl Acad Sci U S A. 2003;100(25):15077–82. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Qu X, Yu J, Bhagat G, Furuya N, Hibshoosh H, Troxel A, Rosen J, Eskelinen EL, Mizushima N, Ohsumi Y, Cattoretti G, Levine B. Promotion of tumorigenesis by heterozygous disruption of the beclin 1 autophagy gene. J Clin Invest. 2003;112(12):1809–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Edinger AL, Thompson CB. Defective autophagy leads to cancer. Cancer Cell. 2003;4(6):422–4. [DOI] [PubMed] [Google Scholar]
- 115.Takahashi Y, Coppola D, Matsushita N, Cualing HD, Sun M, Sato Y, Liang C, Jung JU, Cheng JQ, Mule JJ, Pledger WJ, Wang HG. Bif-1 interacts with Beclin 1 through UVRAG and regulates autophagy and tumorigenesis. Nat Cell Biol. 2007;9(10):1142–51. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 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. [DOI] [PubMed] [Google Scholar]
- 117.Balakrishnan A, von Neuhoff N, Rudolph C, Kamphues K, Schraders M, Groenen P, van Krieken JH, Callet-Bauchu E, Schlegelberger B, Steinemann D. Quantitative microsatellite analysis to delineate the commonly deleted region 1p22.3 in mantle cell lymphomas. Genes Chromo Cancer. 2006;45(10):883–92. [DOI] [PubMed] [Google Scholar]
- 118.Lee JW, Jeong EG, Soung YH, Nam SW, Lee JY, Yoo NJ, Lee SH. Decreased expression of tumour suppressor Baxinteracting factor-1 (Bif-1), a Bax activator, in gastric carcinomas. Pathology. 2006;38(4):312–5. [DOI] [PubMed] [Google Scholar]
- 119.Takamura A, Komatsu M, Hara T, Sakamoto A, Kishi C, Waguri S, Eishi Y, Hino O, Tanaka K, Mizushima N. Autophagy-deficient mice develop multiple liver tumors. Genes Dev. 2011;25(8):795–800. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Narendra D, Tanaka A, Suen DF, Youle RJ. Parkin is recruited selectively to impaired mitochondria and promotes their autophagy. J Cell Biol. 2008;183(5):795–803. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Youle RJ, Narendra DP. Mechanisms of mitophagy. Nat Rev Mol Cell Biol. 2011;12(1):9–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Veeriah S, Morris LG, Solit D, Chan TA. The familial Parkinson disease gene PARK2 is a multisite tumor suppressor on chromosome 6q25.2–27 that regulates cyclin E. Cell Cycle. 2010;9(8):1451–2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 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 frequently in sporadic colorectal cancer and accelerate adenoma development in Apc mutant mice. Proc Natl Acad Sci U S A. 2010;107(34):15145–50. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Moscat J, Diaz-Meco MT. p62: a versatile multitasker takes on cancer. Trends Biochem Sci. 2012;37(6):230–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Komatsu M, Kurokawa H, Waguri S, Taguchi K, Kobayashi 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. [DOI] [PubMed] [Google Scholar]
- 126.Nioi P, McMahon M, Itoh K, Yamamoto M, Hayes JD. Identification of a novel Nrf2-regulated antioxidant response element (ARE) in the mouse NAD(P)H:quinone oxidoreductase 1 gene: reassessment of the ARE consensus sequence. Biochem J. 2003;374(Pt 2):337–48. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 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. [DOI] [PubMed] [Google Scholar]
- 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 detoxification and tumorigenesis. Nature. 2011;475(7354):106–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 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. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 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. [DOI] [PubMed] [Google Scholar]
- 131.Konstantinopoulos PA, Spentzos D, Fountzilas E, Francoeur N, Sanisetty S, Grammatikos AP, Hecht JL, Cannistra SA. Keap1 mutations and Nrf2 pathway activation in epithelial ovarian cancer. Cancer Res. 2011;71(15): 5081–9. [DOI] [PubMed] [Google Scholar]
- 132.Mathew R, Karantza-Wadsworth V, White E. Role of autophagy in cancer. Nat Rev Cancer. 2007;7(12):961–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Suzuki SW, Onodera J, Ohsumi Y. Starvation induced cell death in autophagy-defective yeast mutants is caused by mitochondria dysfunction. PLoS One. 2011;6(2):e17412. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Onodera J, Ohsumi Y. Autophagy is required for maintenance of amino acid levels and protein synthesis under nitrogen starvation. J Biol Chem. 2005;280(36): 31582–6. [DOI] [PubMed] [Google Scholar]
