Autophagy is an evolutionarily conserved, catabolic process that involves the entrapment of cytoplasmic components within characteristic vesicles for their delivery to and degradation within lysosomes. Autophagy is regulated via a group of genes called AuTophaGy-related genes and is executed at basal levels in virtually all cells as a homeostatic mechanism for maintaining cellular integrity. The levels and cargos of autophagy can be modulated in response to a variety of intra- and extracellular cues to bring about specific and selective events. Autophagy is a multifaceted process and alterations in autophagic signalling pathways are frequently found in cancer and many other diseases. During tumour development and in cancer therapy, autophagy has paradoxically been reported to have roles in promoting both cell survival and cell death. In addition, autophagy has been reported to control other processes relevant to the aetiology of malignant disease, including oxidative stress, inflammation and both innate and acquired immunity. It is the aim of this review to describe the molecular basis and the signalling events that control autophagy in mammalian cells and to summarize the cellular functions that contribute to tumourigenesis when autophagy is perturbed.

Introduction

In 1963, de Duve introduced the term autophagy, which is derived from the Greek words ‘auto’ and ‘phagy’ meaning ‘self’ and ‘eating’, to describe the occurrence of distinct intracellular, membranous vesicles that contained degraded cytoplasmic material (1). Since then, autophagy has been characterized as an adaptive, catabolic process that serves to deliver cytoplasmic proteins and organelles to lysosomes for digestion. Depending on the route of delivery to the lysosome, three different types of autophagy are defined: microautophagy, chaperone-mediated autophagy and macroautophagy (2). This review solely focuses on macroautophagy, which is hereafter simply termed autophagy.

Autophagy is orchestrated by a number of highly conserved AuTophaGy-related genes (ATGs), which were originally identified in yeast, with many of these genes having orthologues in mammalian cells (3,4). In mammalian cells, double-membrane-bound autophagosomes develop in a multi-step process from a precursor structure called the phagophore or isolation membrane. Autophagosomes subsequently fuse with lysosomes to form a degradative, single-membrane-bound vesicle called an autolysosome. In a process named autophagic lysosome reformation, lysosomes are then re-derived from autolysosomes (5).

The role of autophagy extends beyond the general homeostatic removal, degradation and recycling of damaged proteins and organelles to many specific physiological and pathological processes such as development, immunity, energy homeostasis, cell death, tumourigenesis and many more (2). The involvement of autophagy in tumour development is unquestioned but is at present incompletely understood. Deregulation of autophagy is known to affect many processes that can control the formation and existence of a cancer cell, but paradoxes still exist in what we currently understand about the relationship between autophagy and cancer (4,6). This review aims therefore to describe the molecular control of autophagy and to delineate how the deregulation of autophagy can contribute in context-specific ways to the development of cancer.

The molecular basis of autophagic vesicle formation

Autophagy is activated in response to a whole host of stimuli including nutrient depletion, hypoxia and activated oncogenes. The majority of pro-autophagic events converge on the serine/threonine protein kinase mTOR (MTOR, mammalian/mechanistic target of rapamycin) (79). Another important, nutrient-sensitive entry route to ATG signalling is the class III phosphatidylinositol 3-kinase complex (PI3K-III) consisting of hVps34 (PIK3C3, the orthologue of yeast Vps34), Beclin 1 (yeast Atg6) and p150/hVps35 (PIK3R4; yeast Vps15) (9,10). Many of the aforementioned ATGs are restricted to a certain type of autophagy (see above). The ‘core’ autophagic machinery encompasses only those ATGs that are necessary for autophagosome formation in all subtypes and these can be divided into several distinct groups: (i) the unc-51-like kinase 1/2 (ULK1/2) complex (ii) the multi-spanning membrane protein Atg9, (iii) the PI3K-III complex and (iv) the ubiquitin-like ATG12 and microtubule-associated protein 1 light chain 3 alpha (MAP1LC3A) conjugation systems. The following sections detail how these proteins regulate the various stages of the autophagy process.

The autophagy machinery.

Initiation and nucleation are terms used to describe the events that lead to the formation of the initial autophagic structure: the phagophore or isolation membrane. RAPTOR forms the catalytic subunit of two different protein complexes: mTORC1 and mTORC2. The former contains mTOR and RAPTOR (regulatory-associated protein of MTOR), whereas mTORC2 contains among others, mTOR and RICTOR (rapamycin-insensitive companion of MTOR) (11). In nutrient-rich states, mTORC1 but not mTORC2 forms a complex with ULK1/2 (orthologues of yeast Atg1), mAtg13, FIP200 (RB1CC1; mammalian orthologue of Atg17) and the newly identified ATG101, as a result of interaction between RAPTOR and ULK1 (8,1214). mTOR phosphorylates ULK1 and Atg13 and thereby keeps the kinase activity of ULK1 in check. Upon treatment with rapamycin or in fasting conditions, mTORC1 breaks free from the ULK complex and the inhibitory phosphorylation of ULK1 is lost. ULK1 then autophosphorylates and activates Atg13 and FIP200. The activated ULK complex localizes to the developing phagophore. The relationship between mTOR and activation of the ULK complex is shown in Figure 1. Startlingly, the inverse relationship between mTOR activity and autophagy is not universal, as autophagy induced by 6-thioguanine has been reported to require activation and not inhibition of mTOR (15).

Autophagic core machinery. The ULK kinase complex, the PI3K-III complex, mAtg9 and the two ubiquitination systems are indispensable for autophagy. Members of the core machinery are shown in coloured boxes. Modulators that are not part of the core machinery are shown in white boxes. For details, see text.
Fig. 1.

Autophagic core machinery. The ULK kinase complex, the PI3K-III complex, mAtg9 and the two ubiquitination systems are indispensable for autophagy. Members of the core machinery are shown in coloured boxes. Modulators that are not part of the core machinery are shown in white boxes. For details, see text.

There are two mammalian orthologues of Atg9: ATG9L1 (mAtg9) is ubiquitously expressed, whereas expression of ATG9L2 is restricted to the placenta and pituitary gland. The exact function of mAtg9 currently remains elusive but it is required for LC3 lipidation and knockout mice die after birth as do Atg5- and Atg7-knockout animals (1618).

Phosphatidylinositol 3-phosphate (PtdIns(3)P) is a prerequisite for the nucleation process. It is produced by the PI3K-III-hVps34 complex when bound to its core partners Beclin 1 and p150/hVps35 (10). The complex is found on the phagophore and thought to facilitate recruitment of other ATGs to the developing vesicle. Importantly, autophagy can be positively and negatively modulated at the level of the Beclin 1–hVps34–p150/hVps35 complex depending on additional, regulatory binding partners of Beclin 1 (Figure 1) (4,10). ATG14/BARKOR (Beclin 1-associated autophagy-related key regulator), UVRAG (protein product of the ultraviolet radiation resistance gene) and activating molecule in Beclin 1-regulated autophagy are pro-autophagic regulators of the PI3K-III complex. Simultaneous binding of both UVRAG and RUBICON (RUN domain and cysteine-rich domain containing) inhibits the autophagy-promoting activity of the complex (1923).

