Skip to main content
Erschienen in: Molecular Cancer 1/2020

Open Access 01.12.2020 | Review

Autophagy and autophagy-related proteins in cancer

verfasst von: Xiaohua Li, Shikun He, Binyun Ma

Erschienen in: Molecular Cancer | Ausgabe 1/2020

Abstract

Autophagy, as a type II programmed cell death, plays crucial roles with autophagy-related (ATG) proteins in cancer. Up to now, the dual role of autophagy both in cancer progression and inhibition remains controversial, in which the numerous ATG proteins and their core complexes including ULK1/2 kinase core complex, autophagy-specific class III PI3K complex, ATG9A trafficking system, ATG12 and LC3 ubiquitin-like conjugation systems, give multiple activities of autophagy pathway and are involved in autophagy initiation, nucleation, elongation, maturation, fusion and degradation. Autophagy plays a dynamic tumor-suppressive or tumor-promoting role in different contexts and stages of cancer development. In the early tumorigenesis, autophagy, as a survival pathway and quality-control mechanism, prevents tumor initiation and suppresses cancer progression. Once the tumors progress to late stage and are established and subjected to the environmental stresses, autophagy, as a dynamic degradation and recycling system, contributes to the survival and growth of the established tumors and promotes aggressiveness of the cancers by facilitating metastasis. This indicates that regulation of autophagy can be used as effective interventional strategies for cancer therapy.
Hinweise
Xiaohua Li and Shikun He contributed equally to this work.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
As2O3
Arsenic Trioxide
ATG
autophagy-related proteins, such as ATG1, ATG4, ATG5 ATG7 etc.
BH3
Bcl-2 homology 3 domain
CCD
Coiled-coil domain
Cvt
Cytoplasm to vacuole targeting
ECD
Evolutionarily conserved domain
ECM
Extracellular matrix
ER
Endoplasmic reticulum
FIP200
FAK-family interacting protein of 200 kDa
GABARAP
γ-aminobutyric-acid-type-A-receptor-associated protein
GATE16
Golgi-associated ATPase enhancer of 16 kDa
HCC
Hepatocellular carcinoma
LC3
Microtubule-associated protein 1A/1B-light chain 3
LC3-I
The cytosolic form of LC3
LC3-II
The conjugate form of LC3-I with phosphatidylethanolamine (PE) (LC3-I/PE)
LIR
LC3 interaction region
p62/SQSTM1
a ubiquitin-binding protein p62, sequestosome 1
PAS
Pre-autophagosomal structure
PI
Phosphatidylinositol
PI3KC3-CI and PI3KC3-CII
Class III phosphatidylinositol 3-kinase complex I and II
ProLC3
The precursor protein form of LC3
PtdIns(3)P
Phosphatidylinositol 3-phosphate (PI3P)
RB1CC1
RB1-inducible coiled-coil protein 1
TGN
Trans-Golgi network
TMZ
Temozolomide
TOR
Target of rapamycin
ULK1/2
Unc51-like kinase 1 and 2
UVRAG
UV radiation resistance-associated gene
VPS15
Vacuolar protein sorting 15
VPS34
Vacuolar protein sorting 34
WIPI1/2
WD-repeat protein interacting with phosphoinositides proteins 1 and 2

Introduction

Fifty years ago, Christian de Duve, a Belgian scientist, firstly coined the term autophagy at the Ciba Foundation symposium on lysosomes in 1963 [1, 2], for which he shared the Nobel Prize in Physiology or Medicine in 1974 with Albert Claude and George E. Palade. There are three morphologically and mechanistically distinct types of autophagy in cells: macroautophagy, microautophagy and chaperone mediated autophagy [3], and usually macroautophagy is referred to as autophagy [4].
Autophagy is an intracellular evolutionarily conserved catabolic degradation process in which cytoplasmic macromolecules, aggregated proteins, damaged organelles or pathogen are delivered to lysosomes, and digested by lysosomal hydrolases to generate nucleotides, amino acids, fatty acids, sugars, and ATP, and ultimately recycled into the cytosol [513] (Fig. 1). This cellular self-digestion mediated by lysosome sustains, on the one hand, cell metabolism and survival during starvation and stress, and eliminates, on the other hand, damaged proteins and organelles to maintain protein and organelle quality and quantity [14, 15].
Although autophagy was found over 50 years ago, only within decade lots of studies elucidated the functions and roles of this ubiquitous process. Recent studies have indicated that autophagy plays a greater variety of pathophysiological roles in many disease processes, including cancer, neurodegeneration, autoimmune diseases, aging, cell death, heart disease and infection, and aids cell to clear damaged proteins, organelles, pathogens or aggregates, and has been proposed as a cell death mechanism, programmed cell death type II [1621], whereas apoptosis is distinctively programmed cell death type I [2224]. The potential ability of autophagy to modulate cell death makes it a therapeutic target in cancer [25, 26].
With its basic role in the turnover of proteins and organelles, autophagy has multiple physiological and pathophysiological functions. During tumorigenesis, autophagy plays an important role. In this review, the molecular basis of autophagy and its roles in cancer are summarized.

Molecular basis of autophagy

Only a small amount of autophagy in cells is involved in maintaining homeostasis in physiological condition. When cells are stimulated by intracellular and extracellular factors e.g. starvation, hypoxia [27], some small molecular compounds [28], oxidation, and pathogen invasion [3, 29], a large number of autophagy is induced by the transduction of cellular signaling pathways, and many important autophagy-related proteins and their complex involved in the autophagic process [30].

Process of autophagy

Physiologically, autophagy is an evolutionarily conserved, self-degradative, normal physiological process in cells, which is composed of several closely related steps including induction of autophagy, assembly and formation of autophagosome, autophagosome docking and fusion with lysosomal membranes, and degradation and recirculation of intra-autophagosomal contents in autophagolyosome [17, 31] (Fig. 1a-d).

Induction of autophagy

Induction of autophagy can be triggered by several intracellular and extracellular stimulus, e.g. nutrient starvation including depletion of total amino acids and serum starvation that strongly induces a high level of autophagy [27], oxidative stress that induces autophagy in order to recycle damaged organelles (e.g. mitochondria) and eliminate proteins aggregates [32], and inhibitors of TOR such as rapamycin and CCI-779 [17]. Under nutrient-rich condition, the active mTORC1 kinase hyperphosphorylates ATG13 and blocks the interaction of ATG13 with ULK1and FIP200. When cells are induced by those intracellular and extracellular stimulating factors, the ATG13 anchors ULK1 to a pre-autophagosomal structure (PAS), and then the almost all autophagy-related (Atg) proteins gather hierarchically onto the PAS (Fig. 1a), which is reported to be a crucial site of the cytoplasm to vacuole targeting (Cvt) and autophagosome formation [2, 33, 34].
As a dock structure for recruitment of ATG proteins, PAS plays a critical role during induction of autophagy [34, 35] Under autophagy-inducing conditions, the functional unit ULK1/Atg1 (including ULK1, ATG13, FIP200, and ATG101) acts as autophagy initiation complex, in which the ATG13 is a crucial protein for the PAS localization of ULK1 (Atg1 in yeast) and the interaction of FIP200 with ULK1, while the FIP200 (Atg11 and Atg17 in yeast) functions as a scaffold for downstream ATG protein assembly at the PAS. Once the ATG13 and ULK1 target to the PAS, all of these multiple ATG proteins are initially engaged and localized to the PAS, that is the initiation of autophagy [2, 33, 34] (Fig. 1a). Subsequently, the other functional units, including ULK1 complex, PI3K complex, ATG9A system, ATG12-conjugation system, and LC3-conjugation system, are targeted to the PAS in a hierarchical manner and involved in assembly and formation of autophagosome [12, 3639].

Assembly and formation of autophagosome

Final formation of mature autophagosome includes nucleation of the multiple Atg proteins at PAS, elongation of the isolation membrane, and maturation of autophagosome, and four functional units are involved in these processes (Fig. 1b). The multiple Atg proteins gathering onto the PAS lead to the formation of a phagophore (or an isolation membrane) [40, 41]. The PAS is a potential nucleating site for forming the isolation membrane and recruits multiple Atg proteins. This nucleation process is initiated by the ULK1/Atg1 complex [42]. In response to nutrient starvation, the ULK1/Atg1 protein forms a complex with Atg13, FIP200/Atg17, Atg29, and Atg31, and this complex further associates with itself to generate the PAS scaffold complex, then the PI3K complex is gathered to the PAS and involved in forming phagophore through ATG14L interacting and binding to the ATG13 at PAS; and the ATG9A positive membrane vesicles associating with ATG2-WIPI complex (Atg2-Atg18 complex in yeast) are tethered to the PAS via interacting with the FIP200 (Atg17 and Atg11 in yeast). The multiple Atg proteins coordinate to generate the isolation membrane [42]. Once the first small ATG9A positive vesicles are fused at the PAS to form a phagophore, the bowl-shaped membrane is elongated continuously, and wraps and engulfs portions of cytoplasm and organelles. Finally, the isolation membrane, mediated by two ubiquitin-like ATG conjugation pathways, Atg12-Atg5 and Atg8/LC3 conjugation systems, forms a closed bilayer membrane structure, mature autophagosome with an inner and outer membrane [43] (Fig. 1b).

Autophagosome fusion with lysosomal membranes

Autophagosome docking and fusion with lysosomal membranes require the mature autophagosomes which will be transported to the perinuclear region for the autophagosome-lysosome fusion [44]. Autophagosomes can be formed randomly throughout the cytoplasm, whereas lysosomes are predominantly found in the perinuclear region. Therefore, once mature autophagosomes have been generated, they need to be delivered to the perinuclear region [45]. As long as autophagosomes arrive at the perinuclear region, they dock and fuse with lysosome immediately, and then form autophagolyosome (Fig. 1c).

Degradation and recirculation of autophagosomal contents

When autophagosome fuses with lysosomes to form autophagolyosome, many enzymes in lysosomes, e.g. lysosomal hydrolases, can degrade the inner membrane of the autophagosome and the cytoplasm-derived macromolecules, e.g. proteins and organelles, in the autophagosome into amino acids or peptides for reuse by cells (Fig. 1d).
Although autophagic structures by electron microscopy examination were firstly reported by Christian de Duve under 60 years ago, the molecular mechanism of autophagy regulation remained mostly unknown until discovery of yeast Atg genes in the 1990s, which greatly promoted the mechanistic understanding of autophagy and clarified the fact that autophagy plays important roles in various biological processes [4649]. Functionally, multiple autophagy-related proteins regulate and control various stages of the autophagy formation, including initiation of autophagy, nucleation of the multiple Atg proteins at PAS, elongation of the isolation membrane, and maturation of autophagosome, trafficking of mature autophagosomes, autophagosome docking and fusion with lysosomal membranes, and degradation of intra-autophagosomal contents in autophagolyosome by a hierarchical manner [17, 31].
So far, more than 40 genes encoding Atg proteins have been identified in yeast [49], and most of the genes (e.g. Atg1-Atg10, Atg12-Atg14, Atg16-Atg18) are conserved between yeast and mammalian, which indicates that autophagy is an evolutionarily conserved process [50]. Klionsky et al. (2003) collectively named the genes encoding these proteins as ATG (AuTophaGy), which is used to represent the autophagy gene and its encoding protein [50] (Table 1; Fig. 1).
Table 1
Autophagy-related (Atg) genes and their protein function in autophagy
Genes
Protein function description
References
Mammals
Yeast
ULK1/2
(Unc51-like kinase 1 and 2)
Atg1
Is part of the ULK-ATG13-ATG101-FIP200 complex and phosphorylates Beclin1; interacts with Atg13; is involved in initiation of autophagy, membrane targeting, membrane curvature sensing, and lipid vesicle tethering
[5154]
ATG2A/B
Atg2
Is part of the ATG9/ATG12-WIPI complex, which is important for ATG9 recruitment to expand autophagosome
[55, 56]
ATG3
Atg3
E2-like enzyme in LC3 lipidation; autocatalyzes itself to form ATG12-ATG3 complex for maintaining mitochondrial homeostasis
[5759]
ATG4A-D
Atg4
Cysteine protease to process Atg8 by removing its last amino acid; and deconjugate Atg8–PE; involved in LC3 activation and delipidation
[60, 61]
ATG5
Atg5
Is part of the ATG12-ATG5 complex involved in autophagosome formation/elongation, acting as an E3-like enzyme in LC3 lipidation; interacts with Atg16 and plays crucial roles in autophagy.
[62, 63]
Beclin1
Atg6
Is subunit of the VPS34-PI3K complex; recruits Atg14 or Vps38; interacts with Bcl-2; and lipid binding and membrane deformation
[64, 65]
ATG7
Atg7
E1-like enzyme interacting with E2 enzyme Atg10 or Atg3 involved in LC3 and ATG12 conjugation; and forms a thioester bond with Atg8
[6668]
MAP 1 LC3A-C, GABARAPs, GATE-16
Atg8
Modifier; Ubiquitin-like module conjugated to PE and used as autophagosome marker; recognizes the cargo-specific adaptors; and in vitro membrane tethering
[6971]
ATG9L1/L2
Atg9
Transmembrane protein; interacts with ATG2-WIPI complex; shuttles between PAS and peripheral organelles to deliver lipids/factors during phagophore expansion; and self-interaction
[72, 73]
ATG10
Atg10
E2-like enzyme in ATG12 conjugation with Atg5
[7476]
ATG12
Atg12
Modifier; ubiquitin-like module conjugated to Atg5; forms an E3 complex with Atg5 and Atg16; and interacts with Atg3
[59, 62, 77]
ATG13
Atg13
Is part of the ULK-ATG13-ATG101-FIP200 complex involved in initiation of autophagy; targets mTOR signaling pathway; interact with Atg1 and bridges Atg1 and Atg17-Atg31-Atg29; recruits the Vps34 complex via Atg14; binds to LC3; and interacts with Atg101
[7880]
ATG14L
(Barkor)
Atg14
Is subunit of VPS34-PI3K complex; interacts with Beclin1 to assemble the autophagic-specific complex; membrane targeting and membrane curvature sensing; and promote membrane fusion
[8183]
ATG16L1/L2
Atg16
Binds to ATG5-ATG12 complex acting as part of the E3 enzyme complex
[8486]
RB1CC1/ FIP200
Atg17
Is part of the ULK-ATG13-ATG101-FIP200 complex involved in initiation of autophagy; interacts with Atg13 and Atg9; forms ternary complex with Atg31 and Atg29; and senses membrane curvature
[53, 78, 87]
WIPI1–4
Atg18
Is part of the ATG2-WIPI complex which is important for ATG9 recruitment to autophagosome; binds to PI3P; required for the retrograde transport of Atg9; and complexes with Atg2
[88, 89]
ATG101
Interact with Atg13 and forms the ULK-ATG13-ATG101-FIP200 complex
[90, 91]
-, This protein has not been identified