- 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 autophagy in neural cells causes neurodegenerative disease in mice. Nature. 2006;441(7095):885–9. [DOI] [PubMed] [Google Scholar]
- 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. [DOI] [PubMed] [Google Scholar]
- 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 period. Nature. 2004;432(7020):1032–6. [DOI] [PubMed] [Google Scholar]
- 138.Rabinowitz JD, White E. Autophagy and metabolism. Science. 2010;330(6009):1344–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 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 autophagy for tumor growth. Genes Dev. 2011;25(7):717–29. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Lum JJ, Bauer DE, Kong M, Harris MH, Li C, Lindsten T, Thompson CB. Growth factor regulation of autophagy and cell survival in the absence of apoptosis. Cell. 2005;120(2):237–48. [DOI] [PubMed] [Google Scholar]
- 141.Degenhardt K, Mathew R, Beaudoin B, Bray K, Anderson 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, inflammation, and tumorigenesis. Cancer Cell. 2006;10(1):51–64. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142.Hu YL, DeLay M, Jahangiri A, Molinaro AM, Rose SD, Carbonell WS, Aghi MK. Hypoxia-induced autophagy promotes tumor cell survival and adaptation to antiangiogenic treatment in glioblastoma. Cancer Res. 2012;72(7):1773–83. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 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 Phosphorylation of ATG4B regulates autophagic activity, tumorigenicity, and radio-resistance in glioblastoma. Cancer cell. 2017;32(6):840–55. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144.Strohecker AM, White E. Autophagy promotes BrafV600E-driven lung tumorigenesis by preserving mitochondrial metabolism. Autophagy. 2014;10(2):384–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 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, Rabinowitz JD, White E. Activated Ras requires autophagy to maintain oxidative metabolism and tumorigenesis. Genes Dev. 2011;25(5):460–70. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146.Larki P, Gharib E, Yaghoob Taleghani M, Khorshidi F, Nazemalhosseini-Mojarad E, Asadzadeh Aghdaei H. Coexistence 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. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147.Hu YM, Lu WQ, Chen G, Wang P, Chen Z, Zhou Y, Ogasawara 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 glycolysis. Cell Res. 2012;22(2):399–412. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148.Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell. 2011;144(5):646–74. [DOI] [PubMed] [Google Scholar]
- 149.Schmelzle T, Hall MN. TOR, a central controller of cell growth. Cell. 2000;103(2):253–62. [DOI] [PubMed] [Google Scholar]
- 150.Noda T, Ohsumi Y. Tor, a phosphatidylinositol kinase homologue, controls autophagy in yeast. J Biol Chem. 1998;273(7):3963–6. [DOI] [PubMed] [Google Scholar]
- 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 clearance of protein aggregates in neurodegeneration. Cell Signal. 2014;26(12):2694–701. [DOI] [PubMed] [Google Scholar]
- 152.Feng Z, Zhang H, Levine AJ, Jin S. The coordinate regulation of the p53 and mTOR pathways in cells. Proc Natl Acad Sci U S A. 2005;102(23):8204–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 153.Balaburski GM, Hontz RD, Murphy ME. p53 and ARF: unexpected players in autophagy. Trends Cell Biol. 2010;20(6):363–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 154.Zhong Z, Sanchez-Lopez E, Karin M. Autophagy, inflammation, and immunity: a troika governing cancer and its treatment. Cell. 2016;166(2):288–98. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 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. Epithelial-to-mesenchymal transition is not required for lung metastasis but contributes to chemoresistance. Nature. 2015;527(7579):472–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 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 anoikis resistance and colonization of HCC cells. Autophagy. 2013;9(12):2056–68. [DOI] [PubMed] [Google Scholar]