Elongation and closure describe the development of the characteristic double-membrane-bound autophagosome from its precursor structure and require two ubiquitin-like conjugation systems. The ubiquitin-like ATG12 is conjugated to ATG5 via the E1-like protein ATG7 and the E2-like ATG10. ATG16 then enters the complex and directs the large (L), newly formed ATG16L (ATG12-ATG5-ATG16) complex to the isolation membrane. The Atg16L complex is required for autophagosome formation, guides LC3 to the phagophore and promotes lipidation of LC3 (Figure 1) (2426).

The ubiquitin-like yeast protein Atg8 has several orthologues in mammalian cells: MAP1LC3 (LC3), GABARAPL2 (GATE16), GABARAP and GABARAPL1 (ATG8L). LC3 is the most thoroughly investigated of these proteins and its modification during autophagy is exploited as a marker for autophagy (27,28). Newly synthesized LC3 is immediately cleaved at its C-terminal end by the protease Atg4 into the cytoplasmic form LC3-I. If autophagy is active, LC3-I is then conjugated to phosphatidylethanolamine via ATG7 and the E2-like ATG3 (29). In its conjugated form, LC3 is called LC3-II and is recruited via its lipid moiety to the inner and outer surfaces of the autophagosomal membrane, i.e. unlike LC3-I, LC3-II is not freely dispersed in the cytoplasm (Figure 1).

The exact order of Atg activation is not clear and there is intensive crosstalk between the different Atg systems. However, it is generally accepted that the ULK1 kinase complex and the PI3K-III complex act upstream of the ubiquitination systems (30).

The maturation process encompasses the fusion of autophagosomes with lysosomes to form autolysosomes. Autolysosomes are single-membrane-bound, acidic vesicles comprised of the outer membrane of autophagosomes and the lysosome that degrade the autophagosomal cargo via acidic hydrolases provided by the lysosome. The process is less well understood but involves the action of lysosomal proteins, such as lysosomal-associated membrane protein 1/2 (LAMP 1/2) and also again, Beclin 1 (3,4).

Work by Yu et al. (5) has recently shed light on the ultimate fate of autolysosomes. During autophagy-initiation mTOR is inhibited but becomes reactivated at later stages as a result of the release of cellular constituents into the cytoplasm following the breakdown of macromolecules within autolysosomes. Increased mTOR activity then inhibits autophagy and leads to the formation of proto-lysosomal extensions (LAMP1+, LC3−) from autolysosomes (LAMP1+, LC3+) (30). Ultimately, these proto-lysosomal extensions detach from the autolysosome and mature into functional lysosomes. Inhibition of mTOR, or (auto-)lysosomal function, prevents autophagic lysosome reformation (Figure 2). Autophagy is therefore controlled by a negative feedback mechanism that is regulated by mTOR (5,30).

 Autophagic vesicle generation and recycling. The first steps of autophagosome formation are initiation and nucleation. The earliest detectable autophagic structure is the double-membrane-bound phagophore/isolation membrane that evolves from the ER, mitochondria or the plasma membrane following activation of the ULK1 and Beclin 1 complexes (initiation/nucleation). Subsequently, the ATG16L complex, LC3-II and mAtg9 are recruited to the developing isolation membrane. The membraneous structure evolves (elongation) and encapsulates marcomolecules to become the closed hallmark structure of autophagy, the autophagosome. After fusion with a lysosome (maturation), the intra-vesicular constituents of the autophagosome get degraded and released into the cytosol, thereby creating a local rise in nutrient availability. This leads to reactivation of mTOR and regeneration of a mature lysosome from autolysosomes in a process called autophagic lysosome regeneration. Members of the core autophagic machinery that are involved in each step and can be found on the corresponding structure/vesicle are shown in coloured boxes. For details, see text.
Fig. 2.

Autophagic vesicle generation and recycling. The first steps of autophagosome formation are initiation and nucleation. The earliest detectable autophagic structure is the double-membrane-bound phagophore/isolation membrane that evolves from the ER, mitochondria or the plasma membrane following activation of the ULK1 and Beclin 1 complexes (initiation/nucleation). Subsequently, the ATG16L complex, LC3-II and mAtg9 are recruited to the developing isolation membrane. The membraneous structure evolves (elongation) and encapsulates marcomolecules to become the closed hallmark structure of autophagy, the autophagosome. After fusion with a lysosome (maturation), the intra-vesicular constituents of the autophagosome get degraded and released into the cytosol, thereby creating a local rise in nutrient availability. This leads to reactivation of mTOR and regeneration of a mature lysosome from autolysosomes in a process called autophagic lysosome regeneration. Members of the core autophagic machinery that are involved in each step and can be found on the corresponding structure/vesicle are shown in coloured boxes. For details, see text.

Until recently, it was believed that the two ubiquitination systems are indispensible for autophagy. However, Nishida et al. (31) introduced the term ‘alternative macroautophagy’ to describe a degradative process in response to starvation and etoposide treatment that involves autophagosome-like structures that are not decorated by LC3-II. Strikingly, this process is independent of both ATG5 and ATG7 but critically relies on ULK1 and Beclin 1. Double-membrane-bound vesicles that included cytoplasmic material were generated in a RAB9 (RAB9A, member RAS oncogene family)-dependent fashion by the fusion of isolation membranes and vesicles derived from the trans-Golgi and late endosomes (31). Since this process occurs without involvement of crucial regulators for ‘conventional’ or ‘canonical’ autophagy, it is debated whether this phenomenon is something altogether different from autophagy (32).

Origin of the phagophore/isolation membrane.

The first detectable structure during autophagy in mammalian cells is the isolation membrane or phagophore. Considerable insight has been gained in the last 2 years in relation to its sites of origin. Current consensus favours that in mammalian cells, the isolation membrane develops from at least three different, preformed sources: the endoplasmic reticulum (ER), the plasma membrane and mitochondria (3336). Axe et al. (33) proposed that the phagophore is derived from so-called omegasomes (cup-shaped protrusions from the ER). Moreover, it has recently been confirmed that isolation membranes are physically connected to the ER and are cradled by two ER membranes, which is reminiscent of the omegasome (37,38). Upon starvation, the hVps34 kinase is recruited via ATG14L to the ER, where it creates a local increase in PtdIns(3)P, that is essential for autophagosome development (39). Proteins that specifically recognize PtdIns(3)P are then recruited to the omegasome/cradle, WD repeat domain, phosphoinositide interacting 2 and ZFYVE1, zinc finger, FYVE domain containing 1, the latter of which can be used to pinpoint the location of the omegasome/cradle (40,41). The phagophore extends from the PtdIns(3)P-rich region and is cradled by two ER membranes (42). Small sections of ER are encapsulated within autophagosomes by this mechanism. The ULK complex and LC3 localize to the omegasome, as well as the ATG16L complex via ATG16 (42) (Figure 2).