Regulation and signaling of autophagy

In mammal cells, the starvation-induced autophagy is regulated by about 20 core ATG proteins, which can be classified into several functional units: (1) the ULK kinase core complex including ULK1/2, ATG13, RB1CC1/FIP200, and ATG101, (2) the autophagy-specific class III phosphatidylinositol 3-kinase (PI3K) complex including VPS34, VPS15, Beclin1, and ATG14L, (3) the ATG9A trafficking system including ATG9A, WIPI1/2, and ATG2A, (4) the ATG12 ubiquitin-like conjugation system including ATG12, ATG7, ATG10, ATG5, and ATG16L1, and (5) the LC3 ubiquitin-like conjugation system including LC3A/B/C, ATG7, ATG3, and ATG4A/B/C/D. These ATG proteins are recruited hierarchically proximal to the vacuole and organize the pre-autophagosomal structure (PAS) that is essential for autophagosome formation [12, 3639] (Fig. 1e-i; Table 2).
Table 2
ATG proteins of mammals in the core machinery of autophagosome formation
Complex
Components
Roles of the proteins in the core machinery
The ULK kinase core complex
ULK1/2
Protein kinase and recruitment of ATG proteins to the PAS
ATG13
ULK-binding protein and linker between ULK1/2 and FIP200
RB1CC1/FIP200
Scaffold protein for ULK1/2 and ATG13
ATG101
ATG13-binding protein
The class III PI3K complex I
VPS34
PtdIns 3-kinase catalytic subunit
VPS15
Serine/Threonine protein kinase
Beclin1
Component of PtdIns3K complex I and II
ATG14L
Component of PtdIns3K complex I
The ATG9A/ATG2-WIPI1/2 trafficking system
ATG9A
Transmembrane protein required for autophagosome formation
WIPI1/2
PtdIns3P-binding protein
ATG2A
Interacts with WIPI1/2
The ATG12-conjugation system
ATG12
Ubiquitin-like protein conjugated to ATG5
ATG7
E1-like enzyme
ATG10
E2-like enzyme
ATG5
Conjugated by ATG12
ATG16L1
Interacts with ATG12 and ATG5
The LC3-conjugation system
LC3A-C, GABARAPs, GATE-16
Ubiquitin-like protein conjugated to PE
ATG7
E1-like enzyme
ATG3
E2-like enzyme
ATG4A-D
LC3 carboxy-terminal protease, and deconjugating

ULK/Atg1 kinase core complex

During autophagy, autophagosome biogenesis commences at the PAS. In yeast, the Atg1 kinase core complex, consisting of the subunits Atg1, Atg13, Atg17, Atg29, and Atg31, is thought to play an essential and crucial role in the initiation of autophagy at the PAS, and has similar function to the ULK kinase core complex in mammal cells [92]. The ULK/Atg1 complex is mainly involved in receiving signals of cellular stimulation, recruiting ATG/Atg proteins to the PAS, organizing the vesicle cluster to form the phagophore, and governing elongation of the phagophore and formation of autophagosome [54, 93].
In human cells, the ULK1/2 is thought to serve similar and conserved functions as the yeast Atg1 [54]. In yeast, the core subunits of the Atg1 kinase complex are Atg1 and Atg13 [94]. When the cell is stimulated by starvation or other external stress, the target of rapamycin kinase complex (TOR) is inactivated, and then the Atg13 is dephosphorylated and binds greatly to Atg1 to form an activated Atg1-Atg13 dimer [95, 96]. The Atg17, Atg29, and Atg31 can assembly and form a trimeric complex Atg17-Atg31-Atg29 at the PAS [2, 97], and then serve as a preexisting scaffold for the recruitment of Atg1-Atg13 upon activation [98]. In mammal, the ULK1/2, a homologous protein of the kinase Atg1, forms a ULK1/2 kinase complex with ATG13 (homologous to Atg13 in yeast), FIP200 (homologous to Atg17 in yeast) and ATG101 (no homolog in yeast) [99] (Fig. 1e; Table 2).
Atg17 (FIP200) is the earliest protein to arrive at the PAS and adapts a highly elongated crescent shape [37, 93, 100]. The Atg17 is required specifically and associates physically with Atg1-Atg13 (ULK1/2-ATG13) complex, and the interaction between Atg17 and Atg1 is mediated by Atg13, indicating that Atge13 directly binds to both Atg1 and Atg17 and the Atg17-Atg13 complex formation plays an important role in normal autophagosome formation via binding to and activating the Atg1 kinase [33, 93, 96, 101, 102].

The class III PI3K complex I

In mammalian cells, the class III PI3K complex has two distinct types: complex I (PI3KC3-CI) and complex II (PI3KC3-CII). The both complexes share three core subunits: VPS34 (Vps34 in yeast), VPS15 (Vps15 in yeast), and Beclin1 (Atg6/Vps30 in yeast), and each complex contains a unique component: ATG14L/Barkor (Atg14 in yeast) of the autophagy-related complex I and Vps38 of the Vps-related complex II, which determines the localization of its own complex in the cell (Fig. 1f). The ATG14L (Atg14) can associate with and anchor the PI3KC3-CI to the PAS [103106] and the Vps38 can localize the PI3KC3-CII to vacuolar and endosomal membranes [103, 107].
In the autophagy-specific PI3KC3-CI, VPS34, a catalytic PI(3) kinase, catalyzes phosphatidylinositol (PI) phosphorylation to form phosphatidylinositol 3-phosphate (PtdIns(3) P or PI3P). The PtdIns(3) P on autophagic membranes is essential for the elongation and completion of autophagosomes for it can bind and recruit the membrane-bound protein ATG18 to the bilayer membrane [108, 109].
In yeast, Atg6 mediates interaction with Atg14 that is crucial for localizing the PI3KC3-CI to PAS [110, 111]. The sole Class III PI3K, Vps34, is associated with the protein kinase Vps15, which functions as a Vps34 regulatory subunit [112]. Recently, it is reported that the interaction of Vps15-Vps34 with Atg14-Atg6 is mediated by Atg38, which was shown to play a crucial role in the complex integrity [113]. In mammalian cells, Beclin1 is a central regulator, which interacts with a multitude of proteins including ATG14L, UVRAG, Rubicon, and Bcl-2, etc. [114117]. The Beclin1 has three functional domains including a N-terminal Bcl-2 homology 3 (BH3) domain, interacting with the Bcl-2 family protein Bcl-XL [118120], a central coiled-coil domain (CCD), mediating interaction of Beclin1 with ATG14L and UVRAG [121], and a C-terminal evolutionarily conserved domain (ECD), mediating the interaction of Beclin1 with VPS34 and activation of VPS34 kinase activity to regulate the size and number of autophagosomes [110, 111, 116, 122] (Fig. 1f; Table 2).

The ATG9A/Atg9 trafficking system

After the ULK/Atg1 complex is formed, the next step is recruitment of ATG9A/Atg9-containing cytoplasmic vesicles (ATG9A/Atg9 vesicles), which is a crucial step of autophagosome formation and plays an essential role in the nucleation step of autophagosome formation in eukaryotes (from yeast to mammals) [73, 123, 124]. The ATG9A/Atg9 can be phosphorylated by ULK/Atg1, and then the phosphorylated ATG9A/Atg9 is required for the recruitment of LC3/Atg8 and WIPI1/2/Atg18 to the site of autophagosome formation and the expansion and elongation of phagophore [125].
Mammalian ATG9A, the yeast Atg9 homolog, is the sole multi-spanning transmembrane protein within the core machinery of autophagosome formation and has 6 highly conserved transmembrane helices and 2 cytosolic NH2- and COOH-terminal domains that are involved in interactions with other ATG components in both yeast and mammals [126128].
The ATG9A/Atg9 self-interacts and self-associates within membranes into a higher-order assembly [129]. Recent studies indicated that the majority of Atg9 in the yeast are incorporated on small cytoplasmic vesicles with diameters of 30–60 nm, namely Atg9 vesicles [123, 130]. It is estimated that 3 Atg9 vesicles contain approximately 30 molecules of Atg9 each assemble at the PAS [123, 131], and subsequently the Atg9 is integrated into the outer autophagosomal membrane. Once the autophagosomes fuse with vacuole, the Atg9 are recycled as new Atg9 vesicles [102, 123, 132]. The level of Atg9 expressed in cells correlates with the frequency of autophagosome formation and the number of autophagosome [133]. The Atg9 vesicles are originated and transported from the Golgi apparatus [73, 102, 123, 127]. In normal physiological conditions, Atg9 localizes to and cycles between the trans-Golgi network (TGN) and early and late post-Golgi endosomes [134, 135]. A recent study suggests that autophagosome formation occurs where ATG9 vesicles coalesce with the ER [136].
In yeast, Atg9 recycling from PAS is regulated by the Atg2-Atg18 complex [37, 102]. The Atg18 and Atg2 are peripheral membrane proteins. The Atg18 as a downstream effector of class III PI (3) K localizes to PAS via binding to PtdIns(3) P [137]. Recent reports showed that the Atg18-Atg2 complex may play an important role in transporting the membrane structures during autophagosome formation through binding to Atg9 and form an Atg9·Atg2-Atg18 complex on the surface of the PAS and further regulate cycling of Atg9 [102, 138, 139].
In mammals, the WIPI (WD-repeat protein interacting with phosphoinositides) proteins, including WIPI1, WIPI2, WIPI3, and WIPI4, have a similar function to the Atg18 [88, 140, 141]. The WIPI1/2-ATG2 complex is involved in forming ATG9A·WIPI1/2-ATG2 trafficking system, mediating and regulating cycling of ATG9A, and promoting formation of LC3-positive autophagosomes in autophagy [140]. The WIPI1/2-ATG2 (Atg18-Atg2 in yeast) complex localizes to the expanding edge of the isolation membrane and plays a key role in the elongation and/or closure of the isolation membrane [43, 142] (Fig. 1g; Table 2).

The ATG12/Atg12-conjugation system

During autophagosome formation, two ubiquitin-like conjugation systems are required including the ATG12/Atg12 and LC3/Atg8 conjugation systems, and as many as eight ATG proteins are involved in both conjugation systems, which contribute to and are tightly associated with expansion of autophagosomal membrane.
In yeast, the Atg12, a ubiquitin-like protein, is covalently linked to its substrate Atg5 and forms an irreversible Atg12~Atg5 conjugate [38]. The Atg12-conjugation system is similar to the E1-E2-E3 activation and ligase present in the ubiquitination pathway, in which Atg12 is activated by Atg7, an E1-like enzyme [143], and then is transferred to Atg10, an E2-like enzyme [144], and is finally conjugated to its substrate protein Atg5 [38]. The Atg12~Atg5 conjugation has no typical E3 enzyme. The Atg5 of the Atg12~Atg5 conjugate further interacts with a small coiled-coil protein, Atg16, to form a ~ 350-kDa Atg12~Atg5-Atg16 complex [145, 146].
In mammals, the ATG12, activating by the E1 enzyme ATG7, is conjugated to ATG5 via the E2 enzyme Atg10 and then the ATG12-ATG5 conjugate can be stabilized by ATG16L proteins and further form ATG12-ATG5- ATG16L complex of approximately 800 kDa, which is important for the formation of the LC3 conjugation system [58, 62, 147] (Fig. 1h; Table 2).

The LC3/Atg8 -conjugation system

The LC3/Atg8-conjugation system is located downstream of the ATG12/Atg12 system in the context of Atg protein organization.
In yeast, the Atg8, another ubiquitin-like protein in yeast, is covalently linked to phosphatidylethanolamine (PE) after its C-terminal Arg117 residue is removed by a cysteine protease, Atg4, to expose to Gly116 [148]. The Atg8 is activated by the E1-like enzyme Atg7 [143], and then transferred to the E2-like enzyme Atg3 [149], and eventually the Atg3 conjugates Atg8 with the PE through an amide bond [39]. The Atg8-PE conjugate can be cleaved by Atg4 to release free Atg8, indicating that the Atg8-PE is reversible [148]. Most of Atg8 exist in the unconjugated form under normal conditions, but when autophagy is induced by starvation, most of Atg8 are activated, transferred, and converted to the PE-conjugated form [150].
In mammalian cells, there are several homologues of yeast Atg8 including LC3, GATE16, GABARAP and ATG8L. The LC3 has been best investigated and characterized as an autophagosome marker in mammalian cells [151155], which forms an Atg8-like conjugation system, called the LC3-conjugation system (Fig. 1i; Table 2).
LC3, microtubule-associated protein light chain 3, is a soluble protein with a molecular mass of approximately 17 kDa. The LC3 is firstly synthesized as a precursor protein (proLC3) [156], then the C-terminal peptide of the proLC3 precursor is cleaved by mammalian ATG4B homologues to form LC3-I with an exposed C-terminal glycine [151, 157, 158]. Catalyzed by mammalian ATG7 and ATG3 homologues, cytosolic LC3-I is then activated by the E1 enzyme ATG7 and transferred to the E2 enzyme ATG3, and finally is modified to a membrane-bound protein, LC3-II, by conjugating to the amino group of the lipid phosphatidylethanolamine (PE) (LC3-I/PE). Then, the Atg12-Atg5-Atg16 complex acts as an E3 enzyme for the conjugation reaction of LC3-II (LC3-I/PE) [154, 155, 157] (Fig. 1i), which corresponds to the Atg8-PE-conjugated form in yeast [151, 159]. The ATG4B has been reported that it is the sole enzyme to efficiently cleave LC3 precursors and LC3-I/PE among four human homologues of yeast Atg4 (Atg4A-D) [151]. Thus, the both ubiquitin-like systems are intimately involved in formation of PAS, assembly and formation of autophagosome, and subsequent biogenesis of autophagy.

Autophagy in cancer

Physiologically, autophagy, by eliminating damaged proteins and organelles during stress and aging, plays critical roles in regulating organismal development, cooperating with the adaptive immune system, sustaining energy homeostasis and maintaining protein and organelle quality control [11, 160164].
In diseases, such as neurodegenerative diseases [165, 166], infectious diseases [11, 167, 168], and metabolic diseases [14], dysfunctional autophagy leads to the accumulation of abnormal and damaged proteins and organelles and formation of intracellular aggregates, and then prevents the ability of autophagy to battle and eliminate infectious pathogens [11, 161, 167, 169].
In cancer, autophagy can play neutral, tumor-suppressive, or tumor-promoting roles in different contexts and stages of cancer development [25, 170173], which is determined by nutrient availability, microenvironment stress, pathogenic conditions, and the presence of an immune system.

Dual role of autophagy in cancer

In cancer development, autophagy plays a dual role depending on type, stage or genetic context of the cancers [174179]. On the one hand, via its protein and organelle quality control function, autophagy can maintain genome stability, prevent chronic tissue damage, cell injury, and inflammation, and inhibit accumulation of oncogenic p62 protein aggregates, and then prevent tumor initiation, proliferation, invasion, and metastasis, thereby function as a tumor suppressive mechanism, especially in the early stage of tumorigenesis [180182]. Autophagy is important for the quality control of the cells such as removing damaged mitochondria, and its defective proteins (e.g., heterozygous knockdown Beclin1 and Atg7 in mice) promote the malignant transformation and spontaneous tumors [183185]; on the other hand, once the tumors progress to late stage, autophagy can function as a cellular protective, survival, and defense mechanism, maintain functional mitochondria, reduce DNA damage, and enhance the survival and resistance of the cancer cells against stress (e.g., nutrient deprivation, hypoxia, DNA damage metabolic stress, and chemotherapy), and then sustain tumor metabolism, growth, and survival and then mediate tumor promotion and development, finally promotes tumorigenesis and causes resistance to therapeutic agents [180, 182, 186]. It is reported that autophagy can contribute to the aggressiveness of the cancers by facilitating metastasis [187189] (Fig. 2). The effect of autophagy on cancers is dependent on multiple factors including tumor microenvironment, cancer type and stage, and genetic background.