- 157.Chen S, Han Q, Wang X, Yang M, Zhang Z, Li P, Chen A, Hu C, Li S. IBP-mediated suppression of autophagy promotes growth and metastasis of breast cancer cells via activating mTORC2/Akt/FOXO3a signaling pathway. Cell Death Dis. 2013;4:e842. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 158.Okura R, Nakamura M. Overexpression of autophagy-related beclin-1 in cutaneous squamous cell carcinoma with lymph-node metastasis. Eur J Dermatol. 2011;21(6):1002–3. [DOI] [PubMed] [Google Scholar]
- 159.Kenific CM, Thorburn A, Debnath J. Autophagy and metastasis: another double-edged sword. Curr Opin Cell Biol. 2010;22(2):241–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 160.Li J, Yang B, Zhou Q, Wu Y, Shang D, Guo Y, Song Z, Zheng Q, Xiong J. Autophagy promotes hepatocellular carcinoma cell invasion through activation of epithelial-mesenchymal transition. Carcinogenesis. 2013;34(6):1343–51. [DOI] [PubMed] [Google Scholar]
- 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-mesenchymal transition of colon cancer cells. J Cell Biochem. 2018. DOI: 10.1002/jcb.26912. [DOI] [PubMed] [Google Scholar]
- 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 signaling pathway regulates epithelial-mesenchymal transition in A549 lung cancer cells. FEBS Lett. 2016;590(9): 1365–74. [DOI] [PubMed] [Google Scholar]
- 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 sensitivity in nasopharyngeal carcinoma. Oncol Lett. 2017;13(6): 4147–54. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 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 suppresses epithelial-to-mesenchymal transition involving PI3K/Akt/mTOR and sirtuin 1-mediated signaling pathways in human pancreatic cancer cells. Drug Des Devel Ther. 2015;9:575–601. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 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 inhibitor, induces apoptosis and autophagy but inhibits epithelial to mesenchymal transition in human epithelial ovarian cancer cells. Drug Des Devel Ther. 2015;9:425–64. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 166.Zi D, Zhou ZW, Yang YJ, Huang L, Zhou ZL, He SM, He ZX, Zhou SF. Danusertib induces apoptosis, cell cycle arrest, and autophagy but inhibits epithelial to mesenchymal transition involving PI3K/Akt/mTOR Signaling pathway in human ovarian cancer cells. Int J Mol Sci. 2015;16(11):27228–51. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 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 epithelial-to-mesenchymal transition in human breast cancer cells. Drug Des Devel Ther. 2015;9:1027–62. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 168.Zhao Z, Zhao J, Xue J, Zhao X, Liu P. Autophagy inhibition promotes epithelial-mesenchymal transition through ROS/HO-1 pathway in ovarian cancer cells. Am J Cancer Res. 2016;6(10):2162–77. [PMC free article] [PubMed] [Google Scholar]
- 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 impairs migration and invasion by reversing EMT in glioblastoma cells. Mol Oncol. 2015;9(8):1612–25. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 170.Gugnoni M, Sancisi V, Gandolfi G, Manzotti G, Ragazzi 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. [DOI] [PubMed] [Google Scholar]
- 171.Lo HW, Hsu SC, Xia W, Cao X, Shih JY, Wei Y, Abbruzzese JL, Hortobagyi GN, Hung MC. Epidermal growth factor receptor cooperates with signal transducer and activator of transcription 3 to induce epithelial-mesenchymal transition in cancer cells via up-regulation of TWIST gene expression. Cancer Res. 2007;67(19):9066–76. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 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. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 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-mesenchymal transition in human breast cancer. Cancer Res. 2012;72(13):3238–50. [DOI] [PubMed] [Google Scholar]
- 174.Qiang L, He YY. Autophagy deficiency stabilizes TWIST1 to promote epithelial-mesenchymal transition. Autophagy. 2014;10(10):1864–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 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. Metastatic prostate cancer-associated P62 inhibits autophagy flux and promotes epithelial to mesenchymal transition by sustaining the level of HDAC6. Prostate. 2018;78(6): 426–34. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 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. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 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 identifies nasopharyngeal carcinoma patients at high risk of metastasis for additional induction chemotherapy. Clin Cancer Res. 2018;24(3):648–58. [DOI] [PubMed] [Google Scholar]