ATG16L1 has also been reported to be associated with the plasma membrane (36). This association was mediated by an interaction between ATG16L1 and the heavy chain of clathrin and it is believed that this interaction is required for the formation of early autophagosome precursors (Figure 2). Inhibition of clathrin-mediated internalization reduces the formation of these pre-autophagosomal structures as well as mature autophagosomes (36). It was proposed by the authors of this study that due to the size of the plasma membrane, this source of autophagosomes may be particularly important during intense autophagic activity (36). A switch may therefore occur from sources of membrane utilized under basal conditions to the plasma membrane under stressed conditions in order to perhaps maintain intracellular organelle integrity.

Mitochondria have also recently been proposed as an alternative route of phagophore generation (34). Under starving conditions, ATG5 and LC3 localize to the outer membrane of mitochondria, which serves as a cornerstone for phagophore development. Mitofusin 2 connects mitochondria to the ER and thereby enables transfer of phosphatidylserine from the ER to mitochondria, which seems to be essential for autophagosome generation. In mitochondria, phosphatidylserine then gets processed to phosphatidylethanolamine, which becomes an essential component of the developing autophagosome as described previously (29). Figure 2 illustrates the development and recycling of autolysosomes from early precursors and the involvement of the core autophagic machinery in each step.

It seems appropriate to underline that the proposed models of autophagosome generation are not mutually exclusive and probably coexist. It is possible that depending on cellular context and activating triggers, one or all routes are initiated. However, in mammalian cells, each model points away from the assembly model (where phagophores develop de novo), unlike in yeast where the phagophore develops from a phagophore assembly site (43).

Autophagy and cancer

Autophagy is known to protect us against various forms of human disease (44). In some cases, for example neurodegenerative disease, it is clear that autophagy facilitates the removal of aggregate-prone proteins that lead to Huntington’s and Parkinson’s disease (45). The role of autophagy in cancer, however, is more complex. There is evidence that autophagy may be oncogenic in some contexts, whereas in others, it clearly contributes to tumour suppression (4,6,46,47). In the following sections, we detail the cellular functions, such as inflammation, cell death and immunology that may contribute to tumourigenesis when autophagy is impaired and we discuss situations where dichotomies and paradoxes exist in our understanding of the role of autophagy in both the genesis and treatment of human cancer. Numerous studies have described the effects of various specific oncogenes and tumour suppressor genes on the rate of autophagy, but due to the length of this review, these will not be discussed here. For summaries of these studies, we refer the reader to the following excellent and extensive recent reviews (4,48).

Links of autophagy to cancer: cell survival, quality control, inflammation and oxidative stress.

Cancer is caused by the successive acquisition of mutations and epigenetic changes that override failsafe mechanisms such as cell death and cell cycle arrest, which normally prevent tumour development (49,50). The link between autophagy and cancer is broad based. Several studies using genetically engineered mice have established a link between autophagy and cancer development (5153). For example, hemizygosity of Becn1 (the gene encoding Beclin 1) or complete loss of the gene encoding the UVRAG-binding protein BIF-1 (Bax-interacting protein-1, also known as Endophilin B) results in tumour susceptibility in mice (51,52,54). In addition, the ectopic expression of Beclin 1 or UVRAG have both been shown to repress the growth of human cancer cell xenografts (23,55). In line with these studies, many autophagy genes have also been found to be inactivated in human cancers (5559). Furthermore, both chemotherapy and radiation treatments for cancer have also been reported to modulate autophagy (60).

Autophagy has an evolutionarily conserved role in buffering metabolic stress caused by limited nutrients or oxygen. During tumour growth, periods of poor vascularization lead to constraints on nutrient availability and hypoxic regions within tumours (61). Since autophagy is known to be activated in these regions and could be utilized to keep these tumour cells alive, it seems paradoxical therefore how inactivation of a process that could keep tumour cells alive could benefit tumour development. The answer to this apparently conflicting issue simply lies in the basal function of autophagy: the alleviation of stress in order to preserve cellular integrity (46). Insufficient autophagy can no longer provide nutrients, prevent the accumulation of defective proteins and organelles, manage oxidative stress and limit inflammation. As a result intra- and extracellular events occur that favour malignant transformation.

Autophagy-dependent modulation of cell death and senescence is critical for tumourigenesis.

Perhaps, the largest area in which dichotomies exist in relation to autophagy in cancer is in the control of cell death. For sometime, it has been heavily debated whether autophagy can actually be considered an independent form of programmed cell death, the so-called type II cell death (62). The Nomenclature Committee of Cell Death 2009 points out that the latter term ‘may misleadingly suggest a form of death occurring through autophagy, as this process often promotes cell survival’ (63). The committee took a morphological approach to more clearly define the observations of many laboratories and proposed the broader description of ‘cell death occurring with autophagy’ rather than ‘autophagic cell death’ (63). In fact, it is often the case as outlined above that autophagy is activated to keep cells alive. In many cases, this effect on cell survival would enhance tumour cell viability and would be tumour promoting, whereas in other cases, the death signals are maybe too strong for the activated autophagy to counter this effect and the tumour cell therefore dies—leading to tumour suppression. Critically therefore, despite being present in these cell death scenarios, autophagy does not constitute a positive contributing factor towards cellular demise.

In contrast to its role in cell survival, several studies in Drosophila, Dictyostelium discoideum and Caenorhabditis elegans have, however, implicated autophagy as an executor of cell death and not just a mere co-phenomenon (6468). To the best of our knowledge, convincing in vivo evidence that autophagy alone can execute cell death in mammalian cells is lacking, even though this possibility is proposed in several cell culture-based systems (69,70). One view that supports a role for autophagy in promoting cell death dictates that autophagy is not an executor of cell death per se but that it is a required process in certain settings in combination with other pro-death signals. One important example that may be relevant to cancer is DRAM1 (damage-regulated autophagy modulator 1) (71,72). DRAM1 belongs to the recently described DRAM family and was identified due to the fact that it is activated by DNA damage and the tumour suppressor p53 (7173). DRAM1 is a positive modulator of autophagy and is required for the full execution of p53-induced death. When expressed alone, however, DRAM1 can modulate autophagy but does not cause cell death, indicating that autophagy is required but is not sufficient to bring about this response (71,72).

In addition to the relationship between autophagy and apoptosis, it is important to note that in many scenarios, autophagy serves to protect against necrotic cell death with possible detrimental consequences as outlined in subsequent sections (46). Autophagy also prevents anoikis, cell death that occurs after cell detachment from the extracellular matrix (46,74). Consequently, autophagy may promote survival of cells that leave their physiological context and could therefore represent a mechanism that supports metastasis. Taken together, autophagy has conflicting roles in the regulation of cell death that may have both positive and negative effects on tumour development.