Autophagy suppresses tumorigenesis

Autophagy, the lysosome-mediated cellular self-digestion, acts as a cellular quality-control mechanism to sustain cell metabolism and its protein and organelle quality control during starvation, eliminates damaged proteins and organelles that accumulate during stress, and suppress chronic tissue damage, then prevent tumor initiation, especially in the early stage of tumorigenesis [11, 180]. Several indirect evidences indicate that autophagy acts as a tumor suppressor (Fig. 2).

Defective autophagy contributes to tumorigenesis

Through the identification of Beclin1, an essential autophagy gene, autophagy is first linked to human cancer. The Beclin1, as a haploid-insufficient tumor suppressor, is mono-allelically deleted in human hepatocellular carcinoma (HCC), breast, ovarian, and prostate cancers [114, 115, 190, 191] and in mice tumor prone [192]. It is reported that the expression of Beclin1 in cancer tissues was down-regulated in 44 patients with hepatocellular carcinoma, and it was concluded that autophagy might inhibit tumorigenesis [190]. The spontaneous frequency of malignancies is higher in the Beclin1+/− mouse model [192, 193], indicating that autophagy is a tumor-suppression mechanism [11, 25, 190, 194].
A number of studies on the ATG genes relevance to human cancers showed that other ATG genes are also oncogenically associated, including ATG2B, ATG5, ATG9B, ATG12 and ATG16L1. The frameshift mutations with mononucleotide repeats have been found in ATG2B, ATG5, ATG9B and ATG12 genes in gastric cancer and colorectal cancer, which may be involved in cancer development by deregulating the autophagy process [195]. The homozygote deletion of ATG5 predisposed to liver tumors with high penetrance mouse model [196]; the somatic point mutations of ATG5 are also identified in 135 patient samples of gastric cancer, colorectal cancer, and hepatocellular carcinoma [197]. The compartment-specific expression of ATG16L1 in epithelial cancer cells inhibited tumor growth [198].
Taken together, whether the expression of the intact autophagy genes is downregulated in cancers or the spontaneous frequency of cancer malignancies is increased due to autophagy-related gene deficiency, indicating that the intact autophagy functions as a cancer suppression mechanism by limiting genome damage and mutation and constraining tumor initiation.

Autophagy inhibits necrosis and inflammation

Autophagy is a central regulator of the inflammasome, and the chronic inflammation is a common future of early cancer development [199202]. The oncogene activation can cause neoplasia and inflammation, and the inflammatory conditions can increase cancer risk. The autophagy-deficient tumors display an increased level of necrosis and inflammation, indicating that the intact autophagy can inhibit neoplasia, inflammation and cancer [203, 204]. Defective autophagy lead to tissue damage, necrosis, chronic inflammation, and genetic instability, which can increase the incidence of cancer by altering the tumor microenvironment, elevating oxidative stress and creating cancer-causing mutations [204, 205]. In autophagy-defective cells and tissues, the failure to eliminate damaged proteins and organelles leads cellular dysfunction and death, and then stimulates an inflammation condition, and creates ultimately a cancer-prone environment [206].
Among mammal ATG proteins, Beclin-1, ATG5, ATG7, ATG12, ATG16L1 and LC3B are the most studied with respect to inflammation [207], and defects in autophagy are linked to many inflammatory diseases [208, 209] and cancer [210].

Accumulation of p62/SQSTM1 promotes tumorigenesis

The p62, also called sequestosome 1 (SQSTM1) in humans, a multifunctional adaptor protein, is a selective substrate of autophagy. In intact autophagy, the p62/SQSTM1 possesses a short LC3 interaction region (LIR) that facilitates direct interaction with LC3 and causes p62 to be specifically degraded by autophagy, while defective autophagy is a mechanism for p62 upregulation commonly observed in human tumors, so the level of p62 has been used as a marker for inhibition of autophagy or defects in autophagic degradation [211213].
The aberrant accumulation of p62 has been detected in the cases of gastrointestinal cancer [214], prostate cancer [215, 216], hepatocellular carcinoma [217219], breast cancer [220, 221], lung adenocarcinoma [222], suggesting that p62 accumulation correlates with cancer progression and autophagy suppresses tumorigenesis by limiting p62 accumulation [213, 223, 224].

Autophagy mediates cancer promotion

Once the tumors progress to late stage, autophagy can promote the survival and growth of the established tumors by removing toxic oxygen radicals or damaged proteins, maintaining mitochondrial function, sustaining metabolism and survival in stress, and preventing diversion of tumor progression to benign oncocytomas [180182]. Many investigations have shown that autophagy is the major contributor for cancer cells substantially survival [225227]. It is reported that autophagy can contribute to the aggressiveness of the cancers by facilitating metastasis [187189]. Moreover, autophagy as a cellular defense mechanism may reduce the effect of treatments of most chemotherapeutic agents (Fig. 2).

Autophagy prevents cancer cell damage

Autophagy is robustly activated in cancer cells under a multitude of stress conditions, including starvation, growth factor deprivation, hypoxia, damaging stimuli and proteasome inhibition, so elevated levels of autophagy have been observed in many tumor types, e.g. the essential autophagy gene Beclin1 was upregulated in colorectal cancer, gastric cancer, liver cancer, breast cancer, and cervical cancer [228231], suggesting that the enhancement of autophagy can promote tumorigenesis and overexpression of the Beclin1 plays a crucial role in tumor formation.
Autophagy functioning as a cancer promotion mechanism is mainly based on its role involved in removing damaged mitochondria, inhibiting DNA damage, maintaining genome stability, limiting inflammation, and finally preventing cancer cell damage under the conditions of stress [14, 213, 232]. Normal mitochondrial function, e.g. mitochondrial respiration, is required for tumorigenesis [233], t the accumulation of morphologically abnormal mitochondria and mitochondrial dysfunction have been found in the autophagy-defective tumors [196, 234236], indicating that intact autophagy can remove damaged mitochondria and contribute to tumorigenesis. The activation of the DNA damage response, gene amplification, DNA copy number variations and an elevated mutation rate has been found in the autophagy-deficient cancer cells [237]. Autophagy prevents genome damage and promotes tumor cell survival in a model of mammary cancer [194]. Autophagy is induced in hypoxic tumor regions and is required for tumor cell survival and for limiting inflammation [226]. (preventing cancer cell damage). All of these evidences indicate that the survival function of autophagy can be commandeered by tumors to prevent cell damage and promote tumorigenesis under conditions of metabolic stress.

Autophagy promotes cancer metastasis

During cancer progression, metastasis is an extremely complex process that indicates a more advanced stage and a poorer prognosis and accounts for most cancer-related deaths [238]. The metastasis of primary tumor can be divided into a series of stages including invasion of tumor cells from the primary tumor site, intravasation and survival in blood circulation systems, dissemination of the malignant cancer cells through the circulation systems to reach a capillary bed and adhere to the vessel walls, extravasation of the cancer cells at a distant site, and finally colonization of disseminated tumor cells at their destination organs [239242].
Autophagy plays a complex and stage-specific role and promotes multiple steps during cancer metastasis [243]. During the early stage of metastasis, the autophagy may act as a suppressor of metastasis by preventing tumor necrosis and restricting inflammatory cell infiltration [243]; on the other hand, in the advanced stages of metastasis, the autophagy may act as a promoter of metastasis by promoting dissemination of the malignant cancer cells in the circulation [244], enhancing colonization of detached metastatic cell in the destination organs [245], and inducing metastatic cells to enter dormancy and survive in the new environment [246].
Autophagy is upregulated during cancer metastasis. Once the metastatic cancer cells successfully establish distant colonies in their destination organs, autophagy begins to play a critical role and the autophagic flux is induced to respond various environmental stress including hypoxia, nutrient deprivation and detachment from the ECM [187, 240, 247, 248]. Using the autophagy marker, LC3B, various studies have identified an association between increased autophagy and metastasis in several types of cancer including breast cancer metastasis [249, 250], melanoma metastases [251], hepatocellular carcinoma [189], and glioblastoma [252]. These evidences indicate that autophagy promotes cancer metastasis and enhances the aggressiveness of cancer cells [253].

Autophagy inhibit cancer therapy

Cancer cells have common characteristics including increased metabolic demands, high level cellular proliferation, evading growth suppressors, resisting cell death, enabling replicative immortality, inducing angiogenesis, activated invasion and metastasis, and enhanced cellular stress, which require autophagy to be activated to maintain energy, enhance stress tolerance, limit damage, and prevent death in these cells.
Autophagy plays a cytoprotective or pro-survival role in cancer cells and can be induced by most cancer treatments including radiation therapy [254256], chemotherapy [257, 258], histone deacetylase inhibitors in colon cancer cells [259], arsenic trioxide (As2O3) in malignant glioma cells [260, 261], Temozolomide (TMZ) in malignant glioma cells [262], γ-irradiation in breast cancer, prostate cancer, colon cancer and malignant glioma [263265], resveratrol in ovarian cancer [266], TNFα in breast cancer cells [267], IFNγ in Hepatocellular carcinoma (HCC) [268], imatinib lung carcinoma cell [269], rapamycin in malignant glioma cells [270], and tamoxifen in breast cancer and Glioblastoma [271, 272], and the autophagy, in turn, functions as a cellular defense and protection mechanism to prevent cancer cell death upon treatment, enable a state of dormancy in residual cancer cells post treatment, contribute to cancer recurrence and metastasis, and inhibit cancer therapy and tumor cell killing [246, 273].
Given the pro-survival role, the inhibition of the autophagy has been shown to enhance and increase the efficacy of anticancer therapy, implying that autophagy inhibition is a potential valuable approach in combination with other anticancer therapeutic approaches to enhance cancer treatment [181, 182].

Conclusions and perspectives

Autophagy, as a cell survival pathway, plays an important role in cancer, and can help to prevent bioenergetic failure by metabolic stress and maintain protein and organelle quality and quantity, and contributes to all aspects of tumorigenesis, including tumor initiation, progression and development, and maintenance of the malignant state. Cancer cells divide relentlessly, and they are also metabolically stressed. As cancer cells grow, spread, and form solid tumors or flood the blood with abnormal cells, they always face an acute problem increasing hypoxia and nutrient deprivation, which may promote their death and prevent their growth, progression, and development, and autophagy is essential for surviving these stresses and maintaining tumorigenesis. Autophagy also plays key roles in controlling the tumor microenvironment, in suppressing tumor during the early stage and promoting cancer during the late stage, and in the therapeutic response.
Autophagy has a dual role both in progression and inhibition of cancer. Hitherto many data support a dynamic role of autophagy in cancer, both as a tumor suppressor early in progression and as a cancer promotor later in tumor maintenance and therapeutic resistance. In the early tumorigenesis, autophagy, as a survival pathway and quality-control mechanism, contributes to normal cell physiology metabolism and provides biological materials and energy in response to stress, and as a dynamic degradation and quality-control mechanism, eliminates damaged proteins and organelles and prevents tumor initiation. Once the tumors progress to late stage and are established and subjected to the environmental stresses including limited angiogenesis, nutrient deprivation, and hypoxia, autophagy, as a dynamic degradation and recycling system, contributes to the survival and growth of the established tumors and promotes aggressiveness of the cancers by facilitating metastasis.
Regulation of autophagy can be used as effective interventional strategies for cancer prevention and therapy by preventing cancer development, limiting tumor progression, and increasing the efficiency of cancer treatment. On the one hand, autophagy, as one type of programmed cell death, is ubiquitous in various cancer, functions as a tumor suppressor pathway, facilitates the degradation of oncogenic molecules, and finally prevents development of cancers. So defective or inadequate levels of autophagy can lead to cancer. Investigations showed that all chemotherapeutic agents and radiotherapies induce cancer metabolic stress and concomitant inhibition of autophagy, indicating that the autophagy regulation represents a significant direction in the development of anticancer therapies. On the other hand, autophagy, the type II programmed cell death, is involved in several signaling pathways during tumorigenesis via coordinating with apoptosis, the type I programmed cell death. Under stress conditions such as hypoxic or low-nutrition environments autophagy facilitates the survival of tumor cells, and at same time, apoptosis prevents the survival of cancer cells, indicating that autophagy and apoptosis, as two catabolic pathways, are essential for organismal homeostasis and tumor microenvironment. Investigations have now shown that autophagy and apoptosis are interconnected and coordinated by several molecular nodes of crosstalk, such as interaction of Beclin1 with Bcl-2, UVRAG with Bif-1, and ATG12 with the Mcl-1, etc.
So far, some standard cancer treatments have saved, or at least prolonged, many lives. However, the most severe clinical issue is the frequent tumors progression and cancer recurrence after treatment, mainly due to therapeutic resistance. It can be sure that autophagy can facilitate the tumor cells survival and deal with anticancer therapy. Therefore, in the near future, standard cancer treatment combining with regulation of autophagy activity, promoting or preventing by autophagy inducers or inhibitors based on tumorigenesis and cancer stages, can be considered as a potential anticancer therapy. However, further investigations should be done to understand and clarify how autophagy contributes to the development and treatment of cancer, how the autophagy pathway can be targeted and regulated, and how the activity of autophagy pathway can be monitored and quantified during cancer prevention and therapy.

Acknowledgements

Not applicable.
Not applicable.
Not applicable.