- 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 progression by suppressing cyclin A expression. Oncogene. 2017;36(7):885–98. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 179.Pang MF, Georgoudaki AM, Lambut L, Johansson J, Tabor V, Hagikura K, Jin Y, Jansson M, Alexander JS, Nelson 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 chemotaxis. Oncogene. 2016;35(6):748–60. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 180.Kiyono K, Suzuki HI, Matsuyama H, Morishita Y, Komuro A, Kano MR, Sugimoto K, Miyazono K. Autophagy is activated by TGF-beta and potentiates TGF-beta-mediated growth inhibition in human hepatocellular carcinoma cells. Cancer Res. 2009;69(23):8844–52. [DOI] [PubMed] [Google Scholar]
- 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. Oncotarget. 2018;9(5):6433–49. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 182.Zhang C, Zhang X, Xu R, Huang B, Chen AJ, Li C, Wang 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. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- 183.McGowan PM, Kirstein JM, Chambers AF. Micrometastatic disease and metastatic outgrowth: clinical issues and experimental approaches. Future Oncol. 2009;5(7): 1083–98. [DOI] [PubMed] [Google Scholar]
- 184.Chaffer CL, Weinberg RA. A perspective on cancer cell metastasis. Science. 2011;331(6024):1559–64. [DOI] [PubMed] [Google Scholar]
- 185.Kroemer G, Marino G, Levine B. Autophagy and the integrated stress response. Mol Cell. 2010;40(2):280–93. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 186.Lazova R, Camp RL, Klump V, Siddiqui SF, Amaravadi RK, Pawelek JM. Punctate LC3B expression is a common feature of solid tumors and associated with proliferation, metastasis, and poor outcome. Clin Cancer Res. 2012;18(2):370–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 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. [DOI] [PubMed] [Google Scholar]
- 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 vasculogenic mimicry. Tohoku J Exp Med. 2011;223(4):243–51. [DOI] [PubMed] [Google Scholar]
- 189.DeNardo DG, Barreto JB, Andreu P, Vasquez L, Tawfik D, Kolhatkar N, Coussens LM. CD4(+) T cells regulate pulmonary metastasis of mammary carcinomas by enhancing protumor properties of macrophages. Cancer Cell. 2009;16(2):91–102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 190.Bingle L, Brown NJ, Lewis CE. The role of tumour-associated macrophages in tumour progression: implications for new anticancer therapies. J Pathol. 2002;196(3):254–65. [DOI] [PubMed] [Google Scholar]
- 191.Guo W, Giancotti FG. Integrin signalling during tumour progression. Nat Rev Mol Cell Biol. 2004;5(10):816–26. [DOI] [PubMed] [Google Scholar]
- 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. [DOI] [PubMed] [Google Scholar]
- 193.Sosa MS, Bragado P, Aguirre-Ghiso JA. Mechanisms of disseminated cancer cell dormancy: an awakening field. Nat Rev Cancer. 2014;14(9):611–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 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 tumor dormancy in human ovarian cancer cells. J Clin Invest. 2008;118(12):3917–29. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 195.Mowers EE, Sharifi MN, Macleod KF. Autophagy in cancer metastasis. Oncogene. 2017;36(12):1619–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 196.Vander Heiden MG, DeBerardinis RJ. Understanding the intersections between metabolism and cancer biology. Cell. 2017;168(4):657–69. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 197.Vander Heiden MG, Cantley LC, Thompson CB. Understanding the Warburg effect: the metabolic requirements of cell proliferation. Science. 2009;324(5930):1029–33. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 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. [DOI] [PubMed] [Google Scholar]