It is lastly also important to mention the role of autophagy in senescence. Although not a cell death per se, it is certainly an important end point in tumour suppression. Senescent cells are marked by sustained cell arrest and the expression and release of secretory cytokines that attract the immune system (75). Young et al. (76) showed that autophagy occurs during senescence and shapes the senescence-associated secretory phenotype. Impaired autophagy delayed cytokine production and thereby might modulate the clearance of senescent cells by the immune system.

Autophagy is important for protein and organelle quality control to prevent cell damage.

Autophagy removes injured organelles, misfolded and aggregated proteins. If damaged proteins persist, they are akin to a non-inheritable mutation and are a source of increased oxidative stress, both of which can have detrimental effects on cells. Stressed organelles such as the ER also force the cell to execute adaptive programmes that allow for survival and ultimately promote tumour growth. Autophagy can therefore be considered to have dual roles with respect to the management of protein quality control. On the one hand, the removal of damaged protein and organelles may promote the survival of tumour cells, whereas on the other hand, the accumulation of damaged cellular constituents may result in the production of a hostile cellular environment that may ultimately be tumour promoting. In this regard, ER stress and the unfolded protein response are found in, and are required for, a large number of tumours (77). Impaired autophagy is unable to relieve ER stress and the resulting unfolded protein response. Therefore, altered signalling persists and potentially causes a reprogramming of the cell, including nuclear factor-kappaB activation. Kongara et al. (78) also recently found that defective autophagy itself induces ER stress in mammary cells, which might contribute to the development of breast cancer in a manner independent of genotoxic stress and genomic instability. They also underlined the impact of autophagy on the role of the chaperone protein Sequestome 1/p62 in cancer (see below) because autophagy-mediated keratin homeostasis is impaired when p62 levels are altered and keratin is found to be overexpressed in breast cancer (78).

A-1 Antitrypsin deficiency causes chronic liver disease and illustrates how insufficient aggregate and organelle removal might lead to cancer (79). The disease is characterized by the occurrence of intra-hepatocytic globules that represent large aggregates of a mutant protein called Alpha-1-antitrypsin (AT) with substitution of lysine for glutamate at residue 342 within the ER. In affected livers, autophagy is significantly upregulated and responsible for the clearance of aggregates and damaged mitochondria that are injured from the stressed ER. Possibly, ER accumulation of Alpha-1-antitrypsin (AT) with substitution of lysine for glutamate at residue 342 induces the regulator of G signalling 16, which then inhibits the G protein Gαi3 and thereby alleviates Gαi3 inhibitory function on autophagy (79). It is believed that globule-containing cells transmit proliferative signals to globule-devoid cells that can then grow into adenomas and carcinomas upon chronic exposure. Furthermore, globule-containing cells are less proliferative and more resistant to apoptosis (79). Impaired autophagy would no longer be able to clear the aggregates and thus increase the transmission signals of globule-containing cells. This elevates oxidative damage resulting from injured mitochondria and increases the pressure to proliferate on globule-devoid cells.

Impaired autophagy shapes the inflammatory and immune responses to assist tumour development.

Chronic inflammation is thought to be a risk factor for the development of cancer and many tumours present with an inflammatory component (80). It is believed that autophagy partly impacts on cancer development through its ability to shape the inflammatory reaction. Orderly removal of dying cells is a vital function of the organism to prevent undesirable oxidative stress and an inflammatory response that would be triggered from secondary necrosis. Impaired autophagy causes metabolically stressed and apoptosis-deficient cells to undergo necrotic cell death (46,61). The concomitant release of pro-inflammatory cytokines thereby creates a pro-proliferative environment for tumour cells (46,61).

Inflammatory bowel disease and pancreatitis are potentially examples how an underlying defect in autophagy initiates inflammation and thereby creates a pro-tumourigenic environment. Impaired autophagy is found in chronic inflammation of the pancreas and the intestine, both of which are risk factors for the development of cancer (81). Crohn’s disease (CD) is an inflammatory bowel disease that usually causes transmural inflammation of the terminal ileum (small intestine) but can affect the whole gastrointestinal tract (82). Mice that are either hypomorphic for Atg16l1 or deficient in either Atg5 or Atg7 in the intestine display severe cellular abnormalities that are confined to intestinal Paneth cells and resemble the changes seen in CD patients that carry the ATG16L1 risk allele (83,84). Paneth cells have elevated levels of genes involved in peroxisome proliferator-activated receptor pathways and lipid metabolism (8385). ATG16L1-deficient macrophages from chimeric mice (lethally irradiated mice with haematopoietic reconstitution from ATG16L1-deficient foetal liver cells) produce drastically elevated levels of the inflammatory cytokines interleukin-1β and-18 (86). It is tempting to speculate for a role of autophagy in the development of at least a subset of intestinal carcinomas because ulcerative colitis, another inflammatory bowel disease, is clearly recognized as a precancerous lesion, whereas in the case of CD, that relationship is probable but less clear (8789).

Development of acute pancreatitis has also been attributed to retarded autophagy that results from a dysfunction of lysosomal cathepsins (90). Increased conversion of trypsinogen to trypsin by Cathepsin B coincides with a decreased action of Cathepsin L that degrades trypsinogen and trypsin. As a result, intra-acinar trypsin accumulates, digests surrounding tissue and thereby elicits an inflammatory response (90). Acute pancreatitis precedes chronic pancreatitis, which is a clear risk factor for the development of pancreatic carcinoma. CD and pancreatitis might therefore be examples of how hampered autophagy induces inflammation that does not largely result from necrotic cell death but still contributes to tumour development.

Autophagy modulates tumour immunology.

Autophagy is important for the balanced execution of the innate and acquired immune response. Inhibition of autophagy via the PI3K-III inhibitor 3-methyladenine or knockdown of Beclin 1 or ATG12 drastically reduced the ability of HEK293T (human embryonic kidney cells) and melanoma cells for presentation of a model antigen or the endogenous tumour antigen gp100, whereas activation of autophagy had the opposite effect (91). Defective autophagy therefore potentially promotes tumourigenesis and inflammation by constraining immune recognition of tumour antigens.

T cells participate in inflammatory reactions and shape the response to tumour antigens (92). Autophagy-deficient T cells frequently undergo apoptosis and are less proliferative than autophagy-proficient cells (93,94). A causative explanation is that they cannot efficiently clear damaged mitochondria and thus have increased reactive oxygen species (ROS) levels and altered protein expression that favours pro-apoptotic genes (95,96).

Major histocompatibility complex class II molecules present antigenic, exogenous proteins on the cell surface of antigen-presenting cells and thereby activate CD4+ T-cells. Thymic epithelial cells require autophagy for proper selection of major histocompatibility complex-II-restricted antigen recognition i.e. to be able to separate host from foreign antigens. As a result, impaired autophagy leads to the production of T cells that recognize host proteins and illicit a strong inflammatory response (97). Epstein–Barr virus is found in a variety of human tumours, especially lymphoproliferative disorders. Autophagy-deficient antigen-presenting cells cannot process certain nuclear viral oncogenes (Epstein–Barr virus-coded nuclear antigen-1) via the major histocompatibility complex-II pathway, which are therefore not present on the cell surface and consequently evade an immune response (98101).