Competing interests

The authors declare that they have no competing interests.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Literatur
2.
Zurück zum Zitat Kawamata T, et al. Organization of the pre-autophagosomal structure responsible for autophagosome formation. Mol Biol Cell. 2008;19(5):2039–50.PubMedPubMedCentralCrossRef Kawamata T, et al. Organization of the pre-autophagosomal structure responsible for autophagosome formation. Mol Biol Cell. 2008;19(5):2039–50.PubMedPubMedCentralCrossRef
3.
Zurück zum Zitat Xie Z, Klionsky DJ. Autophagosome formation: core machinery and adaptations. Nat Cell Biol. 2007;9(10):1102–9.PubMedCrossRef Xie Z, Klionsky DJ. Autophagosome formation: core machinery and adaptations. Nat Cell Biol. 2007;9(10):1102–9.PubMedCrossRef
5.
Zurück zum Zitat Levine B, Klionsky DJ. Development by self-digestion: molecular mechanisms and biological functions of autophagy. Dev Cell. 2004;6(4):463–77.CrossRefPubMed Levine B, Klionsky DJ. Development by self-digestion: molecular mechanisms and biological functions of autophagy. Dev Cell. 2004;6(4):463–77.CrossRefPubMed
6.
8.
Zurück zum Zitat Klionsky DJ. Autophagy: from phenomenology to molecular understanding in less than a decade. Nat Rev Mol Cell Biol. 2007;8(11):931–7.PubMedCrossRef Klionsky DJ. Autophagy: from phenomenology to molecular understanding in less than a decade. Nat Rev Mol Cell Biol. 2007;8(11):931–7.PubMedCrossRef
10.
12.
Zurück zum Zitat Mizushima N, Yoshimori T, Ohsumi Y. The role of Atg proteins in autophagosome formation. Annu Rev Cell Dev Biol. 2011;27:107–32.PubMedCrossRef Mizushima N, Yoshimori T, Ohsumi Y. The role of Atg proteins in autophagosome formation. Annu Rev Cell Dev Biol. 2011;27:107–32.PubMedCrossRef
13.
Zurück zum Zitat Rogov V, et al. Interactions between autophagy receptors and ubiquitin-like proteins form the molecular basis for selective autophagy. Mol Cell. 2014;53(2):167–78.PubMedCrossRef Rogov V, et al. Interactions between autophagy receptors and ubiquitin-like proteins form the molecular basis for selective autophagy. Mol Cell. 2014;53(2):167–78.PubMedCrossRef
15.
Zurück zum Zitat Mizushima N, Komatsu M. Autophagy: renovation of cells and tissues. Cell. 2011;147(4):728–41.PubMedCrossRef Mizushima N, Komatsu M. Autophagy: renovation of cells and tissues. Cell. 2011;147(4):728–41.PubMedCrossRef
16.
Zurück zum Zitat Mizushima N. The pleiotropic role of autophagy: from protein metabolism to bactericide. Cell Death Differ. 2005;12(Suppl 2):1535–41.PubMedCrossRef Mizushima N. The pleiotropic role of autophagy: from protein metabolism to bactericide. Cell Death Differ. 2005;12(Suppl 2):1535–41.PubMedCrossRef
18.
Zurück zum Zitat Kundu M, Thompson CB. Autophagy: basic principles and relevance to disease. Annu Rev Pathol. 2008;3:427–55.PubMedCrossRef Kundu M, Thompson CB. Autophagy: basic principles and relevance to disease. Annu Rev Pathol. 2008;3:427–55.PubMedCrossRef
21.
Zurück zum Zitat Edinger AL, Thompson CB. Death by design: apoptosis, necrosis and autophagy. Curr Opin Cell Biol. 2004;16(6):663–9.CrossRefPubMed Edinger AL, Thompson CB. Death by design: apoptosis, necrosis and autophagy. Curr Opin Cell Biol. 2004;16(6):663–9.CrossRefPubMed
27.
Zurück zum Zitat Mazure NM, Pouyssegur J. Hypoxia-induced autophagy: cell death or cell survival? Curr Opin Cell Biol. 2010;22(2):177–80.PubMedCrossRef Mazure NM, Pouyssegur J. Hypoxia-induced autophagy: cell death or cell survival? Curr Opin Cell Biol. 2010;22(2):177–80.PubMedCrossRef
28.
Zurück zum Zitat Tsvetkov AS, et al. A small-molecule scaffold induces autophagy in primary neurons and protects against toxicity in a Huntington disease model. Proc Natl Acad Sci U S A. 2010;107(39):16982–7.PubMedPubMedCentralCrossRef Tsvetkov AS, et al. A small-molecule scaffold induces autophagy in primary neurons and protects against toxicity in a Huntington disease model. Proc Natl Acad Sci U S A. 2010;107(39):16982–7.PubMedPubMedCentralCrossRef
29.
Zurück zum Zitat Zhong Y, et al. Distinct regulation of autophagic activity by Atg14L and Rubicon associated with Beclin 1-phosphatidylinositol-3-kinase complex. Nat Cell Biol. 2009;11(4):468–76.PubMedPubMedCentralCrossRef Zhong Y, et al. Distinct regulation of autophagic activity by Atg14L and Rubicon associated with Beclin 1-phosphatidylinositol-3-kinase complex. Nat Cell Biol. 2009;11(4):468–76.PubMedPubMedCentralCrossRef
32.
Zurück zum Zitat Ureshino RP, et al. Calcium signaling alterations, oxidative stress, and autophagy in aging. Antioxid Redox Signal. 2014;21(1):123–37.PubMedCrossRef Ureshino RP, et al. Calcium signaling alterations, oxidative stress, and autophagy in aging. Antioxid Redox Signal. 2014;21(1):123–37.PubMedCrossRef
33.
Zurück zum Zitat Yamamoto H, et al. The intrinsically disordered protein Atg13 mediates Supramolecular assembly of autophagy initiation complexes. Dev Cell. 2016;38(1):86–99.PubMedCrossRef Yamamoto H, et al. The intrinsically disordered protein Atg13 mediates Supramolecular assembly of autophagy initiation complexes. Dev Cell. 2016;38(1):86–99.PubMedCrossRef
34.
Zurück zum Zitat Suzuki K, et al. The pre-autophagosomal structure organized by concerted functions of APG genes is essential for autophagosome formation. EMBO J. 2001;20(21):5971–81.PubMedPubMedCentralCrossRef Suzuki K, et al. The pre-autophagosomal structure organized by concerted functions of APG genes is essential for autophagosome formation. EMBO J. 2001;20(21):5971–81.PubMedPubMedCentralCrossRef
35.
Zurück zum Zitat Kotani T, et al. The Atg2-Atg18 complex tethers pre-autophagosomal membranes to the endoplasmic reticulum for autophagosome formation. Proc Natl Acad Sci U S A. 2018;115(41):10363–8.PubMedPubMedCentralCrossRef Kotani T, et al. The Atg2-Atg18 complex tethers pre-autophagosomal membranes to the endoplasmic reticulum for autophagosome formation. Proc Natl Acad Sci U S A. 2018;115(41):10363–8.PubMedPubMedCentralCrossRef
36.
Zurück zum Zitat Lamb CA, Yoshimori T, Tooze SA. The autophagosome: origins unknown, biogenesis complex. Nat Rev Mol Cell Biol. 2013;14(12):759–74.PubMedCrossRef Lamb CA, Yoshimori T, Tooze SA. The autophagosome: origins unknown, biogenesis complex. Nat Rev Mol Cell Biol. 2013;14(12):759–74.PubMedCrossRef
37.
Zurück zum Zitat Suzuki K, et al. Hierarchy of Atg proteins in pre-autophagosomal structure organization. Genes Cells. 2007;12(2):209–18.PubMedCrossRef Suzuki K, et al. Hierarchy of Atg proteins in pre-autophagosomal structure organization. Genes Cells. 2007;12(2):209–18.PubMedCrossRef
38.
Zurück zum Zitat Mizushima N, et al. A protein conjugation system essential for autophagy. Nature. 1998;395(6700):395–8.PubMedCrossRef Mizushima N, et al. A protein conjugation system essential for autophagy. Nature. 1998;395(6700):395–8.PubMedCrossRef
39.
Zurück zum Zitat Ichimura Y, et al. A ubiquitin-like system mediates protein lipidation. Nature. 2000;408(6811):488–92.PubMedCrossRef Ichimura Y, et al. A ubiquitin-like system mediates protein lipidation. Nature. 2000;408(6811):488–92.PubMedCrossRef
40.
Zurück zum Zitat Tooze SA, Yoshimori T. The origin of the autophagosomal membrane. Nat Cell Biol. 2010;12(9):831–5.PubMedCrossRef Tooze SA, Yoshimori T. The origin of the autophagosomal membrane. Nat Cell Biol. 2010;12(9):831–5.PubMedCrossRef
41.
Zurück zum Zitat Militello RD, Colombo MI. A membrane is born: origin of the autophagosomal compartment. Curr Mol Med. 2011;11(3):197–203.PubMedCrossRef Militello RD, Colombo MI. A membrane is born: origin of the autophagosomal compartment. Curr Mol Med. 2011;11(3):197–203.PubMedCrossRef
42.
Zurück zum Zitat Cheong H, et al. The Atg1 kinase complex is involved in the regulation of protein recruitment to initiate sequestering vesicle formation for nonspecific autophagy in Saccharomyces cerevisiae. Mol Biol Cell. 2008;19(2):668–81.PubMedPubMedCentralCrossRef Cheong H, et al. The Atg1 kinase complex is involved in the regulation of protein recruitment to initiate sequestering vesicle formation for nonspecific autophagy in Saccharomyces cerevisiae. Mol Biol Cell. 2008;19(2):668–81.PubMedPubMedCentralCrossRef
43.
Zurück zum Zitat Suzuki K, et al. Fine mapping of autophagy-related proteins during autophagosome formation in Saccharomyces cerevisiae. J Cell Sci. 2013;126(Pt 11):2534–44.PubMedCrossRef Suzuki K, et al. Fine mapping of autophagy-related proteins during autophagosome formation in Saccharomyces cerevisiae. J Cell Sci. 2013;126(Pt 11):2534–44.PubMedCrossRef
44.
Zurück zum Zitat Wijdeven RH, et al. Cholesterol and ORP1L-mediated ER contact sites control autophagosome transport and fusion with the endocytic pathway. Nat Commun. 2016;7:11808.PubMedPubMedCentralCrossRef Wijdeven RH, et al. Cholesterol and ORP1L-mediated ER contact sites control autophagosome transport and fusion with the endocytic pathway. Nat Commun. 2016;7:11808.PubMedPubMedCentralCrossRef
45.
Zurück zum Zitat Kimura S, Noda T, Yoshimori T. Dynein-dependent movement of autophagosomes mediates efficient encounters with lysosomes. Cell Struct Funct. 2008;33(1):109–22.PubMedCrossRef Kimura S, Noda T, Yoshimori T. Dynein-dependent movement of autophagosomes mediates efficient encounters with lysosomes. Cell Struct Funct. 2008;33(1):109–22.PubMedCrossRef
46.
Zurück zum Zitat Matsuura A, et al. Apg1p, a novel protein kinase required for the autophagic process in Saccharomyces cerevisiae. Gene. 1997;192(2):245–50.PubMedCrossRef Matsuura A, et al. Apg1p, a novel protein kinase required for the autophagic process in Saccharomyces cerevisiae. Gene. 1997;192(2):245–50.PubMedCrossRef
47.
Zurück zum Zitat Clark SL Jr. Cellular differentiation in the kidneys of newborn mice studies with the electron microscope. J Biophys Biochem Cytol. 1957;3(3):349–62.PubMedPubMedCentralCrossRef Clark SL Jr. Cellular differentiation in the kidneys of newborn mice studies with the electron microscope. J Biophys Biochem Cytol. 1957;3(3):349–62.PubMedPubMedCentralCrossRef
49.
50.
Zurück zum Zitat Klionsky DJ, et al. A unified nomenclature for yeast autophagy-related genes. Dev Cell. 2003;5(4):539–45.PubMedCrossRef Klionsky DJ, et al. A unified nomenclature for yeast autophagy-related genes. Dev Cell. 2003;5(4):539–45.PubMedCrossRef
51.
Zurück zum Zitat Kamada Y, et al. Tor directly controls the Atg1 kinase complex to regulate autophagy. Mol Cell Biol. 2010;30(4):1049–58.PubMedCrossRef Kamada Y, et al. Tor directly controls the Atg1 kinase complex to regulate autophagy. Mol Cell Biol. 2010;30(4):1049–58.PubMedCrossRef
53.
Zurück zum Zitat Hara T, Mizushima N. Role of ULK-FIP200 complex in mammalian autophagy: FIP200, a counterpart of yeast Atg17? Autophagy. 2009;5(1):85–7.PubMedCrossRef Hara T, Mizushima N. Role of ULK-FIP200 complex in mammalian autophagy: FIP200, a counterpart of yeast Atg17? Autophagy. 2009;5(1):85–7.PubMedCrossRef
54.
Zurück zum Zitat Mizushima N. The role of the Atg1/ULK1 complex in autophagy regulation. Curr Opin Cell Biol. 2010;22(2):132–9.PubMedCrossRef Mizushima N. The role of the Atg1/ULK1 complex in autophagy regulation. Curr Opin Cell Biol. 2010;22(2):132–9.PubMedCrossRef
55.
Zurück zum Zitat Tang Z, et al. Atg2A/B deficiency switches cytoprotective autophagy to non-canonical caspase-8 activation and apoptosis. Cell Death Differ. 2017;24(12):2127–38.PubMedPubMedCentralCrossRef Tang Z, et al. Atg2A/B deficiency switches cytoprotective autophagy to non-canonical caspase-8 activation and apoptosis. Cell Death Differ. 2017;24(12):2127–38.PubMedPubMedCentralCrossRef
56.
Zurück zum Zitat Velikkakath AK, et al. Mammalian Atg2 proteins are essential for autophagosome formation and important for regulation of size and distribution of lipid droplets. Mol Biol Cell. 2012;23(5):896–909.PubMedPubMedCentralCrossRef Velikkakath AK, et al. Mammalian Atg2 proteins are essential for autophagosome formation and important for regulation of size and distribution of lipid droplets. Mol Biol Cell. 2012;23(5):896–909.PubMedPubMedCentralCrossRef
57.
Zurück zum Zitat Besteiro S, et al. Autophagy protein Atg3 is essential for maintaining mitochondrial integrity and for normal intracellular development of toxoplasma gondii tachyzoites. PLoS Pathog. 2011;7(12):e1002416.PubMedPubMedCentralCrossRef Besteiro S, et al. Autophagy protein Atg3 is essential for maintaining mitochondrial integrity and for normal intracellular development of toxoplasma gondii tachyzoites. PLoS Pathog. 2011;7(12):e1002416.PubMedPubMedCentralCrossRef
58.
Zurück zum Zitat Metlagel Z, et al. Structural basis of ATG3 recognition by the autophagic ubiquitin-like protein ATG12. Proc Natl Acad Sci U S A. 2013;110(47):18844–9.PubMedPubMedCentralCrossRef Metlagel Z, et al. Structural basis of ATG3 recognition by the autophagic ubiquitin-like protein ATG12. Proc Natl Acad Sci U S A. 2013;110(47):18844–9.PubMedPubMedCentralCrossRef
60.
Zurück zum Zitat Li M, et al. Kinetics comparisons of mammalian Atg4 homologues indicate selective preferences toward diverse Atg8 substrates. J Biol Chem. 2011;286(9):7327–38.PubMedCrossRef Li M, et al. Kinetics comparisons of mammalian Atg4 homologues indicate selective preferences toward diverse Atg8 substrates. J Biol Chem. 2011;286(9):7327–38.PubMedCrossRef
61.
Zurück zum Zitat Lang T, et al. Aut2p and Aut7p, two novel microtubule-associated proteins are essential for delivery of autophagic vesicles to the vacuole. EMBO J. 1998;17(13):3597–607.PubMedPubMedCentralCrossRef Lang T, et al. Aut2p and Aut7p, two novel microtubule-associated proteins are essential for delivery of autophagic vesicles to the vacuole. EMBO J. 1998;17(13):3597–607.PubMedPubMedCentralCrossRef
62.
Zurück zum Zitat Otomo C, et al. Structure of the human ATG12~ATG5 conjugate required for LC3 lipidation in autophagy. Nat Struct Mol Biol. 2013;20(1):59–66.PubMedCrossRef Otomo C, et al. Structure of the human ATG12~ATG5 conjugate required for LC3 lipidation in autophagy. Nat Struct Mol Biol. 2013;20(1):59–66.PubMedCrossRef
63.
Zurück zum Zitat Matsushita M, et al. Structure of Atg5.Atg16, a complex essential for autophagy. J Biol Chem. 2007;282(9):6763–72.PubMedCrossRef Matsushita M, et al. Structure of Atg5.Atg16, a complex essential for autophagy. J Biol Chem. 2007;282(9):6763–72.PubMedCrossRef
64.
Zurück zum Zitat Cao Y, Klionsky DJ. Physiological functions of Atg6/Beclin 1: a unique autophagy-related protein. Cell Res. 2007;17(10):839–49.PubMedCrossRef Cao Y, Klionsky DJ. Physiological functions of Atg6/Beclin 1: a unique autophagy-related protein. Cell Res. 2007;17(10):839–49.PubMedCrossRef
66.
Zurück zum Zitat Yuan W, Stromhaug PE, Dunn WA Jr. Glucose-induced autophagy of peroxisomes in Pichia pastoris requires a unique E1-like protein. Mol Biol Cell. 1999;10(5):1353–66.PubMedPubMedCentralCrossRef Yuan W, Stromhaug PE, Dunn WA Jr. Glucose-induced autophagy of peroxisomes in Pichia pastoris requires a unique E1-like protein. Mol Biol Cell. 1999;10(5):1353–66.PubMedPubMedCentralCrossRef
67.
Zurück zum Zitat Hong SB, et al. Insights into noncanonical E1 enzyme activation from the structure of autophagic E1 Atg7 with Atg8. Nat Struct Mol Biol. 2011;18(12):1323–30.PubMedCrossRef Hong SB, et al. Insights into noncanonical E1 enzyme activation from the structure of autophagic E1 Atg7 with Atg8. Nat Struct Mol Biol. 2011;18(12):1323–30.PubMedCrossRef
68.
Zurück zum Zitat Noda NN, et al. Structural basis of Atg8 activation by a homodimeric E1, Atg7. Mol Cell. 2011;44(3):462–75.PubMedCrossRef Noda NN, et al. Structural basis of Atg8 activation by a homodimeric E1, Atg7. Mol Cell. 2011;44(3):462–75.PubMedCrossRef
69.
Zurück zum Zitat He H, et al. Post-translational modifications of three members of the human MAP 1LC3 family and detection of a novel type of modification for MAP 1LC3B. J Biol Chem. 2003;278(31):29278–87.PubMedCrossRef He H, et al. Post-translational modifications of three members of the human MAP 1LC3 family and detection of a novel type of modification for MAP 1LC3B. J Biol Chem. 2003;278(31):29278–87.PubMedCrossRef
71.
Zurück zum Zitat Weidberg H, et al. LC3 and GATE-16/GABARAP subfamilies are both essential yet act differently in autophagosome biogenesis. EMBO J. 2010;29(11):1792–802.PubMedPubMedCentralCrossRef Weidberg H, et al. LC3 and GATE-16/GABARAP subfamilies are both essential yet act differently in autophagosome biogenesis. EMBO J. 2010;29(11):1792–802.PubMedPubMedCentralCrossRef