- 199.Kimmelman AC, White E. Autophagy and tumor metabolism. Cell Metab. 2017;25(5):1037–43. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 200.Janji B, Berchem G, Chouaib S. Targeting autophagy in the tumor microenvironment: new challenges and opportunities for regulating tumor immunity. Front Immunol. 2018;9:887. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 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. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 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. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 203.Lathia JD, Mack SC, Mulkearns-Hubert EE, Valentim CL, Rich JN. Cancer stem cells in glioblastoma. Genes Dev. 2015;29(12):1203–17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 204.Ojha R, Bhattacharyya S, Singh SK. Autophagy in cancer stem cells: a potential link between chemoresistance, recurrence, and metastasis. Biores Open Access. 2015;4(1):97–108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 205.Lomonaco SL, Finniss S, Xiang C, Decarvalho A, Umansky F, Kalkanis SN, Mikkelsen T, Brodie C. The induction of autophagy by gamma-radiation contributes to the radioresistance of glioma stem cells. Int J Cancer. 2009;125(3):717–22. [DOI] [PubMed] [Google Scholar]
- 206.Cufi 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. [DOI] [PubMed] [Google Scholar]
- 207.Wolf J, Dewi DL, Fredebohm J, Muller-Decker K, Flechtenmacher C, Hoheisel JD, Boettcher M. A mammosphere formation RNAi screen reveals that ATG4A promotes a breast cancer stem-like phenotype. Breast Cancer Res. 2013;15(6):R109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 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. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 209.Paglin S, Hollister T, Delohery T, Hackett N, McMahill M, Sphicas E, Domingo D, Yahalom J. A novel response of cancer cells to radiation involves autophagy and formation of acidic vesicles. Cancer Res. 2001;61(2): 439–44. [PubMed] [Google Scholar]
- 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. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 211.Gewirtz DA. The autophagic response to radiation: relevance for radiation sensitization in cancer therapy. Radiat Res. 2014;182(4):363–7. [DOI] [PubMed] [Google Scholar]
- 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. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 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. [DOI] [PubMed] [Google Scholar]
- 214.Zhao XG, Sun RJ, Yang XY, Liu DY, Lei DP, Jin T, Pan XL. Chloroquine-enhanced efficacy of cisplatin in the treatment of hypopharyngeal carcinoma in xenograft mice. PLoS One. 2015;10(4):e0126147. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 215.Li J, Hou N, Faried A, Tsutsumi S, Kuwano H. Inhibition of autophagy augments 5-fluorouracil chemotherapy in human colon cancer In vitro and in vivo model. Eur J Cancer. 2010;46(10):1900–9. [DOI] [PubMed] [Google Scholar]
- 216.Golden EB, Cho HY, Jahanian A, Hofman FM, Louie SG, Schonthal AH, Chen TC. Chloroquine enhances temozolomide cytotoxicity in malignant gliomas by blocking autophagy. Neurosurg Focus. 2014;37(6):E12. [DOI] [PubMed] [Google Scholar]
- 217.Chittaranjan S, Bortnik S, Dragowska WH, Xu J, Abeysundara N, Leung A, Go NE, DeVorkin L, Weppler SA, Gelmon K, Yapp DT, Bally MB, Gorski SM. Autophagy inhibition augments the anticancer effects of epirubicin treatment in anthracycline-sensitive and -resistant triple-negative breast cancer. Clin Cancer Res. 2014;20(12):3159–73. [DOI] [PubMed] [Google Scholar]
- 218.Tseng HC, Liu WS, Tyan YS, Chiang HC, Kuo WH, Chou FP. Sensitizing effect of 3-methyladenine on radiation-induced cytotoxicity in radio-resistant HepG2 cells In vitro and in tumor xenografts. Chem Biol Interact. 2011;192(3):201–8. [DOI] [PubMed] [Google Scholar]
- 219.Chen Y, Li X, Guo L, Wu X, He C, Zhang S, Xiao Y, Yang Y, Hao D. Combining radiation with autophagy inhibition enhances suppression of tumor growth and angiogenesis in esophageal cancer. Mol Med Rep. 2015;12(2): 1645–52. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 220.Dragowska WH, Weppler SA, Wang JC, Wong LY, Kapanen AI, Rawji JS, Warburton C, Qadir MA, Donohue E, Roberge M, Gorski SM, Gelmon KA, Bally MB. Induction of autophagy is an early response to gefitinib and a potential therapeutic target in breast cancer. PLoS One. 2013;8(10):e76503. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 221.Selvakumaran M, Amaravadi RK, Vasilevskaya IA, O’Dwyer PJ. Autophagy inhibition sensitizes colon cancer cells to antiangiogenic and cytotoxic therapy. Clin Cancer Res. 2013;19(11):2995–3007. [DOI] [PubMed] [Google Scholar]