Oxidative stress is a crucial component of tumourigenesis in stressed autophagy-deficient cells.

The one common denominator of impaired autophagy in nearly all situations is the creation of oxidative stress. Oxidative stress results from a disparity between production and elimination of free radicals and reactive metabolites, so-called ROS. The mitochondrial respiratory chain is the main source of intracellular ROS. ROS impact on all stages of tumour formation: initiation, promotion and progression (81). Notably, physiological levels of ROS are required for many cell signalling events. Physiological concentrations of ROS are also critical for autophagy. H2O2 oxidizes and thereby inhibits Atg4, which leads to increased availability of lipidated LC3 during starvation (102). Cells also use a selective form of autophagy, mitophagy, to remove damaged mitochondria as a source of aberrant ROS production. Two molecular routes of mitophagy have been suggested. phosphatase and tensin homolog-induced kinase 1 recruits the E3 ubiquitin-like ligase PARKIN to compromised mitochondria, which results in ubiquitination of voltage-dependent anion channel 1 on the mitochondrial membrane and recruitment of p62 (103105). P62 may through its LC3-interacting region steer damaged mitochondria to autophagosomes. Alternatively, mitochondrial BNIP3L (BCL2/adenovirus E1B 19kDa interacting protein 3-like (aliases: Nix, BNIP3a)) can via its LC3-interacting region interact with Atg8 homologues and deliver mitochondria to autophagosomes (106,107). Furthermore, p62 has also been implicated in the delivery of oxidized proteins for autophagic degradation. Work from Eileen White’s group has underlined that management of oxidative stress is probably the key element how autophagy deficiency promotes cancer (46,61,108110). It convincingly explains how the loss of a survival mechanism benefits cancer or in other words it explains why autophagy-impaired tumour cells are both more susceptible to cell death while at the same time having enhanced tumourigenicity. Figure 3 summarizes how different cellular functions contribute to tumour formation/progression when autophagy is impaired.

 Cellular and organismal functions that contribute to tumour development when autophagy is impaired. Shown is the impact of autophagy impairment on crucial cellular processes and how their alteration contributes to tumourigenesis. Importantly, oxidative stress is a recurring phenomenon in nearly all autophagy-impaired settings. For details, see text.
Fig. 3.

Cellular and organismal functions that contribute to tumour development when autophagy is impaired. Shown is the impact of autophagy impairment on crucial cellular processes and how their alteration contributes to tumourigenesis. Importantly, oxidative stress is a recurring phenomenon in nearly all autophagy-impaired settings. For details, see text.

P62 critically links impaired autophagy, oxidative stress and tumourigenesis.

Degenhardt et al. (61) showed that autophagy is upregulated in hypoxic tumour regions and that autophagy deficiency indeed promotes cell demise but also necrotic cell death. The resulting, uncontrolled cytokine release creates an inflammatory response and attracts tumour-promoting macrophages. Genomic stress that leads to DNA damage and an increased propensity to acquire growth-promoting mutations are cell intrinsic consequences of insufficient autophagy (109,110). P62 is overexpressed in a large number of tumours (111). Sequestome-1/p62 is a multi-domain adapter protein that has emerged as a key player for autophagy-dependent quality control mechanisms (112). Mice that are deficient for Atg7 accumulate poly-ubiquitinated aggregates in the liver and brain that co-localize with p62 and cause cellular damage (113115). Mice lacking both p62 and Atg7 are free of aggregates, indicating that p62 is required for aggregate formation in autophagy-deficient states and that autophagy is responsible for the clearance of these aggregates (111,116). In a landmark study, Mathew et al. (108) showed that p62 is critically required for tumourigenesis, and depletion of p62 by autophagy suppressed tumour development. Autophagy-deficient cells are unable to clear p62 aggregates that arise as a result of metabolic stress. Increased p62 then fuels a detrimental positive feedback loop by which p62 itself leads to ROS production, enhanced induction of the protein folding machinery in the ER and a DNA damage response (108). Suppressing ROS or p62 accumulation alleviated cellular damage resulting from impaired autophagy. Strikingly, sustained expression of p62 also altered nuclear factor-kappaB signalling and thereby initiated additional molecular pathways that favour tumour formation (108). These data convincingly imply that the consequences of oxidative DNA damage and increased tumourigenesis of autophagy-deficient cells are related to insufficient clearance of p62 (Figure 4).

 Consequences of impaired management of oxidative stress in autophagy-deficient cells. Impaired autophagy results in reduced clearance of p62 and damaged organelles, both of which can fuel ROS levels and thereby aggravate oxidative stress. Furthermore, p62 aggregation alters Nf-κB signalling to favour pro-survival signalling and therefore tumourigenic events. This vicious circle of increased ROS production and decreased clearance of ROS producers and p62 aggregates leads to genetic instability and an increased propensity to mutate DNA. As a result of overburdened ROS levels and DNA damage, the cells either die by necrosis and produce a pro-tumourigenic inflammatory response or a subset of cells acquire growth-promoting mutations. For details, see text.
Fig. 4.

Consequences of impaired management of oxidative stress in autophagy-deficient cells. Impaired autophagy results in reduced clearance of p62 and damaged organelles, both of which can fuel ROS levels and thereby aggravate oxidative stress. Furthermore, p62 aggregation alters Nf-κB signalling to favour pro-survival signalling and therefore tumourigenic events. This vicious circle of increased ROS production and decreased clearance of ROS producers and p62 aggregates leads to genetic instability and an increased propensity to mutate DNA. As a result of overburdened ROS levels and DNA damage, the cells either die by necrosis and produce a pro-tumourigenic inflammatory response or a subset of cells acquire growth-promoting mutations. For details, see text.

Summary and therapeutic perspectives

Since it is clear that autophagy has roles in tumour development, it is natural to speculate if targeting autophagy is a realistic prospect for cancer therapy. As evident from this review, one immediate problem relates to whether we can selectively target the oncogenic role played by autophagy in keeping stressed tumour cells alive, while not inhibiting its role in tumour suppression. It may well be the case that autophagy is oncogenic at certain stages of tumour development and tumour suppressive at other stages and analysis of the role played by autophagy at the different stages of cancer in mouse models will certainly provide insight into this issue. The issue, however, may be even more complicated by the fact that the oncogenic and tumour-suppressive effects of autophagy may coexist not just within one patient but even within any individual tumour. It must not be forgotten too that autophagy has many beneficial roles in our normal tissues and in an ideal world it would be best if autophagy in these contexts was not affected by a systemic therapy aimed at targeting malignant disease. We consider therefore that the identification of cellular signalling pathways, which selectively regulate autophagy in response to specific stimuli, may well hold the key to the selective targeting of autophagy in human disease. Work in our own laboratory has sought to address this point with respect to cancer by searching for signalling pathways that selectively regulate autophagy in response to hypoxia—a state common in many solid tumours, but for the most part absent in normal tissue. We found that autocrine platelet-derived growth factor family signalling—an event common in cancer—was critical for cytoprotective autophagy induced by hypoxia but was, in contrast, seemingly dispensable for autophagy induced by other stimuli (117,118). We feel that our findings, therefore, act as a proof of principle paradigm that disease-associated autophagy could be selectively targeted for therapeutic gain. Ultimately, due to the intense interest in the role of autophagy in human cancer, it is reasonable to be optimistic that new studies will lead to important insights into the significance of autophagy in tumour development that will also make targeting autophagy for cancer therapy a realistic and realizable goal.