73.
74.
Zurück zum Zitat Zhao Q, et al. Dual roles of two isoforms of autophagy-related gene ATG10 in HCV-subgenomic replicon mediated autophagy flux and innate immunity. Sci Rep. 2017;7(1):11250.PubMedPubMedCentralCrossRef Zhao Q, et al. Dual roles of two isoforms of autophagy-related gene ATG10 in HCV-subgenomic replicon mediated autophagy flux and innate immunity. Sci Rep. 2017;7(1):11250.PubMedPubMedCentralCrossRef
75.
Zurück zum Zitat Hong SB, et al. Structure of the autophagic E2 enzyme Atg10. Acta Crystallogr D Biol Crystallogr. 2012;68(Pt 10):1409–17.PubMedCrossRef Hong SB, et al. Structure of the autophagic E2 enzyme Atg10. Acta Crystallogr D Biol Crystallogr. 2012;68(Pt 10):1409–17.PubMedCrossRef
76.
Zurück zum Zitat Yamaguchi M, et al. Structural insights into Atg10-mediated formation of the autophagy-essential Atg12-Atg5 conjugate. Structure. 2012;20(7):1244–54.PubMedCrossRef Yamaguchi M, et al. Structural insights into Atg10-mediated formation of the autophagy-essential Atg12-Atg5 conjugate. Structure. 2012;20(7):1244–54.PubMedCrossRef
77.
78.
Zurück zum Zitat Alers S, et al. Atg13 and FIP200 act independently of Ulk1 and Ulk2 in autophagy induction. Autophagy. 2011;7(12):1423–33.PubMedCrossRef Alers S, et al. Atg13 and FIP200 act independently of Ulk1 and Ulk2 in autophagy induction. Autophagy. 2011;7(12):1423–33.PubMedCrossRef
79.
80.
Zurück zum Zitat Hosokawa N, et al. Nutrient-dependent mTORC1 association with the ULK1-Atg13-FIP200 complex required for autophagy. Mol Biol Cell. 2009;20(7):1981–91.PubMedPubMedCentralCrossRef Hosokawa N, et al. Nutrient-dependent mTORC1 association with the ULK1-Atg13-FIP200 complex required for autophagy. Mol Biol Cell. 2009;20(7):1981–91.PubMedPubMedCentralCrossRef
81.
Zurück zum Zitat Kim HJ, et al. Beclin-1-interacting autophagy protein Atg14L targets the SNARE-associated protein Snapin to coordinate endocytic trafficking. J Cell Sci. 2012;125(Pt 20):4740–50.PubMedPubMedCentralCrossRef Kim HJ, et al. Beclin-1-interacting autophagy protein Atg14L targets the SNARE-associated protein Snapin to coordinate endocytic trafficking. J Cell Sci. 2012;125(Pt 20):4740–50.PubMedPubMedCentralCrossRef
83.
Zurück zum Zitat Matsunaga K, et al. Two Beclin 1-binding proteins, Atg14L and Rubicon, reciprocally regulate autophagy at different stages. Nat Cell Biol. 2009;11(4):385–96.PubMedCrossRef Matsunaga K, et al. Two Beclin 1-binding proteins, Atg14L and Rubicon, reciprocally regulate autophagy at different stages. Nat Cell Biol. 2009;11(4):385–96.PubMedCrossRef
84.
Zurück zum Zitat Fujita N, et al. The Atg16L complex specifies the site of LC3 lipidation for membrane biogenesis in autophagy. Mol Biol Cell. 2008;19(5):2092–100.PubMedPubMedCentralCrossRef Fujita N, et al. The Atg16L complex specifies the site of LC3 lipidation for membrane biogenesis in autophagy. Mol Biol Cell. 2008;19(5):2092–100.PubMedPubMedCentralCrossRef
85.
Zurück zum Zitat Hwang S, et al. Nondegradative role of Atg5-Atg12/ Atg16L1 autophagy protein complex in antiviral activity of interferon gamma. Cell Host Microbe. 2012;11(4):397–409.PubMedPubMedCentralCrossRef Hwang S, et al. Nondegradative role of Atg5-Atg12/ Atg16L1 autophagy protein complex in antiviral activity of interferon gamma. Cell Host Microbe. 2012;11(4):397–409.PubMedPubMedCentralCrossRef
86.
Zurück zum Zitat Romanov J, et al. Mechanism and functions of membrane binding by the Atg5-Atg12/Atg16 complex during autophagosome formation. EMBO J. 2012;31(22):4304–17.PubMedPubMedCentralCrossRef Romanov J, et al. Mechanism and functions of membrane binding by the Atg5-Atg12/Atg16 complex during autophagosome formation. EMBO J. 2012;31(22):4304–17.PubMedPubMedCentralCrossRef
87.
Zurück zum Zitat Hara T, et al. FIP200, a ULK-interacting protein, is required for autophagosome formation in mammalian cells. J Cell Biol. 2008;181(3):497–510.PubMedPubMedCentralCrossRef Hara T, et al. FIP200, a ULK-interacting protein, is required for autophagosome formation in mammalian cells. J Cell Biol. 2008;181(3):497–510.PubMedPubMedCentralCrossRef
88.
Zurück zum Zitat Proikas-Cezanne T, et al. WIPI proteins: essential PtdIns3P effectors at the nascent autophagosome. J Cell Sci. 2015;128(2):207–17.PubMedCrossRef Proikas-Cezanne T, et al. WIPI proteins: essential PtdIns3P effectors at the nascent autophagosome. J Cell Sci. 2015;128(2):207–17.PubMedCrossRef
90.
Zurück zum Zitat Hosokawa N, et al. Atg101, a novel mammalian autophagy protein interacting with Atg13. Autophagy. 2009;5(7):973–9.PubMedCrossRef Hosokawa N, et al. Atg101, a novel mammalian autophagy protein interacting with Atg13. Autophagy. 2009;5(7):973–9.PubMedCrossRef
91.
Zurück zum Zitat Suzuki H, et al. Structure of the Atg101-Atg13 complex reveals essential roles of Atg101 in autophagy initiation. Nat Struct Mol Biol. 2015;22(7):572–80.PubMedCrossRef Suzuki H, et al. Structure of the Atg101-Atg13 complex reveals essential roles of Atg101 in autophagy initiation. Nat Struct Mol Biol. 2015;22(7):572–80.PubMedCrossRef
92.
Zurück zum Zitat Rubinsztein DC, Shpilka T, Elazar Z. Mechanisms of autophagosome biogenesis. Curr Biol. 2012;22(1):R29–34.PubMedCrossRef Rubinsztein DC, Shpilka T, Elazar Z. Mechanisms of autophagosome biogenesis. Curr Biol. 2012;22(1):R29–34.PubMedCrossRef
93.
94.
97.
Zurück zum Zitat Kabeya Y, et al. Characterization of the Atg17-Atg29-Atg31 complex specifically required for starvation-induced autophagy in Saccharomyces cerevisiae. Biochem Biophys Res Commun. 2009;389(4):612–5.PubMedCrossRef Kabeya Y, et al. Characterization of the Atg17-Atg29-Atg31 complex specifically required for starvation-induced autophagy in Saccharomyces cerevisiae. Biochem Biophys Res Commun. 2009;389(4):612–5.PubMedCrossRef
99.
100.
Zurück zum Zitat Chew LH, et al. Structural characterization of the Saccharomyces cerevisiae autophagy regulatory complex Atg17-Atg31-Atg29. Autophagy. 2013;9(10):1467–74.PubMedCrossRef Chew LH, et al. Structural characterization of the Saccharomyces cerevisiae autophagy regulatory complex Atg17-Atg31-Atg29. Autophagy. 2013;9(10):1467–74.PubMedCrossRef
102.
Zurück zum Zitat Reggiori F, et al. The Atg1-Atg13 complex regulates Atg9 and Atg23 retrieval transport from the pre-autophagosomal structure. Dev Cell. 2004;6(1):79–90.PubMedCrossRef Reggiori F, et al. The Atg1-Atg13 complex regulates Atg9 and Atg23 retrieval transport from the pre-autophagosomal structure. Dev Cell. 2004;6(1):79–90.PubMedCrossRef
103.
Zurück zum Zitat Kihara A, et al. Two distinct Vps34 phosphatidylinositol 3-kinase complexes function in autophagy and carboxypeptidase Y sorting in Saccharomyces cerevisiae. J Cell Biol. 2001;152(3):519–30.PubMedPubMedCentralCrossRef Kihara A, et al. Two distinct Vps34 phosphatidylinositol 3-kinase complexes function in autophagy and carboxypeptidase Y sorting in Saccharomyces cerevisiae. J Cell Biol. 2001;152(3):519–30.PubMedPubMedCentralCrossRef
104.
Zurück zum Zitat Obara K, Sekito T, Ohsumi Y. Assortment of phosphatidylinositol 3-kinase complexes--Atg14p directs association of complex I to the pre-autophagosomal structure in Saccharomyces cerevisiae. Mol Biol Cell. 2006;17(4):1527–39.PubMedPubMedCentralCrossRef Obara K, Sekito T, Ohsumi Y. Assortment of phosphatidylinositol 3-kinase complexes--Atg14p directs association of complex I to the pre-autophagosomal structure in Saccharomyces cerevisiae. Mol Biol Cell. 2006;17(4):1527–39.PubMedPubMedCentralCrossRef
105.
Zurück zum Zitat Obara K, Ohsumi Y. Dynamics and function of PtdIns(3) P in autophagy. Autophagy. 2008;4(7):952–4.PubMedCrossRef Obara K, Ohsumi Y. Dynamics and function of PtdIns(3) P in autophagy. Autophagy. 2008;4(7):952–4.PubMedCrossRef
107.
Zurück zum Zitat Burda P, et al. Retromer function in endosome-to-Golgi retrograde transport is regulated by the yeast Vps34 PtdIns 3-kinase. J Cell Sci. 2002;115(Pt 20):3889–900.PubMedCrossRef Burda P, et al. Retromer function in endosome-to-Golgi retrograde transport is regulated by the yeast Vps34 PtdIns 3-kinase. J Cell Sci. 2002;115(Pt 20):3889–900.PubMedCrossRef
108.
Zurück zum Zitat Nagy P, et al. Different effects of Atg2 and Atg18 mutations on Atg8a and Atg9 trafficking during starvation in drosophila. FEBS Lett. 2014;588(3):408–13.PubMedPubMedCentralCrossRef Nagy P, et al. Different effects of Atg2 and Atg18 mutations on Atg8a and Atg9 trafficking during starvation in drosophila. FEBS Lett. 2014;588(3):408–13.PubMedPubMedCentralCrossRef
109.
Zurück zum Zitat Noda T, et al. Regulation of membrane biogenesis in autophagy via PI3P dynamics. Semin Cell Dev Biol. 2010;21(7):671–6.PubMedCrossRef Noda T, et al. Regulation of membrane biogenesis in autophagy via PI3P dynamics. Semin Cell Dev Biol. 2010;21(7):671–6.PubMedCrossRef
110.
Zurück zum Zitat Noda NN, et al. Structure of the novel C-terminal domain of vacuolar protein sorting 30/autophagy-related protein 6 and its specific role in autophagy. J Biol Chem. 2012;287(20):16256–66.PubMedPubMedCentralCrossRef Noda NN, et al. Structure of the novel C-terminal domain of vacuolar protein sorting 30/autophagy-related protein 6 and its specific role in autophagy. J Biol Chem. 2012;287(20):16256–66.PubMedPubMedCentralCrossRef
111.
Zurück zum Zitat Fogel AI, et al. Role of membrane association and Atg14-dependent phosphorylation in beclin-1-mediated autophagy. Mol Cell Biol. 2013;33(18):3675–88.PubMedPubMedCentralCrossRef Fogel AI, et al. Role of membrane association and Atg14-dependent phosphorylation in beclin-1-mediated autophagy. Mol Cell Biol. 2013;33(18):3675–88.PubMedPubMedCentralCrossRef
112.
Zurück zum Zitat Backer JM. The regulation and function of class III PI3Ks: novel roles for Vps34. Biochem J. 2008;410(1):1–17.PubMedCrossRef Backer JM. The regulation and function of class III PI3Ks: novel roles for Vps34. Biochem J. 2008;410(1):1–17.PubMedCrossRef
113.
114.
Zurück zum Zitat Aita VM, et al. Cloning and genomic organization of beclin 1, a candidate tumor suppressor gene on chromosome 17q21. Genomics. 1999;59(1):59–65.PubMedCrossRef Aita VM, et al. Cloning and genomic organization of beclin 1, a candidate tumor suppressor gene on chromosome 17q21. Genomics. 1999;59(1):59–65.PubMedCrossRef
115.
Zurück zum Zitat Liang XH, et al. Induction of autophagy and inhibition of tumorigenesis by beclin 1. Nature. 1999;402(6762):672–6.PubMedCrossRef Liang XH, et al. Induction of autophagy and inhibition of tumorigenesis by beclin 1. Nature. 1999;402(6762):672–6.PubMedCrossRef
116.
Zurück zum Zitat Furuya N, et al. The evolutionarily conserved domain of Beclin 1 is required for Vps34 binding, autophagy and tumor suppressor function. Autophagy. 2005;1(1):46–52.PubMedCrossRef Furuya N, et al. The evolutionarily conserved domain of Beclin 1 is required for Vps34 binding, autophagy and tumor suppressor function. Autophagy. 2005;1(1):46–52.PubMedCrossRef
117.
Zurück zum Zitat Itakura E, et al. Beclin 1 forms two distinct phosphatidylinositol 3-kinase complexes with mammalian Atg14 and UVRAG. Mol Biol Cell. 2008;19(12):5360–72.PubMedPubMedCentralCrossRef Itakura E, et al. Beclin 1 forms two distinct phosphatidylinositol 3-kinase complexes with mammalian Atg14 and UVRAG. Mol Biol Cell. 2008;19(12):5360–72.PubMedPubMedCentralCrossRef
118.
Zurück zum Zitat Liang XH, et al. Protection against fatal Sindbis virus encephalitis by beclin, a novel Bcl-2-interacting protein. J Virol. 1998;72(11):8586–96.PubMedPubMedCentralCrossRef Liang XH, et al. Protection against fatal Sindbis virus encephalitis by beclin, a novel Bcl-2-interacting protein. J Virol. 1998;72(11):8586–96.PubMedPubMedCentralCrossRef
119.
Zurück zum Zitat Feng W, et al. Molecular basis of Bcl-xL's target recognition versatility revealed by the structure of Bcl-xL in complex with the BH3 domain of Beclin-1. J Mol Biol. 2007;372(1):223–35.PubMedCrossRef Feng W, et al. Molecular basis of Bcl-xL's target recognition versatility revealed by the structure of Bcl-xL in complex with the BH3 domain of Beclin-1. J Mol Biol. 2007;372(1):223–35.PubMedCrossRef
120.
Zurück zum Zitat Oberstein A, Jeffrey PD, Shi Y. Crystal structure of the Bcl-XL-Beclin 1 peptide complex: Beclin 1 is a novel BH3-only protein. J Biol Chem. 2007;282(17):13123–32.PubMedCrossRef Oberstein A, Jeffrey PD, Shi Y. Crystal structure of the Bcl-XL-Beclin 1 peptide complex: Beclin 1 is a novel BH3-only protein. J Biol Chem. 2007;282(17):13123–32.PubMedCrossRef
121.
Zurück zum Zitat Li X, et al. Imperfect interface of Beclin1 coiled-coil domain regulates homodimer and heterodimer formation with Atg14L and UVRAG. Nat Commun. 2012;3:662.PubMedCrossRef Li X, et al. Imperfect interface of Beclin1 coiled-coil domain regulates homodimer and heterodimer formation with Atg14L and UVRAG. Nat Commun. 2012;3:662.PubMedCrossRef
122.
123.
Zurück zum Zitat Yamamoto H, et al. Atg9 vesicles are an important membrane source during early steps of autophagosome formation. J Cell Biol. 2012;198(2):219–33.PubMedPubMedCentralCrossRef Yamamoto H, et al. Atg9 vesicles are an important membrane source during early steps of autophagosome formation. J Cell Biol. 2012;198(2):219–33.PubMedPubMedCentralCrossRef
124.
Zurück zum Zitat Orsi A, et al. Dynamic and transient interactions of Atg9 with autophagosomes, but not membrane integration, are required for autophagy. Mol Biol Cell. 2012;23(10):1860–73.PubMedPubMedCentralCrossRef Orsi A, et al. Dynamic and transient interactions of Atg9 with autophagosomes, but not membrane integration, are required for autophagy. Mol Biol Cell. 2012;23(10):1860–73.PubMedPubMedCentralCrossRef
125.
126.
Zurück zum Zitat Noda T, et al. Apg9p/Cvt7p is an integral membrane protein required for transport vesicle formation in the Cvt and autophagy pathways. J Cell Biol. 2000;148(3):465–80.PubMedPubMedCentralCrossRef Noda T, et al. Apg9p/Cvt7p is an integral membrane protein required for transport vesicle formation in the Cvt and autophagy pathways. J Cell Biol. 2000;148(3):465–80.PubMedPubMedCentralCrossRef
127.
Zurück zum Zitat Young AR, et al. Starvation and ULK1-dependent cycling of mammalian Atg9 between the TGN and endosomes. J Cell Sci. 2006;119(Pt 18):3888–900.PubMedCrossRef Young AR, et al. Starvation and ULK1-dependent cycling of mammalian Atg9 between the TGN and endosomes. J Cell Sci. 2006;119(Pt 18):3888–900.PubMedCrossRef
128.
Zurück zum Zitat Yamada T, et al. Endothelial nitric-oxide synthase antisense (NOS3AS) gene encodes an autophagy-related protein (APG9-like2) highly expressed in trophoblast. J Biol Chem. 2005;280(18):18283–90.PubMedCrossRef Yamada T, et al. Endothelial nitric-oxide synthase antisense (NOS3AS) gene encodes an autophagy-related protein (APG9-like2) highly expressed in trophoblast. J Biol Chem. 2005;280(18):18283–90.PubMedCrossRef
129.
Zurück zum Zitat He C, et al. Self-interaction is critical for Atg9 transport and function at the phagophore assembly site during autophagy. Mol Biol Cell. 2008;19(12):5506–16.PubMedPubMedCentralCrossRef He C, et al. Self-interaction is critical for Atg9 transport and function at the phagophore assembly site during autophagy. Mol Biol Cell. 2008;19(12):5506–16.PubMedPubMedCentralCrossRef
131.
Zurück zum Zitat Feng Y, et al. Phosphorylation of Atg9 regulates movement to the phagophore assembly site and the rate of autophagosome formation. Autophagy. 2016;12(4):648–58.PubMedPubMedCentralCrossRef Feng Y, et al. Phosphorylation of Atg9 regulates movement to the phagophore assembly site and the rate of autophagosome formation. Autophagy. 2016;12(4):648–58.PubMedPubMedCentralCrossRef
132.
Zurück zum Zitat Reggiori F, et al. Atg9 cycles between mitochondria and the pre-autophagosomal structure in yeasts. Autophagy. 2005;1(2):101–9.PubMedCrossRef Reggiori F, et al. Atg9 cycles between mitochondria and the pre-autophagosomal structure in yeasts. Autophagy. 2005;1(2):101–9.PubMedCrossRef
133.
134.
Zurück zum Zitat Jia S, et al. Mammalian Atg9 contributes to the post-Golgi transport of lysosomal hydrolases by interacting with adaptor protein-1. FEBS Lett. 2017;591(24):4027–38.PubMedCrossRef Jia S, et al. Mammalian Atg9 contributes to the post-Golgi transport of lysosomal hydrolases by interacting with adaptor protein-1. FEBS Lett. 2017;591(24):4027–38.PubMedCrossRef