- 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 radiosensitizes In vitro, yet reduces radioresponses in vivo due to deficient immunogenic signalling. Cell Death Differ. 2014;21(1):92–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 223.Mohan N, Chakrabarti M, Banik NL, Ray SK. Combination of LC3 shRNA plasmid transfection and genistein treatment inhibited autophagy and increased apoptosis in malignant neuroblastoma in cell culture and animal models. PLoS One. 2013;8(10):e78958. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 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 carcinoma contributes to the tolerance of oxaliplatin via reactive oxygen species modulation. Clin Cancer Res. 2011;17(19):6229–38. [DOI] [PubMed] [Google Scholar]
- 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. [DOI] [PubMed] [Google Scholar]
- 226.Firat E, Weyerbrock A, Gaedicke S, Grosu AL, Niedermann G. Chloroquine or chloroquine-PI3K/Akt pathway inhibitor combinations strongly promote gamma-irradiation-induced cell death in primary stem-like glioma cells. PLoS One. 2012;7(10):e47357. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 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. [DOI] [PubMed] [Google Scholar]
- 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. [DOI] [PubMed] [Google Scholar]
- 229.Levy JMM, Towers CG, Thorburn A. Targeting autophagy in cancer. Nat Rev Cancer. 2017;17(9):528–42. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 230.Galluzzi L, Buque A, Kepp O, Zitvogel L, Kroemer G. Immunological effects of conventional chemotherapy and targeted anticancer agents. Cancer Cell. 2015;28(6):690–714. [DOI] [PubMed] [Google Scholar]
- 231.Galluzzi L, Pietrocola F, Bravo-San Pedro JM, Amaravadi 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. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 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, Galluzzi 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. [DOI] [PubMed] [Google Scholar]
- 233.Galluzzi L, Bravo-San Pedro JM, Demaria S, Formenti SC, Kroemer G. Activating autophagy to potentiate immunogenic chemotherapy and radiation therapy. Nat Rev Clin Oncol. 2017;14(4):247–58. [DOI] [PubMed] [Google Scholar]
- 234.Fulda S Targeting autophagy for the treatment of cancer. Biol Chem. 2018. [DOI] [PubMed] [Google Scholar]
- 235.Fulda S Autophagy in cancer therapy. Front Oncol. 2017;7:128. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 236.Kimmelman AC. The dynamic nature of autophagy in cancer. Genes Dev. 2011;25(19):1999–2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 237.Chude CI, Amaravadi RK. Targeting autophagy in cancer: update on clinical trials and novel inhibitors. Int J Mol Sci. 2017;18(6):160.e1–160.e13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 238.Chen YS, Song HX, Lu Y, Li X, Chen T, Zhang Y, Xue JX, Liu H, Kan B, Yang G, Fu T. Autophagy inhibition contributes to radiation sensitization of esophageal squamous carcinoma cells. Dis Esophagus. 2011;24(6): 437–43. [DOI] [PubMed] [Google Scholar]
- 239.Wang J, Gu S, Huang J, Chen S, Zhang Z, Xu M. Inhibition of autophagy potentiates the efficacy of Gli inhibitor GANT-61 in MYCN-amplified neuroblastoma cells. BMC Cancer. 2014;14:768. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 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. [DOI] [PubMed] [Google Scholar]
- 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 Biochem. 2018;119(8):7022–31. [DOI] [PubMed] [Google Scholar]
- 242.Maycotte P, Aryal S, Cummings CT, Thorburn J, Morgan MJ, Thorburn A. Chloroquine sensitizes breast cancer cells to chemotherapy independent of autophagy. Autophagy. 2012;8(2):200–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 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. Tumor vessel normalization by chloroquine independent of autophagy. Cancer Cell. 2014;26(2):190–206. [DOI] [PubMed] [Google Scholar]
- 244.Balic A, Sorensen MD, Trabulo SM, Sainz B Jr., Cioffi 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. [DOI] [PubMed] [Google Scholar]
- 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. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 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. [DOI] [PubMed] [Google Scholar]
- 247.Rybstein MD, Bravo-San Pedro JM, Kroemer G, Galluzzi L. The autophagic network and cancer. Nat Cell Biol. 2018;20(3):243–51. [DOI] [PubMed] [Google Scholar]