Abbreviations:

     
  • ATG

    AuTophaGy-related genes

  •  
  • CD

    Crohn’s disease

  •  
  • DRAM1

    damage-regulated autophagy modulator 1

  •  
  • ER

    endoplasmatic reticulum

  •  
  • LAMP 1/2

    lysosomal-associated membrane protein 1/2

  •  
  • MAP1LC3A

    microtubule-associated protein 1 light chain 3 alpha

  •  
  • MTOR

    mammalian/mechanistic target of rapamycin

  •  
  • PI3K-III

    class III phosphatidylinositol 3-kinase complex

  •  
  • PtdIns(3)P

    phosphatidylinositol 3-phosphate

  •  
  • ROS

    reactive oxygen species

  •  
  • ULK1/2

    unc-51-like kinase 1/2

  •  
  • UVRAG

    ultraviolet radiation resistance gene

We apologize to researchers whose studies on autophagy we were unable to cite due to the length of this review. We would like to thank members of the Tumour Cell Death Laboratory for critical reading of the manuscript.

Funding

Work in the Tumour Cell Death Laboratory is supported by Cancer Research UK and the Association for International Cancer Research

Conflict of Interest Statement: None declared.

1.

Yang
Z
et al.
Eaten alive: a history of macroautophagy
Nat. Cell Biol.
2010
12
814
822

2.

Mizushima
N
et al.
Autophagy fights disease through cellular self-digestion
Nature.
2008
451
1069
1075

3.

Reggiori
F
et al.
Autophagosomes: biogenesis from scratch?
Curr. Opin. Cell Biol.
2005
17
415
422

4.

Rosenfeldt
MT
et al.
The role of autophagy in tumour development and cancer therapy
Expert Rev. Mol. Med.
2009
11
e36

5.

Yu
L
et al.
Termination of autophagy and reformation of lysosomes regulated by mTOR
Nature.
2010
465
942
946

6.

Levine
B
Cell biology: autophagy and cancer
Nature.
2007
446
745
747

7.

Ganley
IG
et al.
ULK1.ATG13.FIP200 complex mediates mTOR signaling and is essential for autophagy
J. Biol. Chem.
2009
284
12297
12305

8.

Jung
CH
et al.
ULK-Atg13-FIP200 complexes mediate mTOR signaling to the autophagy machinery
Mol. Biol. Cell.
2009
20
1992
2003

9.

Mehrpour
M
et al.
Overview of macroautophagy regulation in mammalian cells
Cell Res.
2010
20
748
762

10.

Funderburk
SF
et al.
The Beclin 1-VPS34 complex—at the crossroads of autophagy and beyond
Trends Cell Biol.
2010
20
355
362

11.

Laplante
M
et al.
mTOR signaling at a glance
J. Cell Sci.
2009
122
3589
3594

12.

Hosokawa
N
et al.
Nutrient-dependent mTORC1 association with the ULK1-Atg13-FIP200 complex required for autophagy
Mol. Biol. Cell.
2009
20
1981
1991

13.

Mercer
CA
et al.
A novel, human Atg13 binding protein, Atg101, interacts with ULK1 and is essential for macroautophagy
Autophagy.
2009
5
649
662

14.

Hosokawa
N
et al.
Atg101, a novel mammalian autophagy protein interacting with Atg13
Autophagy.
2009
5
973
979

15.

Zeng
X
et al.
Mammalian target of rapamycin and S6 kinase 1 positively regulate 6-thioguanine-induced autophagy
Cancer Res.
2008
68
2384
2390

16.

Webber
JL
et al.
New insights into the function of Atg9
FEBS Lett.
2010
584
1319
1326

17.

Kuma
A
et al.
The role of autophagy during the early neonatal starvation period
Nature.
2004
432
1032
1036

18.

Komatsu
M
et al.
Impairment of starvation-induced and constitutive autophagy in Atg7-deficient mice
J. Cell Biol.
2005
169
425
434

19.

Itakura
E
et al.
Beclin 1 forms two distinct phosphatidylinositol 3-kinase complexes with mammalian Atg14 and UVRAG
Mol. Biol. Cell.
2008
19
5360
5372

20.

Sun
Q
et al.
Identification of Barkor as a mammalian autophagy-specific factor for Beclin 1 and class III phosphatidylinositol 3-kinase
Proc. Natl Acad. Sci. USA.
2008
105
19211
19216

21.

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
468
476

22.

Fimia
GM
et al.
Ambra1 regulates autophagy and development of the nervous system
Nature.
2007
447
1121
1125

23.

Liang
C
et al.
Autophagic and tumour suppressor activity of a novel Beclin1-binding protein UVRAG
Nat. Cell Biol.
2006
8
688
699

24.

Fujita
N
et al.
The Atg16L complex specifies the site of LC3 lipidation for membrane biogenesis in autophagy
Mol. Biol. Cell.
2008
19
2092
2100

25.

Hanada
T
et al.
The Atg12-Atg5 conjugate has a novel E3-like activity for protein lipidation in autophagy
J. Biol. Chem.
2007
282
37298
37302

26.

Sou
YS
et al.
The Atg8 conjugation system is indispensable for proper development of autophagic isolation membranes in mice
Mol. Biol. Cell.
2008
19
4762
4775

27.

Klionsky
DJ
et al.
Guidelines for the use and interpretation of assays for monitoring autophagy in higher eukaryotes
Autophagy.
2008
4
151
175

28.

Kabeya
Y
et al.
LC3, a mammalian homologue of yeast Apg8p, is localized in autophagosome membranes after processing
EMBO J.
2000
19
5720
5728

29.

Kabeya
Y
et al.
LC3, GABARAP and GATE16 localize to autophagosomal membrane depending on form-II formation
J. Cell Sci.
2004
117
2805
2812

30.

Simonsen
A
et al.
Coordination of membrane events during autophagy by multiple class III PI3-kinase complexes
J. Cell Biol.
2009
186
773
782

31.

Nishida
Y
et al.
Discovery of Atg5/Atg7-independent alternative macroautophagy
Nature.
2009
461
654
658

32.

Klionsky
DJ
et al.
Alternative macroautophagy
Autophagy.
2010
6
201

33.

Axe
EL
et al.
Autophagosome formation from membrane compartments enriched in phosphatidylinositol 3-phosphate and dynamically connected to the endoplasmic reticulum
J. Cell Biol.
2008
182
685
701

34.