135.
Zurück zum Zitat Shirahama-Noda K, et al. TRAPPIII is responsible for vesicular transport from early endosomes to Golgi, facilitating Atg9 cycling in autophagy. J Cell Sci. 2013;126(Pt 21):4963–73.PubMedCrossRef Shirahama-Noda K, et al. TRAPPIII is responsible for vesicular transport from early endosomes to Golgi, facilitating Atg9 cycling in autophagy. J Cell Sci. 2013;126(Pt 21):4963–73.PubMedCrossRef
136.
Zurück zum Zitat Karanasios E, et al. Autophagy initiation by ULK complex assembly on ER tubulovesicular regions marked by ATG9 vesicles. Nat Commun. 2016;7:12420.PubMedPubMedCentralCrossRef Karanasios E, et al. Autophagy initiation by ULK complex assembly on ER tubulovesicular regions marked by ATG9 vesicles. Nat Commun. 2016;7:12420.PubMedPubMedCentralCrossRef
137.
Zurück zum Zitat Obara K, et al. The Atg18-Atg2 complex is recruited to autophagic membranes via phosphatidylinositol 3-phosphate and exerts an essential function. J Biol Chem. 2008;283(35):23972–80.PubMedPubMedCentralCrossRef Obara K, et al. The Atg18-Atg2 complex is recruited to autophagic membranes via phosphatidylinositol 3-phosphate and exerts an essential function. J Biol Chem. 2008;283(35):23972–80.PubMedPubMedCentralCrossRef
138.
Zurück zum Zitat Yang Z, Klionsky DJ. Mammalian autophagy: core molecular machinery and signaling regulation. Curr Opin Cell Biol. 2010;22(2):124–31.PubMedCrossRef Yang Z, Klionsky DJ. Mammalian autophagy: core molecular machinery and signaling regulation. Curr Opin Cell Biol. 2010;22(2):124–31.PubMedCrossRef
140.
Zurück zum Zitat Polson HE, et al. Mammalian Atg18 (WIPI2) localizes to omegasome-anchored phagophores and positively regulates LC3 lipidation. Autophagy. 2010;6(4):506–22.PubMedCrossRef Polson HE, et al. Mammalian Atg18 (WIPI2) localizes to omegasome-anchored phagophores and positively regulates LC3 lipidation. Autophagy. 2010;6(4):506–22.PubMedCrossRef
141.
Zurück zum Zitat Muller AJ, Proikas-Cezanne T. Function of human WIPI proteins in autophagosomal rejuvenation of endomembranes? FEBS Lett. 2015;589(14):1546–51.PubMedCrossRef Muller AJ, Proikas-Cezanne T. Function of human WIPI proteins in autophagosomal rejuvenation of endomembranes? FEBS Lett. 2015;589(14):1546–51.PubMedCrossRef
142.
Zurück zum Zitat Suzuki H, et al. Structural biology of the core autophagy machinery. Curr Opin Struct Biol. 2017;43:10–7.PubMedCrossRef Suzuki H, et al. Structural biology of the core autophagy machinery. Curr Opin Struct Biol. 2017;43:10–7.PubMedCrossRef
145.
Zurück zum Zitat Kuma A, et al. Formation of the similar to 350-kDa Apg12-Apg5 center dot Apg16 multimeric complex, mediated by Apg16 oligomerization, is essential for autophagy in yeast. J Biol Chem. 2002;277(21):18619–25.PubMedCrossRef Kuma A, et al. Formation of the similar to 350-kDa Apg12-Apg5 center dot Apg16 multimeric complex, mediated by Apg16 oligomerization, is essential for autophagy in yeast. J Biol Chem. 2002;277(21):18619–25.PubMedCrossRef
146.
Zurück zum Zitat Mizushima N, Noda T, Ohsumi Y. Apg16p is required for the function of the Apg12p-Apg5p conjugate in the yeast autophagy pathway. EMBO J. 1999;18(14):3888–96.PubMedPubMedCentralCrossRef Mizushima N, Noda T, Ohsumi Y. Apg16p is required for the function of the Apg12p-Apg5p conjugate in the yeast autophagy pathway. EMBO J. 1999;18(14):3888–96.PubMedPubMedCentralCrossRef
147.
Zurück zum Zitat Mizushima N, et al. Mouse Apg16L, a novel WD-repeat protein, targets to the autophagic isolation membrane with the Apg12-Apg5 conjugate. J Cell Sci. 2003;116(Pt 9):1679–88.PubMedCrossRef Mizushima N, et al. Mouse Apg16L, a novel WD-repeat protein, targets to the autophagic isolation membrane with the Apg12-Apg5 conjugate. J Cell Sci. 2003;116(Pt 9):1679–88.PubMedCrossRef
148.
Zurück zum Zitat Kirisako T, et al. The reversible modification regulates the membrane-binding state of Apg8/Aut7 essential for autophagy and the cytoplasm to vacuole targeting pathway. J Cell Biol. 2000;151(2):263–76.PubMedPubMedCentralCrossRef Kirisako T, et al. The reversible modification regulates the membrane-binding state of Apg8/Aut7 essential for autophagy and the cytoplasm to vacuole targeting pathway. J Cell Biol. 2000;151(2):263–76.PubMedPubMedCentralCrossRef
149.
Zurück zum Zitat Yamada Y, et al. The crystal structure of Atg3, an autophagy-related ubiquitin carrier protein (E2) enzyme that mediates Atg8 lipidation. J Biol Chem. 2007;282(11):8036–43.PubMedCrossRef Yamada Y, et al. The crystal structure of Atg3, an autophagy-related ubiquitin carrier protein (E2) enzyme that mediates Atg8 lipidation. J Biol Chem. 2007;282(11):8036–43.PubMedCrossRef
150.
Zurück zum Zitat Huang WP, et al. The itinerary of a vesicle component, Aut7p/Cvt5p, terminates in the yeast vacuole via the autophagy/Cvt pathways. J Biol Chem. 2000;275(8):5845–51.PubMedCrossRef Huang WP, et al. The itinerary of a vesicle component, Aut7p/Cvt5p, terminates in the yeast vacuole via the autophagy/Cvt pathways. J Biol Chem. 2000;275(8):5845–51.PubMedCrossRef
151.
Zurück zum Zitat Kabeya Y, et al. LC3, GABARAP and GATE16 localize to autophagosomal membrane depending on form-II formation. J Cell Sci. 2004;117(Pt 13):2805–12.PubMedCrossRef Kabeya Y, et al. LC3, GABARAP and GATE16 localize to autophagosomal membrane depending on form-II formation. J Cell Sci. 2004;117(Pt 13):2805–12.PubMedCrossRef
152.
Zurück zum Zitat Tanida I, et al. GATE-16 and GABARAP are authentic modifiers mediated by Apg7 and Apg3. Biochem Biophys Res Commun. 2003;300(3):637–44.PubMedCrossRef Tanida I, et al. GATE-16 and GABARAP are authentic modifiers mediated by Apg7 and Apg3. Biochem Biophys Res Commun. 2003;300(3):637–44.PubMedCrossRef
153.
Zurück zum Zitat Tanida I, et al. Atg8L/Apg8L is the fourth mammalian modifier of mammalian Atg8 conjugation mediated by human Atg4B, Atg7 and Atg3. FEBS J. 2006;273(11):2553–62.PubMedCrossRef Tanida I, et al. Atg8L/Apg8L is the fourth mammalian modifier of mammalian Atg8 conjugation mediated by human Atg4B, Atg7 and Atg3. FEBS J. 2006;273(11):2553–62.PubMedCrossRef
154.
Zurück zum Zitat Tanida I, et al. Human Apg3p/Aut1p homologue is an authentic E2 enzyme for multiple substrates, GATE-16, GABARAP, and MAP-LC3, and facilitates the conjugation of hApg12p to hApg5p. J Biol Chem. 2002;277(16):13739–44.PubMedCrossRef Tanida I, et al. Human Apg3p/Aut1p homologue is an authentic E2 enzyme for multiple substrates, GATE-16, GABARAP, and MAP-LC3, and facilitates the conjugation of hApg12p to hApg5p. J Biol Chem. 2002;277(16):13739–44.PubMedCrossRef
155.
Zurück zum Zitat Tanida I, et al. The human homolog of Saccharomyces cerevisiae Apg7p is a protein-activating enzyme for multiple substrates including human Apg12p, GATE-16, GABARAP, and MAP-LC3. J Biol Chem. 2001;276(3):1701–6.PubMedCrossRef Tanida I, et al. The human homolog of Saccharomyces cerevisiae Apg7p is a protein-activating enzyme for multiple substrates including human Apg12p, GATE-16, GABARAP, and MAP-LC3. J Biol Chem. 2001;276(3):1701–6.PubMedCrossRef
157.
Zurück zum Zitat Kabeya Y, et al. LC3, a mammalian homologue of yeast Apg8p, is localized in autophagosome membranes after processing. EMBO J. 2000;19(21):5720–8.PubMedPubMedCentralCrossRef Kabeya Y, et al. LC3, a mammalian homologue of yeast Apg8p, is localized in autophagosome membranes after processing. EMBO J. 2000;19(21):5720–8.PubMedPubMedCentralCrossRef
158.
Zurück zum Zitat Tanida I, Ueno T, Kominami E. Human light chain 3/MAP 1LC3B is cleaved at its carboxyl-terminal Met121 to expose Gly120 for lipidation and targeting to autophagosomal membranes. J Biol Chem. 2004;279(46):47704–10.PubMedCrossRef Tanida I, Ueno T, Kominami E. Human light chain 3/MAP 1LC3B is cleaved at its carboxyl-terminal Met121 to expose Gly120 for lipidation and targeting to autophagosomal membranes. J Biol Chem. 2004;279(46):47704–10.PubMedCrossRef
159.
Zurück zum Zitat Tanida I, et al. Lysosomal turnover of GABARAP-phospholipid conjugate is activated during differentiation of C2C12 cells to myotubes without inactivation of the mTor kinase-signaling pathway. Autophagy. 2006;2(4):264–71.PubMedCrossRef Tanida I, et al. Lysosomal turnover of GABARAP-phospholipid conjugate is activated during differentiation of C2C12 cells to myotubes without inactivation of the mTor kinase-signaling pathway. Autophagy. 2006;2(4):264–71.PubMedCrossRef
160.
Zurück zum Zitat Mizushima N. Physiological functions of autophagy. Curr Top Microbiol Immunol. 2009;335:71–84.PubMed Mizushima N. Physiological functions of autophagy. Curr Top Microbiol Immunol. 2009;335:71–84.PubMed
161.
Zurück zum Zitat Ravikumar B, et al. Regulation of mammalian autophagy in physiology and pathophysiology. Physiol Rev. 2010;90(4):1383–435.PubMedCrossRef Ravikumar B, et al. Regulation of mammalian autophagy in physiology and pathophysiology. Physiol Rev. 2010;90(4):1383–435.PubMedCrossRef
163.
Zurück zum Zitat Wang L, Ye X, Zhao T. The physiological roles of autophagy in the mammalian life cycle. Biol Rev Camb Philos Soc. 2019;94(2):503-16.PubMedCrossRef Wang L, Ye X, Zhao T. The physiological roles of autophagy in the mammalian life cycle. Biol Rev Camb Philos Soc. 2019;94(2):503-16.PubMedCrossRef
164.
Zurück zum Zitat Kuma A, Mizushima N. Physiological role of autophagy as an intracellular recycling system: with an emphasis on nutrient metabolism. Semin Cell Dev Biol. 2010;21(7):683–90.PubMedCrossRef Kuma A, Mizushima N. Physiological role of autophagy as an intracellular recycling system: with an emphasis on nutrient metabolism. Semin Cell Dev Biol. 2010;21(7):683–90.PubMedCrossRef
165.
166.
167.
169.
Zurück zum Zitat Sridhar S, et al. Autophagy and disease: always two sides to a problem. J Pathol. 2012;226(2):255–73.PubMedCrossRef Sridhar S, et al. Autophagy and disease: always two sides to a problem. J Pathol. 2012;226(2):255–73.PubMedCrossRef
171.
173.
Zurück zum Zitat Levy JMM, Towers CG, Thorburn A. Targeting autophagy in cancer. Nat Rev Cancer. 2017;17(9):528-42.PubMedCrossRef Levy JMM, Towers CG, Thorburn A. Targeting autophagy in cancer. Nat Rev Cancer. 2017;17(9):528-42.PubMedCrossRef
174.
Zurück zum Zitat Eskelinen EL. The dual role of autophagy in cancer. Curr Opin Pharmacol. 2011;11(4):294–300.PubMedCrossRef Eskelinen EL. The dual role of autophagy in cancer. Curr Opin Pharmacol. 2011;11(4):294–300.PubMedCrossRef
175.
Zurück zum Zitat Singh SS, et al. Dual role of autophagy in hallmarks of cancer. Oncogene. 2018;37(9):1142–58.PubMedCrossRef Singh SS, et al. Dual role of autophagy in hallmarks of cancer. Oncogene. 2018;37(9):1142–58.PubMedCrossRef
177.
Zurück zum Zitat Jin S, et al. Autophagy regulation and its dual role in blood cancers: a novel target for therapeutic development (review). Oncol Rep. 2018;39(6):2473–81.PubMed Jin S, et al. Autophagy regulation and its dual role in blood cancers: a novel target for therapeutic development (review). Oncol Rep. 2018;39(6):2473–81.PubMed
178.
Zurück zum Zitat Rao S, et al. A dual role for autophagy in a murine model of lung cancer. Nat Commun. 2014;5:3056.PubMedCrossRef Rao S, et al. A dual role for autophagy in a murine model of lung cancer. Nat Commun. 2014;5:3056.PubMedCrossRef
180.
184.
Zurück zum Zitat Ashrafi G, Schwarz TL. The pathways of mitophagy for quality control and clearance of mitochondria. Cell Death Differ. 2013;20(1):31–42.PubMedCrossRef Ashrafi G, Schwarz TL. The pathways of mitophagy for quality control and clearance of mitochondria. Cell Death Differ. 2013;20(1):31–42.PubMedCrossRef
186.
187.
188.
189.
Zurück zum Zitat Peng YF, et al. Autophagy inhibition suppresses pulmonary metastasis of HCC in mice via impairing anoikis resistance and colonization of HCC cells. Autophagy. 2013;9(12):2056–68.PubMedCrossRef Peng YF, et al. Autophagy inhibition suppresses pulmonary metastasis of HCC in mice via impairing anoikis resistance and colonization of HCC cells. Autophagy. 2013;9(12):2056–68.PubMedCrossRef
190.
Zurück zum Zitat Ding ZB, et al. Association of autophagy defect with a malignant phenotype and poor prognosis of hepatocellular carcinoma. Cancer Res. 2008;68(22):9167–75.PubMedCrossRef Ding ZB, et al. Association of autophagy defect with a malignant phenotype and poor prognosis of hepatocellular carcinoma. Cancer Res. 2008;68(22):9167–75.PubMedCrossRef
191.
Zurück zum Zitat Jin S, White E. Role of autophagy in cancer: management of metabolic stress. Autophagy. 2007;3(1):28–31.PubMedCrossRef Jin S, White E. Role of autophagy in cancer: management of metabolic stress. Autophagy. 2007;3(1):28–31.PubMedCrossRef
192.
Zurück zum Zitat Yue Z, et al. 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.PubMedPubMedCentralCrossRef Yue Z, et al. 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.PubMedPubMedCentralCrossRef
193.
194.
195.
Zurück zum Zitat Kang MR, et al. Frameshift mutations of autophagy-related genes ATG2B, ATG5, ATG9B and ATG12 in gastric and colorectal cancers with microsatellite instability. J Pathol. 2009;217(5):702–6.PubMedCrossRef Kang MR, et al. Frameshift mutations of autophagy-related genes ATG2B, ATG5, ATG9B and ATG12 in gastric and colorectal cancers with microsatellite instability. J Pathol. 2009;217(5):702–6.PubMedCrossRef
197.
Zurück zum Zitat An CH, et al. Mutational and expressional analyses of ATG5, an autophagy-related gene, in gastrointestinal cancers. Pathol Res Pract. 2011;207(7):433–7.PubMedCrossRef An CH, et al. Mutational and expressional analyses of ATG5, an autophagy-related gene, in gastrointestinal cancers. Pathol Res Pract. 2011;207(7):433–7.PubMedCrossRef
198.
Zurück zum Zitat Capparelli C, et al. Autophagy and senescence in cancer-associated fibroblasts metabolically supports tumor growth and metastasis via glycolysis and ketone production. Cell Cycle. 2012;11(12):2285–302.PubMedPubMedCentralCrossRef Capparelli C, et al. Autophagy and senescence in cancer-associated fibroblasts metabolically supports tumor growth and metastasis via glycolysis and ketone production. Cell Cycle. 2012;11(12):2285–302.PubMedPubMedCentralCrossRef
200.
Zurück zum Zitat Shi CS, et al. Activation of autophagy by inflammatory signals limits IL-1beta production by targeting ubiquitinated inflammasomes for destruction. Nat Immunol. 2012;13(3):255–63.PubMedPubMedCentralCrossRef Shi CS, et al. Activation of autophagy by inflammatory signals limits IL-1beta production by targeting ubiquitinated inflammasomes for destruction. Nat Immunol. 2012;13(3):255–63.PubMedPubMedCentralCrossRef
201.
202.
Zurück zum Zitat Saitoh T, et al. Loss of the autophagy protein Atg16L1 enhances endotoxin-induced IL-1beta production. Nature. 2008;456(7219):264–8.PubMedCrossRef Saitoh T, et al. Loss of the autophagy protein Atg16L1 enhances endotoxin-induced IL-1beta production. Nature. 2008;456(7219):264–8.PubMedCrossRef
203.
Zurück zum Zitat Kwong C, Gilman-Sachs A, Beaman K. Tumor-associated a2 vacuolar ATPase acts as a key mediator of cancer-related inflammation by inducing pro-tumorigenic properties in monocytes. J Immunol. 2011;186(3):1781–9.PubMedCrossRef Kwong C, Gilman-Sachs A, Beaman K. Tumor-associated a2 vacuolar ATPase acts as a key mediator of cancer-related inflammation by inducing pro-tumorigenic properties in monocytes. J Immunol. 2011;186(3):1781–9.PubMedCrossRef
205.
206.
Zurück zum Zitat Balkwill FR, Mantovani A. Cancer-related inflammation: common themes and therapeutic opportunities. Semin Cancer Biol. 2012;22(1):33–40.PubMedCrossRef Balkwill FR, Mantovani A. Cancer-related inflammation: common themes and therapeutic opportunities. Semin Cancer Biol. 2012;22(1):33–40.PubMedCrossRef
211.
Zurück zum Zitat Bjorkoy G, et al. p62/SQSTM1 forms protein aggregates degraded by autophagy and has a protective effect on huntingtin-induced cell death. J Cell Biol. 2005;171(4):603–14.PubMedPubMedCentralCrossRef Bjorkoy G, et al. p62/SQSTM1 forms protein aggregates degraded by autophagy and has a protective effect on huntingtin-induced cell death. J Cell Biol. 2005;171(4):603–14.PubMedPubMedCentralCrossRef
212.
Zurück zum Zitat Pankiv S, et al. p62/SQSTM1 binds directly to Atg8/LC3 to facilitate degradation of ubiquitinated protein aggregates by autophagy. J Biol Chem. 2007;282(33):24131–45.PubMedCrossRef Pankiv S, et al. p62/SQSTM1 binds directly to Atg8/LC3 to facilitate degradation of ubiquitinated protein aggregates by autophagy. J Biol Chem. 2007;282(33):24131–45.PubMedCrossRef
214.
Zurück zum Zitat Su Y, et al. The diversity expression of p62 in digestive system cancers. Clin Immunol. 2005;116(2):118–23.PubMedCrossRef Su Y, et al. The diversity expression of p62 in digestive system cancers. Clin Immunol. 2005;116(2):118–23.PubMedCrossRef
215.
Zurück zum Zitat Kitamura H, et al. Cytosolic overexpression of p62 sequestosome 1 in neoplastic prostate tissue. Histopathology. 2006;48(2):157–61.PubMedCrossRef Kitamura H, et al. Cytosolic overexpression of p62 sequestosome 1 in neoplastic prostate tissue. Histopathology. 2006;48(2):157–61.PubMedCrossRef
216.
Zurück zum Zitat Valencia T, et al. Metabolic reprogramming of stromal fibroblasts through p62-mTORC1 signaling promotes inflammation and tumorigenesis. Cancer Cell. 2014;26(1):121–35.PubMedPubMedCentralCrossRef Valencia T, et al. Metabolic reprogramming of stromal fibroblasts through p62-mTORC1 signaling promotes inflammation and tumorigenesis. Cancer Cell. 2014;26(1):121–35.PubMedPubMedCentralCrossRef
217.
Zurück zum Zitat Stumptner C, et al. Analysis of intracytoplasmic hyaline bodies in a hepatocellular carcinoma. Demonstration of p62 as major constituent. Am J Pathol. 1999;154(6):1701–10.PubMedPubMedCentralCrossRef Stumptner C, et al. Analysis of intracytoplasmic hyaline bodies in a hepatocellular carcinoma. Demonstration of p62 as major constituent. Am J Pathol. 1999;154(6):1701–10.PubMedPubMedCentralCrossRef
218.
Zurück zum Zitat Saito T, et al. p62/Sqstm1 promotes malignancy of HCV-positive hepatocellular carcinoma through Nrf2-dependent metabolic reprogramming. Nat Commun. 2016;7:12030.PubMedPubMedCentralCrossRef Saito T, et al. p62/Sqstm1 promotes malignancy of HCV-positive hepatocellular carcinoma through Nrf2-dependent metabolic reprogramming. Nat Commun. 2016;7:12030.PubMedPubMedCentralCrossRef
219.
Zurück zum Zitat Umemura A, et al. p62, Upregulated during Preneoplasia, induces hepatocellular carcinogenesis by maintaining survival of stressed HCC-initiating cells. Cancer Cell. 2016;29(6):935–48.PubMedPubMedCentralCrossRef Umemura A, et al. p62, Upregulated during Preneoplasia, induces hepatocellular carcinogenesis by maintaining survival of stressed HCC-initiating cells. Cancer Cell. 2016;29(6):935–48.PubMedPubMedCentralCrossRef
220.
Zurück zum Zitat Thompson HG, et al. p62 overexpression in breast tumors and regulation by prostate-derived Ets factor in breast cancer cells. Oncogene. 2003;22(15):2322–33.PubMedCrossRef Thompson HG, et al. p62 overexpression in breast tumors and regulation by prostate-derived Ets factor in breast cancer cells. Oncogene. 2003;22(15):2322–33.PubMedCrossRef
222.
Zurück zum Zitat Inoue D, et al. Accumulation of p62/SQSTM1 is associated with poor prognosis in patients with lung adenocarcinoma. Cancer Sci. 2012;103(4):760–6.PubMedCrossRefPubMedCentral Inoue D, et al. Accumulation of p62/SQSTM1 is associated with poor prognosis in patients with lung adenocarcinoma. Cancer Sci. 2012;103(4):760–6.PubMedCrossRefPubMedCentral
223.
Zurück zum Zitat Huang J, et al. Adipocyte p62/SQSTM1 suppresses tumorigenesis through opposite regulations of metabolism in adipose tissue and tumor. Cancer Cell. 2018;33(4):770–84 e6.PubMedPubMedCentralCrossRef Huang J, et al. Adipocyte p62/SQSTM1 suppresses tumorigenesis through opposite regulations of metabolism in adipose tissue and tumor. Cancer Cell. 2018;33(4):770–84 e6.PubMedPubMedCentralCrossRef
224.
Zurück zum Zitat Parkhitko A, et al. Tumorigenesis in tuberous sclerosis complex is autophagy and p62/sequestosome 1 (SQSTM1)-dependent. Proc Natl Acad Sci U S A. 2011;108(30):12455–60.PubMedPubMedCentralCrossRef Parkhitko A, et al. Tumorigenesis in tuberous sclerosis complex is autophagy and p62/sequestosome 1 (SQSTM1)-dependent. Proc Natl Acad Sci U S A. 2011;108(30):12455–60.PubMedPubMedCentralCrossRef
225.
Zurück zum Zitat Jin S, et al. Metabolic catastrophe as a means to cancer cell death. J Cell Sci. 2007;120(Pt 3):379–83.PubMedCrossRef Jin S, et al. Metabolic catastrophe as a means to cancer cell death. J Cell Sci. 2007;120(Pt 3):379–83.PubMedCrossRef
226.
Zurück zum Zitat Degenhardt K, et al. Autophagy promotes tumor cell survival and restricts necrosis, inflammation, and tumorigenesis. Cancer Cell. 2006;10(1):51–64.PubMedPubMedCentralCrossRef Degenhardt K, et al. Autophagy promotes tumor cell survival and restricts necrosis, inflammation, and tumorigenesis. Cancer Cell. 2006;10(1):51–64.PubMedPubMedCentralCrossRef
227.
Zurück zum Zitat Lum JJ, et al. Growth factor regulation of autophagy and cell survival in the absence of apoptosis. Cell. 2005;120(2):237–48.PubMedCrossRef Lum JJ, et al. Growth factor regulation of autophagy and cell survival in the absence of apoptosis. Cell. 2005;120(2):237–48.PubMedCrossRef
228.
Zurück zum Zitat Ahn CH, et al. Expression of beclin-1, an autophagy-related protein, in gastric and colorectal cancers. APMIS. 2007;115(12):1344–9.PubMedCrossRef Ahn CH, et al. Expression of beclin-1, an autophagy-related protein, in gastric and colorectal cancers. APMIS. 2007;115(12):1344–9.PubMedCrossRef
229.
Zurück zum Zitat Tang H, et al. Hepatitis B virus X protein sensitizes cells to starvation-induced autophagy via up-regulation of beclin 1 expression. Hepatology. 2009;49(1):60–71.PubMedCrossRef Tang H, et al. Hepatitis B virus X protein sensitizes cells to starvation-induced autophagy via up-regulation of beclin 1 expression. Hepatology. 2009;49(1):60–71.PubMedCrossRef
230.
Zurück zum Zitat Karantza-Wadsworth V, White E. Role of autophagy in breast cancer. Autophagy. 2007;3(6):610–3.PubMedCrossRef Karantza-Wadsworth V, White E. Role of autophagy in breast cancer. Autophagy. 2007;3(6):610–3.PubMedCrossRef
231.
Zurück zum Zitat Sun Y, et al. Over-expression of the Beclin1 gene upregulates chemosensitivity to anti-cancer drugs by enhancing therapy-induced apoptosis in cervix squamous carcinoma CaSki cells. Cancer Lett. 2010;294(2):204–10.PubMedCrossRef Sun Y, et al. Over-expression of the Beclin1 gene upregulates chemosensitivity to anti-cancer drugs by enhancing therapy-induced apoptosis in cervix squamous carcinoma CaSki cells. Cancer Lett. 2010;294(2):204–10.PubMedCrossRef
232.
233.
Zurück zum Zitat Weinberg F, et al. Mitochondrial metabolism and ROS generation are essential for Kras-mediated tumorigenicity. Proc Natl Acad Sci U S A. 2010;107(19):8788–93.PubMedPubMedCentralCrossRef Weinberg F, et al. Mitochondrial metabolism and ROS generation are essential for Kras-mediated tumorigenicity. Proc Natl Acad Sci U S A. 2010;107(19):8788–93.PubMedPubMedCentralCrossRef
234.
238.
Zurück zum Zitat Maishman T, et al. Local recurrence and breast oncological surgery in Young women with breast Cancer: the POSH observational cohort study. Ann Surg. 2017;266(1):165–72.PubMedCrossRef Maishman T, et al. Local recurrence and breast oncological surgery in Young women with breast Cancer: the POSH observational cohort study. Ann Surg. 2017;266(1):165–72.PubMedCrossRef
239.
Zurück zum Zitat Alsarraj J, Hunter KW. Bromodomain-containing protein 4: a dynamic regulator of breast Cancer metastasis through modulation of the extracellular matrix. Int J Breast Cancer. 2012;2012:670632.PubMedCrossRef Alsarraj J, Hunter KW. Bromodomain-containing protein 4: a dynamic regulator of breast Cancer metastasis through modulation of the extracellular matrix. Int J Breast Cancer. 2012;2012:670632.PubMedCrossRef
240.
Zurück zum Zitat Chaffer CL, Weinberg RA. A perspective on cancer cell metastasis. Science. 2011;331(6024):1559–64.PubMedCrossRef Chaffer CL, Weinberg RA. A perspective on cancer cell metastasis. Science. 2011;331(6024):1559–64.PubMedCrossRef
243.
Zurück zum Zitat Kenific CM, Thorburn A, Debnath J. Autophagy and metastasis: another double-edged sword. Curr Opin Cell Biol. 2010;22(2):241–5.PubMedCrossRef Kenific CM, Thorburn A, Debnath J. Autophagy and metastasis: another double-edged sword. Curr Opin Cell Biol. 2010;22(2):241–5.PubMedCrossRef
244.
246.
Zurück zum Zitat Lu Z, et al. The tumor suppressor gene ARHI regulates autophagy and tumor dormancy in human ovarian cancer cells. J Clin Invest. 2008;118(12):3917–29.PubMedPubMedCentral Lu Z, et al. The tumor suppressor gene ARHI regulates autophagy and tumor dormancy in human ovarian cancer cells. J Clin Invest. 2008;118(12):3917–29.PubMedPubMedCentral
249.
Zurück zum Zitat Lazova R, et al. 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.PubMedCrossRef Lazova R, et al. 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.PubMedCrossRef
250.
Zurück zum Zitat Zhao H, et al. High expression of LC3B is associated with progression and poor outcome in triple-negative breast cancer. Med Oncol. 2013;30(1):475.PubMedCrossRef Zhao H, et al. High expression of LC3B is associated with progression and poor outcome in triple-negative breast cancer. Med Oncol. 2013;30(1):475.PubMedCrossRef
251.
Zurück zum Zitat Lazova R, Klump V, Pawelek J. Autophagy in cutaneous malignant melanoma. J Cutan Pathol. 2010;37(2):256–68.PubMedCrossRef Lazova R, Klump V, Pawelek J. Autophagy in cutaneous malignant melanoma. J Cutan Pathol. 2010;37(2):256–68.PubMedCrossRef
252.
Zurück zum Zitat Galavotti S, et al. The autophagy-associated factors DRAM1 and p62 regulate cell migration and invasion in glioblastoma stem cells. Oncogene. 2013;32(6):699–712.PubMedCrossRef Galavotti S, et al. The autophagy-associated factors DRAM1 and p62 regulate cell migration and invasion in glioblastoma stem cells. Oncogene. 2013;32(6):699–712.PubMedCrossRef
253.
Zurück zum Zitat Zheng HY, et al. Autophagy enhances the aggressiveness of human colorectal cancer cells and their ability to adapt to apoptotic stimulus. Cancer Biol Med. 2012;9(2):105–10.PubMedPubMedCentral Zheng HY, et al. Autophagy enhances the aggressiveness of human colorectal cancer cells and their ability to adapt to apoptotic stimulus. Cancer Biol Med. 2012;9(2):105–10.PubMedPubMedCentral
255.
Zurück zum Zitat Classen F, et al. Autophagy induced by ionizing radiation promotes cell death over survival in human colorectal cancer cells. Exp Cell Res. 2019;374(1):29–37.PubMedCrossRef Classen F, et al. Autophagy induced by ionizing radiation promotes cell death over survival in human colorectal cancer cells. Exp Cell Res. 2019;374(1):29–37.PubMedCrossRef
256.
Zurück zum Zitat Zois CE, Koukourakis MI. Radiation-induced autophagy in normal and cancer cells: towards novel cytoprotection and radio-sensitization policies? Autophagy. 2009;5(4):442–50.PubMedCrossRef Zois CE, Koukourakis MI. Radiation-induced autophagy in normal and cancer cells: towards novel cytoprotection and radio-sensitization policies? Autophagy. 2009;5(4):442–50.PubMedCrossRef
257.
Zurück zum Zitat Garbar C, et al. Chemotherapy treatment induces an increase of autophagy in the luminal breast cancer cell MCF7, but not in the triple-negative MDA-MB231. Sci Rep. 2017;7(1):7201.PubMedPubMedCentralCrossRef Garbar C, et al. Chemotherapy treatment induces an increase of autophagy in the luminal breast cancer cell MCF7, but not in the triple-negative MDA-MB231. Sci Rep. 2017;7(1):7201.PubMedPubMedCentralCrossRef
258.
260.
Zurück zum Zitat Kanzawa T, et al. Induction of autophagic cell death in malignant glioma cells by arsenic trioxide. Cancer Res. 2003;63(9):2103–8.PubMed Kanzawa T, et al. Induction of autophagic cell death in malignant glioma cells by arsenic trioxide. Cancer Res. 2003;63(9):2103–8.PubMed
261.
Zurück zum Zitat Kanzawa T, et al. Arsenic trioxide induces autophagic cell death in malignant glioma cells by upregulation of mitochondrial cell death protein BNIP3. Oncogene. 2005;24(6):980–91.PubMedCrossRef Kanzawa T, et al. Arsenic trioxide induces autophagic cell death in malignant glioma cells by upregulation of mitochondrial cell death protein BNIP3. Oncogene. 2005;24(6):980–91.PubMedCrossRef
262.
Zurück zum Zitat Kanzawa T, et al. Role of autophagy in temozolomide-induced cytotoxicity for malignant glioma cells. Cell Death Differ. 2004;11(4):448–57.PubMedCrossRef Kanzawa T, et al. Role of autophagy in temozolomide-induced cytotoxicity for malignant glioma cells. Cell Death Differ. 2004;11(4):448–57.PubMedCrossRef
263.
Zurück zum Zitat Ito H, et al. Radiation-induced autophagy is associated with LC3 and its inhibition sensitizes malignant glioma cells. Int J Oncol. 2005;26(5):1401–10.PubMed Ito H, et al. Radiation-induced autophagy is associated with LC3 and its inhibition sensitizes malignant glioma cells. Int J Oncol. 2005;26(5):1401–10.PubMed
264.
Zurück zum Zitat Paglin S, et al. A novel response of cancer cells to radiation involves autophagy and formation of acidic vesicles. Cancer Res. 2001;61(2):439–44.PubMed Paglin S, et al. A novel response of cancer cells to radiation involves autophagy and formation of acidic vesicles. Cancer Res. 2001;61(2):439–44.PubMed
265.
Zurück zum Zitat Yao KC, et al. Molecular response of human glioblastoma multiforme cells to ionizing radiation: cell cycle arrest, modulation of the expression of cyclin-dependent kinase inhibitors, and autophagy. J Neurosurg. 2003;98(2):378–84.PubMedCrossRef Yao KC, et al. Molecular response of human glioblastoma multiforme cells to ionizing radiation: cell cycle arrest, modulation of the expression of cyclin-dependent kinase inhibitors, and autophagy. J Neurosurg. 2003;98(2):378–84.PubMedCrossRef
266.
Zurück zum Zitat Opipari AW Jr, et al. Resveratrol-induced autophagocytosis in ovarian cancer cells. Cancer Res. 2004;64(2):696–703.PubMedCrossRef Opipari AW Jr, et al. Resveratrol-induced autophagocytosis in ovarian cancer cells. Cancer Res. 2004;64(2):696–703.PubMedCrossRef
267.
Zurück zum Zitat Sivaprasad U, Basu A. Inhibition of ERK attenuates autophagy and potentiates tumour necrosis factor-alpha-induced cell death in MCF-7 cells. J Cell Mol Med. 2008;12(4):1265–71.PubMedPubMedCentralCrossRef Sivaprasad U, Basu A. Inhibition of ERK attenuates autophagy and potentiates tumour necrosis factor-alpha-induced cell death in MCF-7 cells. J Cell Mol Med. 2008;12(4):1265–71.PubMedPubMedCentralCrossRef
268.
Zurück zum Zitat Li P, et al. Interferon-gamma induces autophagy with growth inhibition and cell death in human hepatocellular carcinoma (HCC) cells through interferon-regulatory factor-1 (IRF-1). Cancer Lett. 2012;314(2):213–22.PubMedCrossRef Li P, et al. Interferon-gamma induces autophagy with growth inhibition and cell death in human hepatocellular carcinoma (HCC) cells through interferon-regulatory factor-1 (IRF-1). Cancer Lett. 2012;314(2):213–22.PubMedCrossRef
269.
Zurück zum Zitat Ertmer A, et al. The anticancer drug imatinib induces cellular autophagy. Leukemia. 2007;21(5):936–42.PubMedCrossRef Ertmer A, et al. The anticancer drug imatinib induces cellular autophagy. Leukemia. 2007;21(5):936–42.PubMedCrossRef
270.
Zurück zum Zitat Takeuchi H, et al. Synergistic augmentation of rapamycin-induced autophagy in malignant glioma cells by phosphatidylinositol 3-kinase/protein kinase B inhibitors. Cancer Res. 2005;65(8):3336–46.PubMedCrossRef Takeuchi H, et al. Synergistic augmentation of rapamycin-induced autophagy in malignant glioma cells by phosphatidylinositol 3-kinase/protein kinase B inhibitors. Cancer Res. 2005;65(8):3336–46.PubMedCrossRef
272.
Zurück zum Zitat Scarlatti F, et al. Ceramide-mediated macroautophagy involves inhibition of protein kinase B and up-regulation of beclin 1. J Biol Chem. 2004;279(18):18384–91.PubMedCrossRef Scarlatti F, et al. Ceramide-mediated macroautophagy involves inhibition of protein kinase B and up-regulation of beclin 1. J Biol Chem. 2004;279(18):18384–91.PubMedCrossRef
273.
Zurück zum Zitat Kondo Y, et al. The role of autophagy in cancer development and response to therapy. Nat Rev Cancer. 2005;5(9):726–34.PubMedCrossRef Kondo Y, et al. The role of autophagy in cancer development and response to therapy. Nat Rev Cancer. 2005;5(9):726–34.PubMedCrossRef
Metadaten
Titel
Autophagy and autophagy-related proteins in cancer
verfasst von
Xiaohua Li
Shikun He
Binyun Ma
Publikationsdatum
01.12.2020
Verlag
BioMed Central
Erschienen in
Molecular Cancer / Ausgabe 1/2020
Elektronische ISSN: 1476-4598
DOI
https://doi.org/10.1186/s12943-020-1138-4

Weitere Artikel der Ausgabe 1/2020

Molecular Cancer 1/2020 Zur Ausgabe

Umsetzung der POMGAT-Leitlinie läuft

03.05.2024 DCK 2024 Kongressbericht

Seit November 2023 gibt es evidenzbasierte Empfehlungen zum perioperativen Management bei gastrointestinalen Tumoren (POMGAT) auf S3-Niveau. Vieles wird schon entsprechend der Empfehlungen durchgeführt. Wo es im Alltag noch hapert, zeigt eine Umfrage in einem Klinikverbund.

CUP-Syndrom: Künstliche Intelligenz kann Primärtumor finden

30.04.2024 Künstliche Intelligenz Nachrichten

Krebserkrankungen unbekannten Ursprungs (CUP) sind eine diagnostische Herausforderung. KI-Systeme können Pathologen dabei unterstützen, zytologische Bilder zu interpretieren, um den Primärtumor zu lokalisieren.

Sind Frauen die fähigeren Ärzte?

30.04.2024 Gendermedizin Nachrichten

Patienten, die von Ärztinnen behandelt werden, dürfen offenbar auf bessere Therapieergebnisse hoffen als Patienten von Ärzten. Besonders gilt das offenbar für weibliche Kranke, wie eine Studie zeigt.

Adjuvante Immuntherapie verlängert Leben bei RCC

25.04.2024 Nierenkarzinom Nachrichten

Nun gibt es auch Resultate zum Gesamtüberleben: Eine adjuvante Pembrolizumab-Therapie konnte in einer Phase-3-Studie das Leben von Menschen mit Nierenzellkarzinom deutlich verlängern. Die Sterberate war im Vergleich zu Placebo um 38% geringer.

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.