Hailey
DW
et al.
Mitochondria supply membranes for autophagosome biogenesis during starvation
Cell.
2010
141
656
667

35.

Tooze
SA
et al.
The origin of the autophagosomal membrane
Nat. Cell Biol.
2010
12
831
835

36.

Ravikumar
B
et al.
Plasma membrane contributes to the formation of pre-autophagosomal structures
Nat. Cell Biol.
2010
12
747
757

37.

Hayashi-Nishino
M
et al.
A subdomain of the endoplasmic reticulum forms a cradle for autophagosome formation
Nat. Cell Biol.
2009
11
1433
1437

38.

Yla-Anttila
P
et al.
3D tomography reveals connections between the phagophore and endoplasmic reticulum
Autophagy.
2009
5
1180
1185

39.

Matsunaga
K
et al.
Autophagy requires endoplasmic reticulum targeting of the PI3-kinase complex via Atg14L
J. Cell Biol.
2010
190
511
521

40.

Tooze
SA
The role of membrane proteins in mammalian autophagy
Semin. Cell Dev. Biol.
2010
21
677
682

41.

Polson
HE
et al.
Mammalian Atg18 (WIPI2) localizes to omegasome-anchored phagophores and positively regulates LC3 lipidation
Autophagy.
2010
6
506
522

42.

McEwan
DG
et al.
Not all autophagy membranes are created equal
Cell.
2010
141
564
566

43.

Nair
U
et al.
Molecular mechanisms and regulation of specific and nonspecific autophagy pathways in yeast
J. Biol. Chem.
2005
280
41785
41788

44.

Levine
B
et al.
Autophagy in the pathogenesis of disease
Cell.
2008
132
27
42

45.

Sarkar
S
et al.
Autophagic clearance of aggregate-prone proteins associated with neurodegeneration
Meth. Enzymol.
2009
453
83
110

46.

Mathew
R
et al.
Role of autophagy in cancer
Nat. Rev. Cancer.
2007
7
961
967

47.

Wilkinson
S
et al.
Autophagy: an adaptable modifier of tumourigenesis
Curr. Opin. Genet. Dev.
2010
20
57
64

48.

Maiuri
MC
et al.
Control of autophagy by oncogenes and tumor suppressor genes
Cell Death Differ.
2009
16
87
93

49.

Hanahan
D
et al.
The hallmarks of cancer
Cell.
2000
100
57
70

50.

Crighton
D
et al.
Splicing DNA-damage responses to tumour cell death
Biochim. Biophys. Acta.
2004
1705
3
15

51.

Qu
X
et al.
Promotion of tumorigenesis by heterozygous disruption of the beclin 1 autophagy gene
J. Clin. Invest.
2003
112
1809
1820

52.

Yue
Z
et al.
Beclin 1, an autophagy gene essential for early embryonic development, is a haploinsufficient tumor suppressor
Proc. Natl Acad. Sci. USA.
2003
100
15077
15082

53.

Marino
G
et al.
Tissue-specific autophagy alterations and increased tumorigenesis in mice deficient in Atg4C/autophagin-3
J. Biol. Chem.
2007
282
18573
18583

54.

Takahashi
Y
et al.
Bif-1 interacts with Beclin 1 through UVRAG and regulates autophagy and tumorigenesis
Nat. Cell Biol.
2007
9
1142
1151

55.

Liang
XH
et al.
Induction of autophagy and inhibition of tumorigenesis by beclin 1
Nature.
1999
402
672
676

56.

Aita
VM
et al.
Cloning and genomic organization of beclin 1, a candidate tumor suppressor gene on chromosome 17q21
Genomics.
1999
59
59
65

57.

Miracco
C
et al.
Protein and mRNA expression of autophagy gene Beclin 1 in human brain tumours
Int. J. Oncol.
2007
30
429
436

58.

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
702
706

59.

Kim
MS
et al.
Frameshift mutation of UVRAG, an autophagy-related gene, in gastric carcinomas with microsatellite instability
Hum. Pathol.
2008
39
1059
1063

60.

Hoyer-Hansen
M
et al.
Autophagy: an emerging target for cancer therapy
Autophagy.
2008
4
574
580

61.

Degenhardt
K
et al.
Autophagy promotes tumor cell survival and restricts necrosis, inflammation, and tumorigenesis
Cancer Cell.
2006
10
51
64

62.

Kroemer
G
et al.
Autophagic cell death: the story of a misnomer
Nat. Rev. Mol. Cell. Biol.
2008
9
1004
1010

63.

Galluzzi
L
et al.
Guidelines for the use and interpretation of assays for monitoring cell death in higher eukaryotes
Cell Death Differ.
2009
16
1093
1107

64.

Lam
D
et al.
A specific pathway inducing autophagic cell death is marked by an IP3R mutation
Autophagy.
2008
4
349
350

65.

Berry
DL
et al.
Growth arrest and autophagy are required for salivary gland cell degradation in Drosophila
Cell.
2007
131
1137
1148

66.

Berry
DL
et al.
Autophagy functions in programmed cell death
Autophagy.
2008
4
359
360

67.

Denton
D
et al.
Autophagy, not apoptosis, is essential for midgut cell death in Drosophila
Curr. Biol.
2009
19
1741
1746

68.

Kang
C
et al.
Dual roles of autophagy in the survival of Caenorhabditis elegans during starvation
Genes Dev.
2007
21
2161
2171

69.

Yu
L
et al.
Regulation of an ATG7-beclin 1 program of autophagic cell death by caspase-8
Science.
2004
304
1500
1502

70.

Shimizu
S
et al.
Role of Bcl-2 family proteins in a non-apoptotic programmed cell death dependent on autophagy genes
Nat. Cell Biol.
2004
6
1221
1228

71.

Crighton
D
et al.
DRAM, a p53-induced modulator of autophagy, is critical for apoptosis
Cell.
2006
126
121
134

72.

Crighton
D
et al.
DRAM links autophagy to p53 and programmed cell death
Autophagy.
2007
3
72
74

73.

O’Prey
J
et al.
Analysis of DRAM-related proteins reveals evolutionarily conserved and divergent roles in the control of autophagy
Cell Cycle.
2009
8
2260
2265

74.

Fung
C
et al.
Induction of autophagy during extracellular matrix detachment promotes cell survival
Mol. Biol. Cell.
2008
19
797
806

75.

Xue
W
et al.
Senescence and tumour clearance is triggered by p53 restoration in murine liver carcinomas
Nature.
2007
445
656
660

76.

Young
AR
et al.
Autophagy mediates the mitotic senescence transition
Genes Dev.
2009
23
798
803

77.

Wang
G
et al.
Endoplasmic reticulum stress response in cancer: molecular mechanism and therapeutic potential
Am. J. Transl. Res.
2010
2
65
74

78.

Kongara
S
et al.
Autophagy regulates keratin 8 homeostasis in mammary epithelial cells and in breast tumors
Mol. Cancer Res.
2010
8
873
884

79.

Perlmutter
DH
Autophagic disposal of the aggregation-prone protein that causes liver inflammation and carcinogenesis in alpha-1-antitrypsin deficiency
Cell Death Differ.
2009
16
39
45

80.

Colotta
F
et al.
Cancer-related inflammation, the seventh hallmark of cancer: links to genetic instability
Carcinogenesis.
2009
30
1073
1081

81.

Reuter
S
et al.
Oxidative stress, inflammation, and cancer: how are they linked?
Free Radic. Biol. Med.
2010
49
1603
1616

82.

Van Limbergen
J
et al.
The genetics of Crohn’s disease
Annu. Rev. Genomics Hum. Genet.
2009
10
89
116

83.

Cadwell
K
et al.
A key role for autophagy and the autophagy gene Atg16l1 in mouse and human intestinal Paneth cells
Nature.
2008
456
259
263

84.

Cadwell
K
et al.
A common role for Atg16L1, Atg5 and Atg7 in small intestinal Paneth cells and Crohn disease
Autophagy.
2009
5
250
252

85.

Varnat
F
et al.
PPARbeta/delta regulates paneth cell differentiation via controlling the hedgehog signaling pathway
Gastroenterology.
2006
131
538
553

86.

Saitoh
T
et al.
Loss of the autophagy protein Atg16L1 enhances endotoxin-induced IL-1beta production
Nature.
2008
456
264
268

87.

Ikeuchi
H
et al.
Intestinal cancer in Crohn’s disease
Hepatogastroenterology.
2008
55
2121
2124

88.

Ekbom
A
et al.
Increased risk of large-bowel cancer in Crohn’s disease with colonic involvement
Lancet.
1990
336
357
359

89.

Ekbom
A
et al.
Pancreatitis and the risk of pancreatic cancer
N. Engl. J. Med.
1993
329
1502
1503

90.

Mareninova
OA
et al.
Impaired autophagic flux mediates acinar cell vacuole formation and trypsinogen activation in rodent models of acute pancreatitis
J. Clin. Invest.
2009
119
3340
3355

91.

Li
Y
et al.
Efficient cross-presentation depends on autophagy in tumor cells
Cancer Res.
2008
68
6889
6895

92.

Walsh
CM
et al.
The complex interplay between autophagy, apoptosis, and necrotic signals promotes T-cell homeostasis
Immunol. Rev.
2010
236
95
109

93.

Pua
HH
et al.
A critical role for the autophagy gene Atg5 in T cell survival and proliferation
J. Exp. Med.
2007
204
25
31

94.

Pua
HH
et al.
Maintaining T lymphocyte homeostasis: another duty of autophagy
Autophagy.
2007
3
266
267

95.

Pua
HH
et al.
Autophagy is essential for mitochondrial clearance in mature T lymphocytes
J. Immunol.
2009
182
4046
4055

96.

Stephenson
LM
et al.
Identification of Atg5-dependent transcriptional changes and increases in mitochondrial mass in Atg5-deficient T lymphocytes
Autophagy.
2009
5
625
635

97.

Nedjic
J
et al.
Autophagy in thymic epithelium shapes the T-cell repertoire and is essential for tolerance
Nature.
2008
455
396
400

98.

Dengjel
J
et al.
Autophagy promotes MHC class II presentation of peptides from intracellular source proteins
Proc. Natl Acad. Sci. USA.
2005
102
7922
7927

99.

Paludan
C
et al.
Endogenous MHC class II processing of a viral nuclear antigen after autophagy
Science.
2005
307
593
596

100.

Taylor
GS
et al.
A role for intercellular antigen transfer in the recognition of EBV-transformed B cell lines by EBV nuclear antigen-specific CD4+ T cells
J. Immunol.
2006
177
3746
3756

101.

Leung
CS
et al.
Nuclear location of an endogenously expressed antigen, EBNA1, restricts access to macroautophagy and the range of CD4 epitope display
Proc. Natl Acad. Sci. USA.
2010
107
2165
2170

102.

Scherz-Shouval
R
et al.
Reactive oxygen species are essential for autophagy and specifically regulate the activity of Atg4
EMBO J.
2007
26
1749
1760

103.

Narendra
D
et al.
Parkin is recruited selectively to impaired mitochondria and promotes their autophagy
J. Cell Biol.
2008
183
795
803

104.

Narendra
DP
et al.
PINK1 is selectively stabilized on impaired mitochondria to activate Parkin
PLoS Biol.
2010
8
e1000298

105.

Suen
DF
et al.
Parkin overexpression selects against a deleterious mtDNA mutation in heteroplasmic cybrid cells
Proc. Natl Acad. Sci. USA.
2010
107
11835
11840

106.

Novak
I
et al.
Nix is a selective autophagy receptor for mitochondrial clearance
EMBO Rep.
2010
11
45
51

107.

Sandoval
H
et al.
Essential role for Nix in autophagic maturation of erythroid cells
Nature.
2008
454
232
235

108.

Mathew
R
et al.
Autophagy suppresses tumorigenesis through elimination of p62
Cell.
2009
137
1062
1075

109.

Karantza-Wadsworth
V
et al.
Autophagy mitigates metabolic stress and genome damage in mammary tumorigenesis
Genes Dev.
2007
21
1621
1635

110.

Mathew
R
et al.
Autophagy suppresses tumor progression by limiting chromosomal instability
Genes Dev.
2007
21
1367
1381

111.

Moscat
J
et al.
p62 at the crossroads of autophagy, apoptosis, and cancer
Cell.
2009
137
1001
1004

112.

Bjorkoy
G
et al.
p62/SQSTM1: a missing link between protein aggregates and the autophagy machinery
Autophagy.
2006
2
138
139

113.

Komatsu
M
et al.
Homeostatic levels of p62 control cytoplasmic inclusion body formation in autophagy-deficient mice
Cell.
2007
131
1149
1163

114.

Komatsu
M
et al.
Essential role for autophagy protein Atg7 in the maintenance of axonal homeostasis and the prevention of axonal degeneration
Proc. Natl Acad. Sci. USA.
2007
104
14489
14494

115.

Komatsu
M
et al.
Loss of autophagy in the central nervous system causes neurodegeneration in mice
Nature.
2006
441
880
884

116.

Duran
A
et al.
The signaling adaptor p62 is an important NF-kappaB mediator in tumorigenesis
Cancer Cell.
2008
13
343
354

117.

Wilkinson
S
et al.
Hypoxia-selective macroautophagy and cell survival signaled by autocrine PDGFR activity
Genes Dev.
2009
23
1283
1288

118.

Wilkinson
S
et al.
Growth factor signaling permits hypoxia-induced autophagy by a HIF1alpha-dependent, BNIP3/3L-independent transcriptional program in human cancer cells
Autophagy.
2009
5
1068
1069

This is an Open Access article distributed under the terms of the Creative Commons Attribution Non-Commercial License (http://creativecommons.org/licenses/by-nc/2.5), which permits unrestricted non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited.