Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2022 Apr 5.
Published in final edited form as: Dev Cell. 2021 Mar 8;56(7):906–918. doi: 10.1016/j.devcel.2021.02.010

Autophagy and Organelle Homeostasis in Cancer

Dannah R Miller 1, Andrew Thorburn 1
PMCID: PMC8026727  NIHMSID: NIHMS1679647  PMID: 33689692

Abstract

Beginning with the earliest studies of autophagy in cancer, there have been indications that autophagy can both promote and inhibit cancer growth and progression. Autophagy regulation of organelle homeostasis is similarly complicated. In this review, we discuss pro- and anti-tumor effects of organelle-targeted autophagy and how this contributes to several hallmarks of cancer, such as evading cell death, genomic instability, and altered metabolism. Typically, the removal of damaged or dysfunctional organelles prevents tumor development but can also aid in proliferation or drug resistance in established tumors. By better understanding how organelle-specific autophagy takes place and can be manipulated, it may be possible to go beyond the brute force approach of trying to manipulate all autophagy in order to improve therapeutic targeting of this process in cancer.

Thorburn ETOC

Autophagy can both promote and inhibit cancer growth and progression. Miller and Thorburn review the pro- and anti-tumor effects of organelle-targeted autophagy and their relationship to cancer hallmarks, such as evading cell death, genomic instability, and altered metabolism. These insights may help improve therapeutic targeting of autophagy in cancer.

Introduction

The recycling processes that deliver excess or damaged cytoplasmic material to lysosomes for degradation are one of three pathways: micro-autophagy, chaperone-mediated autophagy, and macro-autophagy. Macro-autophagy (hereafter referred to as autophagy) is a complex process that promotes both bulk and selective degradation of damaged proteins and organelles or spare molecules to generate macromolecular building blocks and fuel metabolic pathways (Dikic and Elazar, 2018). Autophagy involves over 20 core autophagy proteins, encoded by ATG genes, that envelop cytoplasmic cargo within a double-membrane vesicle structure called the autophagosome (Suzuki et al., 2001). After engulfment of the cargo, autophagosomes fuse with lysosomes, where pH-sensitive hydrolases mediate degradation of the engulfed material (Dikic and Elazar, 2018). Selective autophagy occurs through targeting of specific cargos, including organelles, to autophagosomes. Micro-autophagy and chaperone-mediated autophagy are considered to be selective forms of autophagy that involve direct delivery mechanisms of targeted material to the lysosome (Anding and Baehrecke, 2017, Mijaljica et al., 2011, Sahu et al., 2011, Kaushik and Cuervo, 2018). Selective autophagy has three distinct steps: designation, targeting and sequestration, and degradation. Designation occurs when a cargo receives a molecular tag for its recognition by the autophagy machinery. Ubiquitination often provides this signal in mammalian cells (Kim et al., 2008, Rogov et al., 2014). Next, targeting and sequestration occur when designated cargoes are delivered to autophagosomes by specific receptors, allowing the autophagosome to engulf and sequester cargo. Numerous Autophagy Cargo Receptors (ACRs) have been identified in mammals, including SQSTM1/p62, NBR1, optineurin (OPTN), NDP52, TAX1BP, BNIP3-like (BNIP3L)/NIX, and TRIM proteins (Rogov et al., 2014, Mandell et al., 2014). A key step in the formation of autophagosomes is the conjugation of the LC3/GABARAP family of proteins to the lipid phosphatidylethanolamine (PE), which is mediated by the autophagy conjugation machinery including ATG3, ATG5, ATG7, and ATG12. Importantly, these conjugation events are frequently used as a method to monitor autophagy levels (the most common method to quantitate the amount of autophagy is to measure the rate of conjugation of LC3B to PE). A common way to block autophagy involves the inactivation by knockout or knockdown of one or other component of the conjugation machinery. These conjugation events are important for proper development of autophagosomes and the conjugated LC3 molecules on the inner autophagosome membrane serve as targets that are recognized by the ACRs. Then, fusion of the autophagosome with the lysosome takes place, followed by degradation of the inner autophagosome membrane, allowing lysosomal hydrolases access to degrade the cargo. Lastly, macromolecular precursors including amino acids, lipids etc. resulting from degradation of the cargo are released to the cytoplasm to fuel metabolic pathways and serve as substrates for the creation of new macromolecules.

Autophagy can both promote and inhibit tumor growth in different contexts (White, 2015, Amaravadi et al., 2019). For example, genetic inhibition of Atg5 or Atg7 showed that deletion of autophagy in tumor-prone mouse models following RAS pathway activation caused an increase in preneoplastic lesions and tumor incidence, indicating that autophagy protects against cancer development (Strohecker et al., 2013, Rao et al., 2014, Rosenfeldt et al., 2013, Yang et al., 2014). In these mice, it is likely that the defective autophagy process causes chronic tissue damage, leading to tumor-initiating inflammation (White et al., 2010). Various other tumor suppressive functions of autophagy have also been proposed, as autophagy plays an important role in tissue homeostasis. Most of these mechanisms likely affect tumor initiation and early steps in cancer progression, including removal of damaging, reactive oxygen species (ROS)–inducing mitochondria, degradation of oncogenic viruses, maintenance of genomic stability, and a role in oncogene-induced senescence via degradation of the nuclear lamina (Levine and Kroemer, 2019, Wang et al., 2016, Dou et al., 2015). However, it is important to note that it is possible to have mutations or expression of alternate splice variants of autophagy genes that negatively regulate autophagy (Wible et al., 2019), this seldom occurs in cancer (Lebovitz et al., 2015). Later in disease progression, it is more common for oncogenic events to activate autophagy or enhance lysosomal biogenesis to withstand nutrient deprivation as well as prevent excessive accumulation of the toxic molecules discussed above (Amaravadi et al., 2019).

Therefore, once a tumor becomes established, autophagy inhibition often results in less aggressive cancers. For example, pancreatic cancers have increased autophagy flux that contributes to their growth (Yang et al., 2011) through multiple tumor cell intrinsic and nontumor-intrinsic mechanisms (Piffoux et al., 2020, Yang et al., 2018). As such, mutations in autophagy genes would thwart the tumor’s rapid growth and metastasis and instead result in death or senescence. This increase in autophagy flux makes pancreatic cancer a strong candidate for therapeutic targeting of autophagy. Roles of autophagy in cancer and the potential of targeting autophagy as a therapeutic strategy for various cancers have been extensively reviewed (Mulcahy Levy et al., 2017, Galluzzi et al., 2015, Amaravadi et al., 2019, Mulcahy Levy and Thorburn, 2020, White, 2015). Because tumors are established at the time of detection and treatment, autophagy inhibition would be effective for this stage of the disease. However, autophagy also has pro- and anti-metastatic effects. For example, autophagy can aid in the ability to endure environmental stress as well as promote an anti-tumor immune response or cell dormancy for migrating cancer cells {Kenific, 2010 #387}. Given these competing roles of autophagy in general, targeting organelle homeostasis rather than general autophagy processes could potentially be a more effective way to treat cancer by allowing more precision in targeting pro-tumor functions while avoiding anti-tumor promoting functions of autophagy.

In this Review, we discuss how the role of autophagy in organelle homeostasis may affect tumor behavior. However, it is important to keep in mind that, although many studies propose critical roles for specific types of organelle-phagy in cancer cell behavior, such conclusions may be over simplified. In many of these studies, a component of the autophagy machinery, often a component of the conjugation machinery, is inhibited as a way to block organelle-specific autophagy. But such studies do not exclude the possibility that the biological effect depends not (only) on the loss of degradation of the specific organelle in question, but also on the absence of degradation of other molecules or organelles. In addition, in many cases it remains possible that the effect is due to autophagy-independent functions autophagy components (Galluzzi and Green, 2019). Therefore, specific manipulation of organelle-specific autophagy pathways is required to confirm that a biological effect arises due to autophagy of that specific organelle. However, this may be difficult due to the redundancy of autophagy adaptors utilized across different types of organelle-targeted autophagy.

Mitophagy and Cancer

Mitochondria are well-known for their role in ATP generation (Green and Kroemer, 2004), but they have several other functions that are intertwined with hallmarks of cancer (Giampazolias and Tait, 2016). Mitochondria are vulnerable to damage from high levels of ROS, a byproduct of aerobic respiration that can also cause DNA mutations and protein damage/misfolding. Mitochondria are also involved in fatty acid synthesis, amino acid production, heme synthesis and iron–sulfur cluster biogenesis, and play an essential role in the execution of apoptosis by releasing cytochrome c (Wang and Youle, 2009). As such, the clearance of mitochondria can have diverse effects on tumor development, growth, and progression.

The autophagy-targeted degradation of mitochondria is accomplished by a selective form of autophagy, named mitophagy (Lemasters, 2005) (Figure 1). Mitophagy can be initiated by stimuli that cause mitochondrial damage, such as hypoxia (Bellot et al., 2009; Zhang et al., 2008), chemical uncouplers (Narendra et al., 2008), or ROS (Frank et al., 2012; Zhang et al., 2008). The best understood mechanism of mitophagy involves PTEN-induced putative kinase 1 (PINK1), a mitochondrially localized kinase, and Parkin, a cytosolic E3 ubiquitin ligase (Narendra et al., 2010), both of which are mutated in early-onset recessive Parkinson’s disease. A small portion of PINK1 is imported into mitochondria through two complexes: the translocase of the outer membrane (TOM) and the translocase of the inner membrane (TIM23). While in the inner mitochondrial membrane, matrix processing peptidase (MPP) clips the intra-membrane section of PINK1 and the inner membrane protease PINK1/PGAM5- associated rhomboid-like protease (PARL) (Jin et al., 2010). After cleavage and activation, PINK1 phosphorylates Parkin to increase its ubiquitin E3 ligase activity (Shiba-Fukushima et al., 2012, Koyano et al., 2014) resulting in activation of Parkin and the addition of ubiquitin to various mitochondrial substrates, eventually forming ubiquitin chains on a variety of these proteins (Okatsu et al., 2015). Several ACRs mediate Parkin-dependent mitophagy in mammalian cells: p62/SQSTM1, NDP52, OPTN, NIX, FUNDC1, BCL2/adenovirus E1B 19 kDa protein-interacting protein 3 (BNIP3), and TAX1BP1 (Lazarou et al., 2015, Heo et al., 2015, Chen et al., 2016). These ARCs direct ubiquitinated mitochondria to phagophores by binding the ubiquitin molecules on the mitochondrial surface and LC3 family members on the inner autophagosome membrane via a LC3-interacting region (LIR) domain (Figure 1).

Figure 1. Dysfunctional Mitochondria can both Promote and Inhibit Cancer Growth and Survival.

Figure 1.

A. Healthy mitochondria produce ATP for cellular energy use. Overworking mitochondria or treatment with anticancer agents such as cisplatin can promote mitochondrial damage. Damaged mitochondria can promote tumor survival and progression through ROS and mitochondrial DNA release as well as promoting the Warburg effect. However, the release of cytochrome C from mitochondria in this state could promote apoptosis. B. Clearance of mitochondria through mitophagy typically occurs through PINK1-mediated phosphorylation of Parkin, which promotes protein ubiquitination. However, mitophagy can also occur in the absence of Parkin, for example via AMBRA1–1 and HUWE1-mediated ubiquitination of MFN2. Also, BNIP3 and NIX can recruit LC3 to the mitochondria in the absence of ubiquitinated mitochondrial membrane proteins.

Although cancer cells often do not express Parkin (Chang et al., 2017, Zhang et al., 2020), they adopt similar mechanisms for removal of mitochondria, usually through alternate ubiquitin ligases (Villa et al., 2018). ARIH1 is a ubiquitin ligase that has the potential to promote mitophagy in the absence of Parkin in various types of cancer cells (Villa et al., 2017). Similarly, Autophagy and Beclin1 Regulator 1 (AMBRA1) localizes to damaged mitochondria through LIR motif-dependent interactions with LC3, promoting both Parkin-dependent and –independent mitochondrial degradation (Strappazzon et al., 2015). This interaction enhances the mitochondrial localization of HECT, UBA And WWE Domain Containing E3 Ubiquitin Protein Ligase 1 (HUWE1), which ubiquitinates mitofusin-2 (MFN2) to promote mitophagy after membrane depolarization (Di Rita et al., 2018). Mitophagy receptors BNIP3 and NIX can also trigger mitophagy in the absence of protein ubiquitination (Novak et al., 2010, Hanna et al., 2012). All these mechanisms rely upon LC3 conjugation to selectively sequester mitochondria in the autophagosome. One major unanswered question is whether mitophagy can still occur in cancer cells when the autophagy apparatus is disabled as occurs when, as discussed below, we design experiments where we inactivate a component of the conjugation machinery in tumors.

Mitophagy as a Pro-Survival Mechanism

Because mitochondrial permeabilization is the key rate limiting step in apoptosis, one obvious mechanism by which mitophagy might allow cancer cells to evade death is by removing mitochondria that could potentially be permeabilized to induce cell death. Degradation of dysfunctional mitochondria has been reported to promote cell survival by preventing the production or release of toxic byproducts such as ROS and cytochrome c to cause apoptosis (Galluzzi et al., 2006, Colell et al., 2007). Consistent with this, mitophagy has been reported to cause cancer cell resistance to radiation (Wang et al., 2016), microtubule inhibitors (Wei et al., 2018), doxorubicin (Zhang et al., 2012, Yan et al., 2017), cisplatin, and etoposide (Villa et al., 2017, Yao et al., 2019), supporting a pro-survival role of mitophagy in cancer cells in response to chemo/radiotherapy (Abdrakhmanov et al., 2019). However, the mechanisms at play in this survival response may be complicated. Cancer cells are primed to undergo apoptosis because they more easily undergo mitochondrial permeabilization compared to normal cells. Indeed, this is the basis for the therapeutic window that allows many cancer therapies to work (Sarosiek et al., 2013). Autophagic degradation of specific proteins can regulate the efficacy of and sensitivity to anti-cancer drugs by altering the ease with which mitochondrial permeabilization can take place (Fitzwalter et al., 2018), potentially setting the apoptotic threshold and thus the therapeutic window for cancer drugs (Tompkins and Thorburn, 2019). This mechanism, though resulting in a similar effect of increased drug sensitivity when autophagy is blocked, is independent of mitophagy itself. This emphasizes the importance of the caveat described above– it is critical to carry out detailed mechanistic studies to determine if the degradation of the organelle itself is responsible for the biological effect. Indeed in the examples cited above where mitophagy has been linked to cancer cell drug resistance, Villa et al. (2017) provided evidence that their effects were mediated by a specific mitophagy ACR, but the others involved more general inhibition of autophagy. Blocking mitophagy could nevertheless be a promising anti-cancer therapeutic strategy to reduce resistance to other anti-cancer treatments (Drake et al., 2017). Moreover, autophagy and mitophagy are involved in dampening the anti-tumor immune response through the clearance of damage-associated molecular patterns (DAMPs), such as mitochondrial DNA (mtDNA) (Nakahira et al., 2011, Saitoh and Akira, 2016). Upon release from a damaged cell, pattern recognition receptors (PRRs) recognize the mtDNA, or other DAMPS, and initiate signaling that will upregulate pro-inflammatory interferons or cytokines or trigger the NLRP3 inflammasome to activate the innate immune system (Riley and Tait, 2020). More recent studies suggest that blocking mitophagy may enhance immunotherapy by allowing accumulation of cytosolic mitochondrial DNA after radiation, which promotes an enhanced Interferon-mediated tumor immune response that can make immune checkpoint inhibitors more effective (Yamazaki et al., 2020).

Accumulating evidence indicates that autophagy supports the metabolic plasticity of cancer cells by providing virtually all essential components of carbon metabolism through the degradation of carbohydrates, proteins, lipids, and nucleotides (Poillet-Perez and White, 2019). KRAS mutations, which frequently occur in several types of cancers, upregulate glycolysis and overwork the mitochondria (Weinberg et al., 2010), creating a need for increased mitochondrial turnover. In such tumors, increased autophagy or mitophagy has been suggested to support aggressive tumor growth (Kim et al., 2011, Rao et al., 2014). It has been well-documented that autophagy-deficient tumors have dysfunctional mitochondria that exhibit altered morphology, ineffective fatty acid oxidation, reduced carbon flux through the Krebs cycle, and lipid accumulation, all of which can act as tumor suppressors (Guo et al., 2011, Strohecker et al., 2013). Given that many tumors exhibit the Warburg effect (Warburg, 1956, Warburg et al., 1927), hyperactivation of mitophagy could solidify their glycolytic addiction by diverting the flux of metabolites away from the mitochondria (Narendra et al., 2008). Further evidence of a pro-tumorigenic role of mitophagy can be seen in oncocytomas, which in mouse models can arise from adenomas with defective autophagy (Guo et al., 2013). In this context, a reduction in autophagy flux may cause accumulation of dysfunctional mitochondria, resulting in switching to the less aggressive oncocytoma from more aggressive adenomas. In human oncocytomas, which also tend to be less aggressive, autophagy is also compromised (Joshi et al., 2015). Therefore, the role of mitophagy in the context of metabolism appears to be primarily pro-tumorigenic through maintaining healthy mitochondria (Figure 1).

Tumor Suppressive Roles of Mitophagy

Mitophagy is also associated with tumor suppression due to reduced ROS levels or energy production, which negatively impacts tumor metastasis and prevents genomic instability (Figure 1). Dysfunctional mitochondria and increased mitochondrial ROS can promote tumorigenesis, cancer progression, metastasis, and drug resistance through DNA, lipid, and protein oxidation (Weinberg et al., 2010, Chourasia et al., 2015, Yan et al., 2017). In a mouse model of breast cancer, depletion of the mitophagy ACR BNIP3 reduced mitophagy leading to increased mitochondrial ROS levels. This contributed to increased Hypoxia-Inducible Factor 1 alpha (HIF1α) stabilization, eventually promoting the Warburg effect and, subsequently, tumor progression (Chourasia et al., 2015). The loss of Parkin in many cancers has been interpreted as suggesting a tumor suppressive function for mitophagy (Chang et al., 2017, Zhang et al., 2020). Indeed, Parkin or PINK1 deletion in mice leads to the spontaneous development of hepatocellular carcinoma (Fujiwara et al., 2008) and increases KRAS-driven pancreatic tumorigenesis (Li et al., 2018). The accumulation of defective mitochondria induced by Parkin deficiency decreases mitochondrial oxidative phosphorylation (OXPHOS), increases ROS production, and increases glycolysis, therefore possibly contributing to the Warburg effect and consequently increasing tumorigenesis (Zhang et al., 2011). Additionally, both Parkin- and PINK1-null mice display an inflammatory phenotype due to Simulator of Interferon Genes (STING)-mediated interferon response (Sliter et al., 2018), suggesting that mitophagy may play a role in tumor suppression through prevention of tumor-promoting inflammation. However, as noted above, because numerous alternative mechanisms can lead to mitophagy in the absence of Parkin or PINK1, it is presently unclear whether the tumor suppressive functions of these molecules are due solely to reduced mitophagy.

Lysophagy

The lysosome is an acidic, membrane-bound organelle containing hydrolytic enzymes such as cathepsins, which are the main executioners of protein degradation in autophagy. During transformation, cancer cells alter their lysosomal compartment, including its size, localization, cathepsin expression, and activity (Gocheva et al., 2006, Nishimura et al., 1998). These changes in lysosomes often correlates with invasive growth, angiogenesis, and drug resistance (Fehrenbacher et al., 2004). Macropinocytosis, which relies on the lysosome for degradation of extracellular material, is also elevated in nutrient-deprived cancers, including RAS-driven bladder or pancreatic cancers (Commisso et al., 2013, Kamphorst et al., 2015, Sukhai et al., 2013) and tumor cells with WNT pathway activation (Tejeda-Munoz et al., 2019). Lysosomes also play a vital role in cancer drug resistance by sequestering drugs, which can prevent them from reaching their intended target molecule and provide a mechanism for removal of drugs (Zhitomirsky and Assaraf, 2016). Conversely, the lysosome can become a dangerous organelle when the lysosomal membrane is permeabilized (LMP), allowing leaking of cathepsins and other molecules into the cytoplasm (Aits and Jaattela, 2013) (Figure 2). This lysosomal cell death is of interest in cancer therapy because it may help avoid some of the oncogenic effects that can arise when apoptosis is the main death mechanism for tumor cells (Cao and Tait, 2018).

Figure 2. Lysophagy Promotes Cell Survival by Removal of Toxic Permeabilized Lysosomes.

Figure 2.

A. Functional lysosomes act as the cell’s recycling system to degrade proteins and other macromolecules. When the lysosome becomes permeabilized by lysosome membrane permeabilization agents or anticancer agents such as paclitaxel, cathepsins and ROS may be released from this organelle, which often promotes cell death. B. Permeabilization of the lysosome is first detected by Galectin proteins. The cell can either repair the membrane damage through increased HSP70 expression to prevent large scale lysosome membrane permeabilization and cathepsin release or target the lysosome for lysophagy. UBE2QL1 promotes K48 ubiquitin chain elongation on lysosomal membrane proteins, which then attracts p97. Part of the p97 deubiquitation complex is the deubiquitinase YOD1, which removes the K48 ubiquitin chains. This promotes K63 ubiquitination through an unknown mechanism, allowing for p62 interaction and engulfment of the lysosome.

Cells degrade damaged or permeabilized lysosomes through lysophagy (Hung et al., 2013, Maejima et al., 2013). Once the lysosome has become permeabilized, several sensors, galectin (Gal)-1, −3, −8 and −9, bind exposed β-galactosides at the inner lysosomal membrane and elicit downstream signaling events (Thurston et al., 2012). Coordinated actions of galectin binding proteins are then thought to elicit lysophagy. Once galectins sense the damaged organelle, Ubiquitin Conjugating Enzyme E2 Q Family Like 1 (UBE2QL1) tags lysosomal membrane proteins with ubiquitin on K48 residues (Koerver et al., 2019, Kravic et al., 2020). After UBE2QL1-mediated protein ubiquitination, the ubiquitin-directed AAA-ATPase p97 is recruited to damaged lysosomes (Papadopoulos et al., 2017, Seczynska and Dikic, 2017, Tanaka et al., 2010). The K48-linked ubiquitin chains are subsequently removed by p97 cofactor and deubiquitinating enzyme YOD1, which then, through an unknown mechanism, promotes K63 ubiquitination, recruitment of p62, and association of the LC3-decorated phagophore (Papadopoulos et al., 2017, Seczynska and Dikic, 2017). It is important to note that different galectins have different roles within this process. For example, Gal3 recruits the tripartite motif (TRIM) protein TRIM16, which serves as a platform for autophagic initiation factors that induce phagophore formation (Chauhan et al., 2016). Meanwhile, Gal8 binds the ARC NDP52, which recruits LC3-positive phagophores that mediate lysophagy (Thurston et al., 2012) (Figure 2).

Because functional lysosomes are critical for cancer cell survival and play a vital role in drug resistance, there is interest in killing cancer cells through the induction of LMP. Several agents that can promote LMP and can also induce cell death include ROS, sphingosine, lysomotrophic toxins, or vacuolar H+-ATPase inhibitors (Aits and Jäättelä, 2013; Maejima et al., 2013). It has also been noted that many anti-cancer agents whose mechanism of action is usually considered to be through different cellular targets, such as TNFα, paclitaxel, vincristine, or siramesine, can also induce LMP (Werneburg et al., 2007, Broker et al., 2004, Groth-Pedersen et al., 2007, Erdal et al., 2005, Ostenfeld et al., 2005, Groth-Pedersen and Jaattela, 2013, Settembre et al., 2013). For such agents, lysophagy may prevent the cell from dying by removing these organelles therefore leading to drug resistance. There are currently no known specific inhibitors of lysophagy that might be used to test feasibility of this strategy. It is important to note that autophagy inhibitors that are used clinically, such as hydroxychloroquine and related molecules such as Lys05 and DQ661, operate through inhibition of lysosome function, specifically through preventing lysosomal palmitoyl protein thioesterase 1 (PPT1)-mediated removal of fatty acids on proteins (Rebecca et al., 2019). Combination therapy of LMP-inducing agents with autophagy/lysosomal inhibition agents such as these may also provide a strategy for cancer therapy.

ER-phagy (Reticulophagy)

The endoplasmic reticulum (ER) has multiple functions, including protein folding, processing, and transport, lipid and steroid synthesis, calcium storage and homeostasis, and detoxification. When the cell demands a high amount of protein synthesis and accumulates misfolded proteins, as often occurs in cancer, the ER becomes stressed, which can trigger cell death (Friedman and Voeltz, 2011, Nixon-Abell et al., 2016) (Figure 3). In addition to constitutive recycling, the ER must undergo more active turnover under certain stress conditions to achieve optimal quality of individual proteins and the entire organelle. A well-known mechanism to combat this event is the unfolded protein response (UPR), which halts protein translation and upregulates various chaperones to aid in correctly organizing misfolded proteins. Another mechanism to relieve ER stress is by the autophagic targeting and degradation of the ER, termed ER-phagy. ER-phagy is thought to aid in ER remodeling to minimize ER stress. However, while toxicities are often attributed to the loss of autophagy following ER stress, it is often unclear whether such effects are due to loss of ER-phagy itself or if other functions of autophagy contribute to the survival of the ER stressed cell. For example, tissue-specific deletion of general autophagy proteins, such as Atg5 in terminally differentiated B-lymphocytes (Pengo et al., 2013) or Atg5 or Atg7 in pancreatic acinar cells, drives excessive ER expansion, which may contribute to cell death (Antonucci et al., 2015), but in such studies, general autophagy is also compromised. To more specifically test if ER-phagy is responsible, one could try to selectively block only this kind of autophagy. However, this is difficult to do because, as with mitophagy, multiple ACRs mediate selective targeting of the ER, including Family With Sequence Similarity 134 Member B (FAM134B), Reticulons (RTN1–4), Alastin-3 (ATL3), SEC62, CCPG1, TEX264, and NDP52/CALCOCO2 (Nthiga et al., 2020). Each receptor has a specific target and function in proteostasis and/or ER fragmentation. FAM134B and RTN1–4 drive ER tubulation, sheet edge curvature, and sheet fenestration to promote the remodeling and scission of ER tubules through its reticulon domain (Schroeder et al., 2019, Voeltz et al., 2006). SEC62 mediates macroER-phagy during cellular recovery from an acute UPR response and delivers portions of ER into autophagosomes following ER stress in a process termed ‘recovER-phagy’ (Fumagalli et al., 2016, Loi et al., 2019). CCPG1 is a type II, single-pass transmembrane protein that delivers portions of the ER to autophagosomes upon ER-stress (Kostenko et al., 2006), typically in response to UPR, thus helping rid the cell of misfolded proteins or protein aggregates (Smith et al., 2018). TEX264 is a single pass, transmembrane receptor for nutrient starvation-induced ER-phagy that targets three-way ER junctions upon nutrient starvation (Chino et al., 2019, An et al., 2019). All adaptors contain an LIR motif, which can bind to the autophagosome to target the ER for degradation (Chen et al., 2019, Smith et al., 2018, Grumati et al., 2017, Khaminets et al., 2015, An et al., 2019, Fumagalli et al., 2016). Also, all adaptors besides ATL3 have long intrinsically disordered regions between the ER transmembrane domain and the LIR that cannot fold into a compact structure, which allows for ER-targeting when ribosomes are on the ER (Chino et al., 2019, Meszaros et al., 2007) (Figure 3).

Figure 3. ER Stress in Cancer Requires Increased ER Turnover.

Figure 3.

A. A healthy ER is crucial for protein translation. However, ER stress occurs in cancer cells upon nutrient deprivation, hypoxia, and anticancer treatments, often due to excessive amounts of protein translation, protein aggregation, and UPR. B. ER-phagy has many known adaptors that promote the degradation of stressed or damaged ER to prevent cell death. SEC62, CCPG1, and FAM134B all function in promoting the degradation of ER sheets, while RTN, ALT3, and TEX364 are utilized for ER tubule degradation. TEX264 is unique in its targeting of three-way junctions. Receptors all have LIR interacting domains allowing for binding to LC3, and can be activated upon nutrient deprivation or UPR.

There is evidence suggesting specific pro-tumor roles of ER-phagy in cancer. Overexpression of FAM134B can transform noncancerous cells (Tang et al., 2007) and mutations and alterations in FAM134B expression have been observed in various malignancies (Islam et al., 2018, Kasem et al., 2014b, Kasem et al., 2014a, Kasem et al., 2014c). Glioma cells require FAM134B-driven ER-phagy to survive proteotoxic stress (Peng et al., 2018), suggesting the potential for this molecule as a therapeutic target in cancer. Additionally, a number of cancers have amplifications in the SEC62 gene, including lung adenocarcinomas, prostate, thyroid, and head and neck squamous cell carcinoma (Greiner et al., 2011, Jung et al., 2006, Linxweiler et al., 2016, Linxweiler et al., 2012, Wemmert et al., 2016, Weng et al., 2012). These studies suggest that SEC62 plays a role in tumor cell invasion and migration in addition to aiding in resistance to anti-cancer ER stress (Peng et al., 2018).

Due to the critical role of ER stress in cancer, promoting even greater amounts of ER stress could be a potential therapeutic strategy for killing tumors (Wang and Kaufman, 2014). The proteasome inhibitor bortezomib was the first FDA-approved drug for cancer therapy that functions as an inducer of ER stress (Takenokuchi et al., 2015). One could also target specific components of the protein-folding or UPR pathways, such as PERK, IRE1α, HSP90, GRP94, and BiP, thereby augmenting ER stress and sensitize cancer cells to cell death (Wang and Kaufman, 2014). Similarly, inhibition of HSP molecular chaperones leads to a disruption in protein processing and induces ER stress. Increased expression of pro-survival HSP90 and HSP70 has been observed in many types of cancers, and their inhibition has emerged as a promising anti-cancer strategy (Calderwood et al., 2006). Stress-inducible HSP70 was considerably upregulated as a pro-survival response after HSP90 inhibition in vitro, allowing for greater capacity for myeloma cells to fold damaged proteins (Davenport et al., 2007).

The sarcoendoplasmic reticulum Ca2+-ATPase (SERCA) is a major source of Ca2+ transport out of the ER. This release of stored Ca2+ is critical for many cellular processes such as proliferation, metabolism, muscle contraction, gene transcription, and cell death (Berridge et al., 2000). Another method of inducing ER stress to kill cancer cells is by depleting ER Ca2+ through SERCA inhibitor thapsigargin and its prodrugs G202 and 12ADT-Asp (Denmeade et al., 2012, Mahalingam et al., 2016). Similarly, because SEC62 relies on Ca2+ for its function, attempts have been made to treat cancers with Ca2+ sensor and effector protein Calmodulin (CaM) antagonists to prevent Ca2+ efflux into the ER lumen. Indeed, CaM antagonists trifluoperazine (TFP) and ophiobolin A inhibited cell migration and proliferation in cancerous cells with high SEC62 expression and sensitized cancer cells to thapsigargin-induced ER-stress (Greiner et al., 2011, Linxweiler et al., 2016, Linxweiler et al., 2012). Other studies have suggested that ER stress is an indirect response to anti-cancer agents such as cisplatin (Mandic et al., 2002), plant alkaloid ellipticine (Hagg et al., 2004), or doxorubicin (Jang et al., 2004). Furthermore, the glycosylation inhibitors tunicamycin and 2-deoxyglucose can induce ER stress through accumulation of misfolded proteins (Wu et al., 2018, Yu and Kim, 2010). However, because ER stress, either endogenous or treatment-induced, can upregulate ER-phagy (Zhang et al., 2019b, Stephani et al., 2020, Smith et al., 2018, Jiang et al., 2020, Loi et al., 2019), these molecules might need subsequent treatment with ER-phagy or autophagy inhibitors to prevent cancer cells from rescuing themselves from the toxicities of protein misfolding and aggregation. One suggested ER-phagy inhibitor, berberine, enhances expression of various ER stress markers as well as reduces ER localization to LC3 suggesting inhibition of ER-phagy (Zhang et al., 2019a). Interestingly, cancer cells that are dependent on autophagy, become vulnerable to proteasome inhibitors upon loss of autophagy, consistent with the idea that blockade of autophagy (and therefore ER-phagy) can increase sensitivity to agents that cause ER stress through different mechanisms (Towers et al., 2019, Towers and Thorburn, 2019).

Other Potentially Important Types of Selective Autophagy of Organelles in Cancer

Pexophagy

Another important player in cancer cell metabolism is the peroxisome, which plays a vital role in the oxidation of many types of molecules, notably fatty acids and amino acids, that can promote cell growth and survival (De Duve and Baudhuin, 1966, Poirier et al., 2006). These oxidation reactions generate hydrogen peroxide that can damage DNA and proteins, which is then detoxified by antioxidant enzyme catalase (Till et al., 2012, Fransen et al., 2012). There are two stimuli that promote pexophagy: nutrient starvation and ROS (Sargent et al., 2016, Zhang et al., 2015, Lee et al., 2018). Although few studies have shown the specific effects of peroxisomes in cancer, their increased activity could contribute to tumor growth through lipid oxidation (Carracedo et al., 2013, Santos and Schulze, 2012, Liu et al., 2017). Peroxisome proliferator-activated receptors (PPARs) are involved in regulating the expression of genes involved in peroxisome metabolism, such as Acetyl-CoA Oxidase 1 (ACOX1) (Hostetler et al., 2006). ACOX1 is upregulated to aid in the oxidation of hepatic fatty acids and results in an increase of H2O2 and subsequent tumor development (Misra and Reddy, 2014, Yeldandi et al., 2000). As such, high ACOX1 levels are associated with poor survival rate in HER2 positive breast cancers (Kim et al., 2015). Alpha-methylacyl-CoA Racemase (AMACR) is another possible pro-tumorigenic PPAR-regulated protein involved in alpha oxidation of molecules to prepare them for beta-oxidation. Elevation of AMACR is associated with poor prognosis in a variety of cancers, including prostate (Box et al., 2016, Lloyd et al., 2008), colon (Jiang et al., 2003, Chen et al., 2005), gastric (Jindal et al., 2016), breast (Witkiewicz et al., 2005), renal and hepatocellular (Went et al., 2006), and in myxofibrosarcomas (Li et al., 2014).

Because peroxisomes provide cancer cells with vital molecules for survival and their activity is increased in response to nutrient or oxidative stress, promoting pexophagy is a potential therapeutic strategy for cancer. While there are no current therapeutics, or even genetic or other experimental strategies, that can directly promote pexophagy in a specific manner, oral treatment with lipolytic agent 3,5-dimethylpyrazole or 5-carboxy-2-methylpyrazine 1-oxide (Acipimox) can reduce both mitochondrial and peroxisome beta-oxidation (Locci Cubeddu et al., 1985), and therefore could be a useful agent to treat cancer. Hypolipidemic drugs, phthalate esters (Yokota and Dariush Fahimi, 2009), or non-classical peroxisome proliferators, such as 4-phenylbutyrate (Sexton et al., 2010) might also indirectly induce pexophagy.

Lipophagy

Lipids are the major component of biological membranes and are also used for energy storage, production, and cellular signaling. Intracellularly, lipids are stored in lipid droplets when energy supplies are high. Degradation of lipid droplets can occur in response to starvation to help meet energy needs of the cell. Lipolysis and lipophagy (autophagic degradation of lipids by acidic lipases) are two pathways the cell uses to metabolize these stored fats (Singh et al., 2009, Zechner et al., 2017). Once lipid droplets are catabolized to free fatty acids, they are broken down through mitochondrial fatty acid β-oxidation pathway to meet the energy requirements of rapidly proliferating cells (Carracedo et al., 2013).

Lipophagy, like mitophagy and the other forms of selective autophagy or organelles, could potentially be both tumor promoting and tumor suppressive. Cancer cells often upregulate fatty acid synthesis and uptake, leading to increased accumulation of lipid droplets (Menard et al., 2016, Tirinato et al., 2015). Lipophagy provides cancer cells with lipid metabolites for synthesis of biomolecules under stressed or nutrient-deprived conditions and this may aid in survival (Gomez de Cedron and Ramirez de Molina, 2016). For example, lipophagy-mediated production of free fatty acids aids and subsequent ROS production promotes senescence in prostate cancer. Senescence is reversed through inhibition of Lysosomal acid lipase (LAL) (Panda et al., 2020). Studies have also linked reduced lipophagy, i.e. lipid droplet accumulation, with increased cancer aggressiveness (Nieva et al., 2012, Du et al., 2015, Kaini et al., 2012) and chemotherapy resistance (Rak et al., 2014). LAL deficiency results in an abnormal hematopoiesis leading to an abundance of immature myeloid-derived suppressor cells, which mediate suppression of immune surveillance and promote tumor evasion of host immunity to stimulate tumorigenesis and metastasis (Qu et al., 2009, Zhao et al., 2015). Conversely, expression of LAL and subsequent enhanced lipid metabolism has also been reported to reduce metastasis in lung and liver cancer (Zhao et al., 2016, Du et al., 2015). Another recent study reported that increased lipophagy mediates ER stress through free fatty acid accumulation to sensitize cancer cells to apoptosis (Mukhopadhyay et al., 2017). Involvement of lipophagy in lipid turnover makes it a crucial player in cancer survival and metastasis. Therefore, targeting this autophagic pathway could be another therapeutic strategy for a variety of cancers.

Concluding Remarks

As is the case with general autophagy, there is a complex relationship between organelle-targeted autophagy and cancer cell growth, survival, or progression. Although we have focused on selective autophagy of the mitochondria, ER, lysosomes, peroxisomes, and lipid droplets, other organelles and parts of organelles can also be targeted for autophagic turnover and may contribute to cancer-related phenotypes. While we typically think that autophagy of organelles is important in cancer through the removal of toxic and/or damaged molecules, there are situations, such as in mitophagy, where increased degradation of these organelles could be both advantageous and detrimental to the cell. However, it is difficult to be certain of many conclusions in the literature that have suggested a particular role for a certain type of organelle-specific autophagy because we lack effective methods to positively or negatively manipulate these processes without also affecting general autophagy. However, despite these limitations and complexities, it is very likely that selective turnover of organelles is critical in cancer cells.

If we were able to manipulate these forms of selective autophagy, it might be feasible to use that approach as a way to better treat or prevent cancer. However, many questions remain regarding organelle homeostasis in cancer. Can we understand the context-dependent functions of organelle homeostasis and design reliable and effective ways to selectively manipulate these processes in experimental models of cancer? An even bigger challenge is to find ways to design interventions that could realistically be applied therapeutically since this would require the development of drugs that can achieve these goals. These problems are difficult because of the nature of evolved biological systems. First, cancer cells adapt in response to selective pressure. Thus, if a cancer cell adapts to perform cellular functions in a different way, this may stymie efforts to interfere with the process that was thought to be critically important. For example, although we believe that mitophagy is critical in cancer growth, if cancer cells can maintain healthy mitochondria through other methods that do not involve autophagy at all, they may be able to circumvent even highly effective methods of mitophagy inhibition. Second, biology often uses different molecules to achieve the same purpose while conversely using the same machinery for different purposes. Thus, if we target one ACR that is critical for a particular type of organelle-phagy, another may take its place. Beyond their role in organelle homeostasis, ACRs are also vital to promoting cancer cell growth and survival. For example, p62 accumulation results in alteration in NFᴋB and NRF2 signaling, which promotes tumor growth through increased proteosome activation as well as enhanced oxidative stress and DNA damage response (Towers et al., 2019, Mathew et al., 2009, Wei et al., 2014). Another example is that accumulation of the autophagy receptorNBR1 enhances breast cancer metastatic growth through an incompletely understood autophagy-independent mechanism (Marsh et al., 2020). Disrupting organelle homeostasis via autophagy inhibition may therefore have unintended consequence, such as accumulation of ACRs, which may then promote tumor signaling via alternate methods. It is also unclear how organelle-specific autophagy pathways intersect with other organelle functions, such as serving as signaling hubs. Such problems cause practical problems in experimental interpretation. However, this complexity also opens up potential opportunities to refine and improve interventions. For example, by interfering with autophagy to alter organelle turnover, it may be possible to manipulate signaling events that take place at these organelles. By better understanding such mechanisms as they relate to cancer cell behaviors, it may be feasible to develop more rational approaches for therapy.

Acknowledgements

This work was supported by the National Institutes of Health under Ruth L. Kirschstein National Research Service Award T32CA190216-04, F32 CA250129 and R01s CA150925 and CA190170. Its contents are solely the responsibility of the authors and do not necessarily represent the official views of the NIH.

Footnotes

Declaration of Interests

The authors declare no competing interests.

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

References

  1. ABDRAKHMANOV A, KULIKOV AV, LUCHKINA EA, ZHIVOTOVSKY B & GOGVADZE V 2019. Involvement of mitophagy in cisplatin-induced cell death regulation. Biol Chem, 400, 161–170. [DOI] [PubMed] [Google Scholar]
  2. AITS S & JAATTELA M 2013. Lysosomal cell death at a glance. J Cell Sci, 126, 1905–12. [DOI] [PubMed] [Google Scholar]
  3. AMARAVADI RK, KIMMELMAN AC & DEBNATH J 2019. Targeting Autophagy in Cancer: Recent Advances and Future Directions. Cancer Discov, 9, 1167–1181. [DOI] [PMC free article] [PubMed] [Google Scholar]
  4. AN H, ORDUREAU A, PAULO JA, SHOEMAKER CJ, DENIC V & HARPER JW 2019. TEX264 Is an Endoplasmic Reticulum-Resident ATG8-Interacting Protein Critical for ER Remodeling during Nutrient Stress. Mol Cell, 74, 891–908 e10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  5. ANDING AL & BAEHRECKE EH 2017. Cleaning House: Selective Autophagy of Organelles. Dev Cell, 41, 10–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  6. ANTONUCCI L, FAGMAN JB, KIM JY, TODORIC J, GUKOVSKY I, MACKEY M, ELLISMAN MH & KARIN M 2015. Basal autophagy maintains pancreatic acinar cell homeostasis and protein synthesis and prevents ER stress. Proc Natl Acad Sci U S A, 112, E6166–74. [DOI] [PMC free article] [PubMed] [Google Scholar]
  7. BERRIDGE MJ, LIPP P & BOOTMAN MD 2000. The versatility and universality of calcium signalling. Nat Rev Mol Cell Biol, 1, 11–21. [DOI] [PubMed] [Google Scholar]
  8. BOX A, ALSHALALFA M, HEGAZY SA, DONNELLY B & BISMAR TA 2016. High alpha-methylacyl-CoA racemase (AMACR) is associated with ERG expression and with adverse clinical outcome in patients with localized prostate cancer. Tumour Biol, 37, 12287–12299. [DOI] [PubMed] [Google Scholar]
  9. BROKER LE, HUISMAN C, SPAN SW, RODRIGUEZ JA, KRUYT FA & GIACCONE G 2004. Cathepsin B mediates caspase-independent cell death induced by microtubule stabilizing agents in non-small cell lung cancer cells. Cancer Res, 64, 27–30. [DOI] [PubMed] [Google Scholar]
  10. CALDERWOOD SK, KHALEQUE MA, SAWYER DB & CIOCCA DR 2006. Heat shock proteins in cancer: chaperones of tumorigenesis. Trends Biochem Sci, 31, 164–72. [DOI] [PubMed] [Google Scholar]
  11. CAO K & TAIT SWG 2018. Apoptosis and Cancer: Force Awakens, Phantom Menace, or Both? Int Rev Cell Mol Biol, 337, 135–152. [DOI] [PubMed] [Google Scholar]
  12. CARRACEDO A, CANTLEY LC & PANDOLFI PP 2013. Cancer metabolism: fatty acid oxidation in the limelight. Nat Rev Cancer, 13, 227–32. [DOI] [PMC free article] [PubMed] [Google Scholar]
  13. CHANG JY, YI HS, KIM HW & SHONG M 2017. Dysregulation of mitophagy in carcinogenesis and tumor progression. Biochim Biophys Acta Bioenerg, 1858, 633–640. [DOI] [PubMed] [Google Scholar]
  14. CHAUHAN S, KUMAR S, JAIN A, PONPUAK M, MUDD MH, KIMURA T, CHOI SW, PETERS R, MANDELL M, BRUUN JA, JOHANSEN T & DERETIC V 2016. TRIMs and Galectins Globally Cooperate and TRIM16 and Galectin-3 Co-direct Autophagy in Endomembrane Damage Homeostasis. Dev Cell, 39, 13–27. [DOI] [PMC free article] [PubMed] [Google Scholar]
  15. CHEN M, CHEN Z, WANG Y, TAN Z, ZHU C, LI Y, HAN Z, CHEN L, GAO R, LIU L & CHEN Q 2016. Mitophagy receptor FUNDC1 regulates mitochondrial dynamics and mitophagy. Autophagy, 12, 689–702. [DOI] [PMC free article] [PubMed] [Google Scholar]
  16. CHEN Q, XIAO Y, CHAI P, ZHENG P, TENG J & CHEN J 2019. ATL3 Is a Tubular ER-Phagy Receptor for GABARAP-Mediated Selective Autophagy. Curr Biol, 29, 846–855 e6. [DOI] [PubMed] [Google Scholar]
  17. CHEN ZM, RITTER JH & WANG HL 2005. Differential expression of alpha-methylacyl coenzyme A racemase in adenocarcinomas of the small and large intestines. Am J Surg Pathol, 29, 890–6. [DOI] [PubMed] [Google Scholar]
  18. CHINO H, HATTA T, NATSUME T & MIZUSHIMA N 2019. Intrinsically Disordered Protein TEX264 Mediates ER-phagy. Mol Cell, 74, 909–921 e6. [DOI] [PubMed] [Google Scholar]
  19. CHOURASIA AH, TRACY K, FRANKENBERGER C, BOLAND ML, SHARIFI MN, DRAKE LE, SACHLEBEN JR, ASARA JM, LOCASALE JW, KARCZMAR GS & MACLEOD KF 2015. Mitophagy defects arising from BNip3 loss promote mammary tumor progression to metastasis. EMBO Rep, 16, 1145–63. [DOI] [PMC free article] [PubMed] [Google Scholar]
  20. COLELL A, RICCI JE, TAIT S, MILASTA S, MAURER U, BOUCHIER-HAYES L, FITZGERALD P, GUIO-CARRION A, WATERHOUSE NJ, LI CW, MARI B, BARBRY P, NEWMEYER DD, BEERE HM & GREEN DR 2007. GAPDH and autophagy preserve survival after apoptotic cytochrome c release in the absence of caspase activation. Cell, 129, 983–97. [DOI] [PubMed] [Google Scholar]
  21. COMMISSO C, DAVIDSON SM, SOYDANER-AZELOGLU RG, PARKER SJ, KAMPHORST JJ, HACKETT S, GRABOCKA E, NOFAL M, DREBIN JA, THOMPSON CB, RABINOWITZ JD, METALLO CM, VANDER HEIDEN MG & BAR-SAGI D 2013. Macropinocytosis of protein is an amino acid supply route in Ras-transformed cells. Nature, 497, 633–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  22. DAVENPORT EL, MOORE HE, DUNLOP AS, SHARP SY, WORKMAN P, MORGAN GJ & DAVIES FE 2007. Heat shock protein inhibition is associated with activation of the unfolded protein response pathway in myeloma plasma cells. Blood, 110, 2641–9. [DOI] [PubMed] [Google Scholar]
  23. DE DUVE C & BAUDHUIN P 1966. Peroxisomes (microbodies and related particles). Physiol Rev, 46, 323–57. [DOI] [PubMed] [Google Scholar]
  24. DENMEADE SR, MHAKA AM, ROSEN DM, BRENNEN WN, DALRYMPLE S, DACH I, OLESEN C, GUREL B, DEMARZO AM, WILDING G, CARDUCCI MA, DIONNE CA, MOLLER JV, NISSEN P, CHRISTENSEN SB & ISAACS JT 2012. Engineering a prostate-specific membrane antigen-activated tumor endothelial cell prodrug for cancer therapy. Sci Transl Med, 4, 140ra86. [DOI] [PMC free article] [PubMed] [Google Scholar]
  25. DI RITA A, PESCHIAROLI A, P DA, STROBBE D, HU Z, GRUBER J, NYGAARD M, LAMBRUGHI M, MELINO G, PAPALEO E, DENGJEL J, EL ALAOUI S, CAMPANELLA M, DOTSCH V, ROGOV VV, STRAPPAZZON F & CECCONI F 2018. HUWE1 E3 ligase promotes PINK1/PARKIN-independent mitophagy by regulating AMBRA1 activation via IKKalpha. Nat Commun, 9, 3755. [DOI] [PMC free article] [PubMed] [Google Scholar]
  26. DIKIC I & ELAZAR Z 2018. Mechanism and medical implications of mammalian autophagy. Nat Rev Mol Cell Biol, 19, 349–364. [DOI] [PubMed] [Google Scholar]
  27. DOU Z, XU C, DONAHUE G, SHIMI T, PAN JA, ZHU J, IVANOV A, CAPELL BC, DRAKE AM, SHAH PP, CATANZARO JM, RICKETTS MD, LAMARK T, ADAM SA, MARMORSTEIN R, ZONG WX, JOHANSEN T, GOLDMAN RD, ADAMS PD & BERGER SL 2015. Autophagy mediates degradation of nuclear lamina. Nature, 527, 105–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  28. DRAKE LE, SPRINGER MZ, POOLE LP, KIM CJ & MACLEOD KF 2017. Expanding perspectives on the significance of mitophagy in cancer. Semin Cancer Biol, 47, 110–124. [DOI] [PMC free article] [PubMed] [Google Scholar]
  29. DU H, ZHAO T, DING X & YAN C 2015. Hepatocyte-Specific Expression of Human Lysosome Acid Lipase Corrects Liver Inflammation and Tumor Metastasis in lal(−/−) Mice. Am J Pathol, 185, 2379–89. [DOI] [PMC free article] [PubMed] [Google Scholar]
  30. ERDAL H, BERNDTSSON M, CASTRO J, BRUNK U, SHOSHAN MC & LINDER S 2005. Induction of lysosomal membrane permeabilization by compounds that activate p53-independent apoptosis. Proc Natl Acad Sci U S A, 102, 192–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  31. FEHRENBACHER N, GYRD-HANSEN M, POULSEN B, FELBOR U, KALLUNKI T, BOES M, WEBER E, LEIST M & JAATTELA M 2004. Sensitization to the lysosomal cell death pathway upon immortalization and transformation. Cancer Res, 64, 5301–10. [DOI] [PubMed] [Google Scholar]
  32. FITZWALTER BE, TOWERS CG, SULLIVAN KD, ANDRYSIK Z, HOH M, LUDWIG M, O’PREY J, RYAN KM, ESPINOSA JM, MORGAN MJ & THORBURN A 2018. Autophagy Inhibition Mediates Apoptosis Sensitization in Cancer Therapy by Relieving FOXO3a Turnover. Dev Cell, 44, 555–565 e3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  33. FRANSEN M, NORDGREN M, WANG B & APANASETS O 2012. Role of peroxisomes in ROS/RNS-metabolism: implications for human disease. Biochim Biophys Acta, 1822, 1363–73. [DOI] [PubMed] [Google Scholar]
  34. FRIEDMAN JR & VOELTZ GK 2011. The ER in 3D: a multifunctional dynamic membrane network. Trends Cell Biol, 21, 709–17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  35. FUJIWARA M, MARUSAWA H, WANG HQ, IWAI A, IKEUCHI K, IMAI Y, KATAOKA A, NUKINA N, TAKAHASHI R & CHIBA T 2008. Parkin as a tumor suppressor gene for hepatocellular carcinoma. Oncogene, 27, 6002–11. [DOI] [PubMed] [Google Scholar]
  36. FUMAGALLI F, NOACK J, BERGMANN TJ, CEBOLLERO E, PISONI GB, FASANA E, FREGNO I, GALLI C, LOI M, SOLDA T, D’ANTUONO R, RAIMONDI A, JUNG M, MELNYK A, SCHORR S, SCHREIBER A, SIMONELLI L, VARANI L, WILSON-ZBINDEN C, ZERBE O, HOFMANN K, PETER M, QUADRONI M, ZIMMERMANN R & MOLINARI M 2016. Translocon component Sec62 acts in endoplasmic reticulum turnover during stress recovery. Nat Cell Biol, 18, 1173–1184. [DOI] [PubMed] [Google Scholar]
  37. GALLUZZI L & GREEN DR 2019. Autophagy-Independent Functions of the Autophagy Machinery. Cell, 177, 1682–1699. [DOI] [PMC free article] [PubMed] [Google Scholar]
  38. GALLUZZI L, LAROCHETTE N, ZAMZAMI N & KROEMER G 2006. Mitochondria as therapeutic targets for cancer chemotherapy. Oncogene, 25, 4812–30. [DOI] [PubMed] [Google Scholar]
  39. GALLUZZI L, PIETROCOLA F, BRAVO-SAN PEDRO JM, AMARAVADI RK, BAEHRECKE EH, CECCONI F, CODOGNO P, DEBNATH J, GEWIRTZ DA, KARANTZA V, KIMMELMAN A, KUMAR S, LEVINE B, MAIURI MC, MARTIN SJ, PENNINGER J, PIACENTINI M, RUBINSZTEIN DC, SIMON HU, SIMONSEN A, THORBURN AM, VELASCO G, RYAN KM & KROEMER G 2015. Autophagy in malignant transformation and cancer progression. EMBO J, 34, 856–80. [DOI] [PMC free article] [PubMed] [Google Scholar]
  40. GIAMPAZOLIAS E & TAIT SW 2016. Mitochondria and the hallmarks of cancer. FEBS J, 283, 803–14. [DOI] [PubMed] [Google Scholar]
  41. GOCHEVA V, ZENG W, KE D, KLIMSTRA D, REINHECKEL T, PETERS C, HANAHAN D & JOYCE JA 2006. Distinct roles for cysteine cathepsin genes in multistage tumorigenesis. Genes Dev, 20, 543–56. [DOI] [PMC free article] [PubMed] [Google Scholar]
  42. GOMEZ DE CEDRON M & RAMIREZ DE MOLINA A 2016. Microtargeting cancer metabolism: opening new therapeutic windows based on lipid metabolism. J Lipid Res, 57, 193–206. [DOI] [PMC free article] [PubMed] [Google Scholar]
  43. GREINER M, KREUTZER B, JUNG V, GROBHOLZ R, HASENFUS A, STOHR RF, TORNILLO L, DUDEK J, STOCKLE M, UNTEREGGER G, KAMRADT J, WULLICH B & ZIMMERMANN R 2011. Silencing of the SEC62 gene inhibits migratory and invasive potential of various tumor cells. Int J Cancer, 128, 2284–95. [DOI] [PubMed] [Google Scholar]
  44. GROTH-PEDERSEN L & JAATTELA M 2013. Combating apoptosis and multidrug resistant cancers by targeting lysosomes. Cancer Lett, 332, 265–74. [DOI] [PubMed] [Google Scholar]
  45. GROTH-PEDERSEN L, OSTENFELD MS, HOYER-HANSEN M, NYLANDSTED J & JAATTELA M 2007. Vincristine induces dramatic lysosomal changes and sensitizes cancer cells to lysosome-destabilizing siramesine. Cancer Res, 67, 2217–25. [DOI] [PubMed] [Google Scholar]
  46. GRUMATI P, MOROZZI G, HOLPER S, MARI M, HARWARDT MI, YAN R, MULLER S, REGGIORI F, HEILEMANN M & DIKIC I 2017. Full length RTN3 regulates turnover of tubular endoplasmic reticulum via selective autophagy. Elife, 6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  47. GUO JY, CHEN HY, MATHEW R, FAN J, STROHECKER AM, KARSLI-UZUNBAS G, KAMPHORST JJ, CHEN G, LEMONS JM, KARANTZA V, COLLER HA, DIPAOLA RS, GELINAS C, RABINOWITZ JD & WHITE E 2011. Activated Ras requires autophagy to maintain oxidative metabolism and tumorigenesis. Genes Dev, 25, 460–70. [DOI] [PMC free article] [PubMed] [Google Scholar]
  48. GUO JY, KARSLI-UZUNBAS G, MATHEW R, AISNER SC, KAMPHORST JJ, STROHECKER AM, CHEN G, PRICE S, LU W, TENG X, SNYDER E, SANTANAM U, DIPAOLA RS, JACKS T, RABINOWITZ JD & WHITE E 2013. Autophagy suppresses progression of K-ras-induced lung tumors to oncocytomas and maintains lipid homeostasis. Genes Dev, 27, 1447–61. [DOI] [PMC free article] [PubMed] [Google Scholar]
  49. HAGG M, BERNDTSSON M, MANDIC A, ZHOU R, SHOSHAN MC & LINDER S 2004. Induction of endoplasmic reticulum stress by ellipticine plant alkaloids. Mol Cancer Ther, 3, 489–97. [PubMed] [Google Scholar]
  50. HANNA RA, QUINSAY MN, OROGO AM, GIANG K, RIKKA S & GUSTAFSSON AB 2012. Microtubule-associated protein 1 light chain 3 (LC3) interacts with Bnip3 protein to selectively remove endoplasmic reticulum and mitochondria via autophagy. J Biol Chem, 287, 19094–104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  51. HEO JM, ORDUREAU A, PAULO JA, RINEHART J & HARPER JW 2015. The PINK1-PARKIN Mitochondrial Ubiquitylation Pathway Drives a Program of OPTN/NDP52 Recruitment and TBK1 Activation to Promote Mitophagy. Mol Cell, 60, 7–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  52. HOSTETLER HA, KIER AB & SCHROEDER F 2006. Very-long-chain and branched-chain fatty acyl-CoAs are high affinity ligands for the peroxisome proliferator-activated receptor alpha (PPARalpha). Biochemistry, 45, 7669–81. [DOI] [PMC free article] [PubMed] [Google Scholar]
  53. HUNG YH, CHEN LM, YANG JY & YANG WY 2013. Spatiotemporally controlled induction of autophagy-mediated lysosome turnover. Nat Commun, 4, 2111. [DOI] [PubMed] [Google Scholar]
  54. ISLAM F, GOPALAN V & LAM AK 2018. RETREG1 (FAM134B): A new player in human diseases: 15 years after the discovery in cancer. J Cell Physiol, 233, 4479–4489. [DOI] [PubMed] [Google Scholar]
  55. JANG YM, KENDAIAH S, DREW B, PHILLIPS T, SELMAN C, JULIAN D & LEEUWENBURGH C 2004. Doxorubicin treatment in vivo activates caspase-12 mediated cardiac apoptosis in both male and female rats. FEBS Lett, 577, 483–90. [DOI] [PubMed] [Google Scholar]
  56. JIANG X, WANG X, DING X, DU M, LI B, WENG X, ZHANG J, LI L, TIAN R, ZHU Q, CHEN S, WANG L, LIU W, FANG L, NECULAI D & SUN Q 2020. FAM134B oligomerization drives endoplasmic reticulum membrane scission for ER-phagy. EMBO J, 39, e102608. [DOI] [PMC free article] [PubMed] [Google Scholar]
  57. JIANG Z, FANGER GR, BANNER BF, WODA BA, ALGATE P, DRESSER K, XU J, REED SG, ROCK KL & CHU PG 2003. A dietary enzyme: alpha-methylacyl-CoA racemase/P504S is overexpressed in colon carcinoma. Cancer Detect Prev, 27, 422–6. [DOI] [PubMed] [Google Scholar]
  58. JIN SM, LAZAROU M, WANG C, KANE LA, NARENDRA DP & YOULE RJ 2010. Mitochondrial membrane potential regulates PINK1 import and proteolytic destabilization by PARL. J Cell Biol, 191, 933–42. [DOI] [PMC free article] [PubMed] [Google Scholar]
  59. JINDAL Y, SINGH A, KUMAR R, VARMA K, MISRA V, MISRA SP & DWIVEDI M 2016. Expression of Alpha Methylacyl CoA Racemase (AMACR) in Gastric Adenocarcinoma and Its Correlation with Helicobacter pylori Infection. J Clin Diagn Res, 10, EC10–EC12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  60. JOSHI S, TOLKUNOV D, AVIV H, HAKIMI AA, YAO M, HSIEH JJ, GANESAN S, CHAN CS & WHITE E 2015. The Genomic Landscape of Renal Oncocytoma Identifies a Metabolic Barrier to Tumorigenesis. Cell Rep, 13, 1895–908. [DOI] [PMC free article] [PubMed] [Google Scholar]
  61. JUNG V, KINDICH R, KAMRADT J, JUNG M, MULLER M, SCHULZ WA, ENGERS R, UNTEREGGER G, STOCKLE M, ZIMMERMANN R & WULLICH B 2006. Genomic and expression analysis of the 3q25-q26 amplification unit reveals TLOC1/SEC62 as a probable target gene in prostate cancer. Mol Cancer Res, 4, 169–76. [DOI] [PubMed] [Google Scholar]
  62. KAINI RR, SILLERUD LO, ZHAORIGETU S & HU CA 2012. Autophagy regulates lipolysis and cell survival through lipid droplet degradation in androgen-sensitive prostate cancer cells. Prostate, 72, 1412–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  63. KAMPHORST JJ, NOFAL M, COMMISSO C, HACKETT SR, LU W, GRABOCKA E, VANDER HEIDEN MG, MILLER G, DREBIN JA, BAR-SAGI D, THOMPSON CB & RABINOWITZ JD 2015. Human pancreatic cancer tumors are nutrient poor and tumor cells actively scavenge extracellular protein. Cancer Res, 75, 544–53. [DOI] [PMC free article] [PubMed] [Google Scholar]
  64. KASEM K, GOPALAN V, SALAJEGHEH A, LU CT, SMITH RA & LAM AK 2014a. JK1 (FAM134B) gene and colorectal cancer: a pilot study on the gene copy number alterations and correlations with clinicopathological parameters. Exp Mol Pathol, 97, 31–6. [DOI] [PubMed] [Google Scholar]
  65. KASEM K, GOPALAN V, SALAJEGHEH A, LU CT, SMITH RA & LAM AK 2014b. The roles of JK-1 (FAM134B) expressions in colorectal cancer. Exp Cell Res, 326, 166–73. [DOI] [PubMed] [Google Scholar]
  66. KASEM K, SULLIVAN E, GOPALAN V, SALAJEGHEH A, SMITH RA & LAM AK 2014c. JK1 (FAM134B) represses cell migration in colon cancer: a functional study of a novel gene. Exp Mol Pathol, 97, 99–104. [DOI] [PubMed] [Google Scholar]
  67. KAUSHIK S & CUERVO AM 2018. The coming of age of chaperone-mediated autophagy. Nat Rev Mol Cell Biol, 19, 365–381. [DOI] [PMC free article] [PubMed] [Google Scholar]
  68. KHAMINETS A, HEINRICH T, MARI M, GRUMATI P, HUEBNER AK, AKUTSU M, LIEBMANN L, STOLZ A, NIETZSCHE S, KOCH N, MAUTHE M, KATONA I, QUALMANN B, WEIS J, REGGIORI F, KURTH I, HUBNER CA & DIKIC I 2015. Regulation of endoplasmic reticulum turnover by selective autophagy. Nature, 522, 354–8. [DOI] [PubMed] [Google Scholar]
  69. KIM JH, KIM HY, LEE YK, YOON YS, XU WG, YOON JK, CHOI SE, KO YG, KIM MJ, LEE SJ, WANG HJ & YOON G 2011. Involvement of mitophagy in oncogenic K-Ras-induced transformation: overcoming a cellular energy deficit from glucose deficiency. Autophagy, 7, 1187–98. [DOI] [PMC free article] [PubMed] [Google Scholar]
  70. KIM PK, HAILEY DW, MULLEN RT & LIPPINCOTT-SCHWARTZ J 2008. Ubiquitin signals autophagic degradation of cytosolic proteins and peroxisomes. Proc Natl Acad Sci U S A, 105, 20567–74. [DOI] [PMC free article] [PubMed] [Google Scholar]
  71. KIM S, LEE Y & KOO JS 2015. Differential expression of lipid metabolism-related proteins in different breast cancer subtypes. PLoS One, 10, e0119473. [DOI] [PMC free article] [PubMed] [Google Scholar]
  72. KOERVER L, PAPADOPOULOS C, LIU B, KRAVIC B, ROTA G, BRECHT L, VEENENDAAL T, POLAJNAR M, BLUEMKE A, EHRMANN M, KLUMPERMAN J, JAATTELA M, BEHRENDS C & MEYER H 2019. The ubiquitin-conjugating enzyme UBE2QL1 coordinates lysophagy in response to endolysosomal damage. EMBO Rep, 20, e48014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  73. KOSTENKO EV, OLABISI OO, SAHAY S, RODRIGUEZ PL & WHITEHEAD IP 2006. Ccpg1, a novel scaffold protein that regulates the activity of the Rho guanine nucleotide exchange factor Dbs. Mol Cell Biol, 26, 8964–75. [DOI] [PMC free article] [PubMed] [Google Scholar]
  74. KOYANO F, OKATSU K, KOSAKO H, TAMURA Y, GO E, KIMURA M, KIMURA Y, TSUCHIYA H, YOSHIHARA H, HIROKAWA T, ENDO T, FON EA, TREMPE JF, SAEKI Y, TANAKA K & MATSUDA N 2014. Ubiquitin is phosphorylated by PINK1 to activate parkin. Nature, 510, 162–6. [DOI] [PubMed] [Google Scholar]
  75. KRAVIC B, BEHRENDS C & MEYER H 2020. Regulation of lysosome integrity and lysophagy by the ubiquitin-conjugating enzyme UBE2QL1. Autophagy, 16, 179–180. [DOI] [PMC free article] [PubMed] [Google Scholar]
  76. LAZAROU M, SLITER DA, KANE LA, SARRAF SA, WANG C, BURMAN JL, SIDERIS DP, FOGEL AI & YOULE RJ 2015. The ubiquitin kinase PINK1 recruits autophagy receptors to induce mitophagy. Nature, 524, 309–314. [DOI] [PMC free article] [PubMed] [Google Scholar]
  77. LEBOVITZ CB, ROBERTSON AG, GOYA R, JONES SJ, MORIN RD, MARRA MA & GORSKI SM 2015. Cross-cancer profiling of molecular alterations within the human autophagy interaction network. Autophagy, 11, 1668–87. [DOI] [PMC free article] [PubMed] [Google Scholar]
  78. LEE JN, DUTTA RK, MAHARJAN Y, LIU ZQ, LIM JY, KIM SJ, CHO DH, SO HS, CHOE SK & PARK R 2018. Catalase inhibition induces pexophagy through ROS accumulation. Biochem Biophys Res Commun, 501, 696–702. [DOI] [PubMed] [Google Scholar]
  79. LEMASTERS JJ 2005. Selective mitochondrial autophagy, or mitophagy, as a targeted defense against oxidative stress, mitochondrial dysfunction, and aging. Rejuvenation Res, 8, 3–5. [DOI] [PubMed] [Google Scholar]
  80. LEVINE B & KROEMER G 2019. Biological Functions of Autophagy Genes: A Disease Perspective. Cell, 176, 11–42. [DOI] [PMC free article] [PubMed] [Google Scholar]
  81. LI C, ZHANG Y, CHENG X, YUAN H, ZHU S, LIU J, WEN Q, XIE Y, LIU J, KROEMER G, KLIONSKY DJ, LOTZE MT, ZEH HJ, KANG R & TANG D 2018. PINK1 and PARK2 Suppress Pancreatic Tumorigenesis through Control of Mitochondrial Iron-Mediated Immunometabolism. Dev Cell, 46, 441–455 e8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  82. LI CF, FANG FM, LAN J, WANG JW, KUNG HJ, CHEN LT, CHEN TJ, LI SH, WANG YH, TAI HC, YU SC & HUANG HY 2014. AMACR amplification in myxofibrosarcomas: a mechanism of overexpression that promotes cell proliferation with therapeutic relevance. Clin Cancer Res, 20, 6141–52. [DOI] [PubMed] [Google Scholar]
  83. LINXWEILER M, BOCHEN F, SCHICK B, WEMMERT S, AL KADAH B, GREINER M, HASENFUS A, BOHLE RM, JUHASZ-BOSS I, SOLOMAYER EF & TAKACS ZF 2016. Identification of SEC62 as a potential marker for 3q amplification and cellular migration in dysplastic cervical lesions. BMC Cancer, 16, 676. [DOI] [PMC free article] [PubMed] [Google Scholar]
  84. LINXWEILER M, LINXWEILER J, BARTH M, BENEDIX J, JUNG V, KIM YJ, BOHLE RM, ZIMMERMANN R & GREINER M 2012. Sec62 bridges the gap from 3q amplification to molecular cell biology in non-small cell lung cancer. Am J Pathol, 180, 473–83. [DOI] [PubMed] [Google Scholar]
  85. LIU Q, LUO Q, HALIM A & SONG G 2017. Targeting lipid metabolism of cancer cells: A promising therapeutic strategy for cancer. Cancer Lett, 401, 39–45. [DOI] [PubMed] [Google Scholar]
  86. LLOYD MD, DARLEY DJ, WIERZBICKI AS & THREADGILL MD 2008. Alpha-methylacyl-CoA racemase--an ‘obscure’ metabolic enzyme takes centre stage. FEBS J, 275, 1089–102. [DOI] [PubMed] [Google Scholar]
  87. LOCCI CUBEDDU T, MASIELLO P, POLLERA M & BERGAMINI E 1985. Effects of antilipolytic agents on rat liver peroxisomes and peroxisomal oxidative activities. Biochim Biophys Acta, 839, 96–104. [DOI] [PubMed] [Google Scholar]
  88. LOI M, RAIMONDI A, MORONE D & MOLINARI M 2019. ESCRT-III-driven piecemeal micro-ER-phagy remodels the ER during recovery from ER stress. Nat Commun, 10, 5058. [DOI] [PMC free article] [PubMed] [Google Scholar]
  89. MAEJIMA I, TAKAHASHI A, OMORI H, KIMURA T, TAKABATAKE Y, SAITOH T, YAMAMOTO A, HAMASAKI M, NODA T, ISAKA Y & YOSHIMORI T 2013. Autophagy sequesters damaged lysosomes to control lysosomal biogenesis and kidney injury. EMBO J, 32, 2336–47. [DOI] [PMC free article] [PubMed] [Google Scholar]
  90. MAHALINGAM D, WILDING G, DENMEADE S, SARANTOPOULAS J, COSGROVE D, CETNAR J, AZAD N, BRUCE J, KURMAN M, ALLGOOD VE & CARDUCCI M 2016. Mipsagargin, a novel thapsigargin-based PSMA-activated prodrug: results of a first-in-man phase I clinical trial in patients with refractory, advanced or metastatic solid tumours. Br J Cancer, 114, 986–94. [DOI] [PMC free article] [PubMed] [Google Scholar]
  91. MANDELL MA, JAIN A, ARKO-MENSAH J, CHAUHAN S, KIMURA T, DINKINS C, SILVESTRI G, MUNCH J, KIRCHHOFF F, SIMONSEN A, WEI Y, LEVINE B, JOHANSEN T & DERETIC V 2014. TRIM proteins regulate autophagy and can target autophagic substrates by direct recognition. Dev Cell, 30, 394–409. [DOI] [PMC free article] [PubMed] [Google Scholar]
  92. MANDIC A, VIKTORSSON K, STRANDBERG L, HEIDEN T, HANSSON J, LINDER S & SHOSHAN MC 2002. Calpain-mediated Bid cleavage and calpain-independent Bak modulation: two separate pathways in cisplatin-induced apoptosis. Mol Cell Biol, 22, 3003–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  93. MARSH T, KENIFIC CM, SURESH D, GONZALEZ H, SHAMIR ER, MEI W, TANKKA A, LEIDAL AM, KALAVACHERLA S, WOO K, WERB Z & DEBNATH J 2020. Autophagic Degradation of NBR1 Restricts Metastatic Outgrowth during Mammary Tumor Progression. Dev Cell, 52, 591–604 e6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  94. MATHEW R, KARP CM, BEAUDOIN B, VUONG N, CHEN G, CHEN HY, BRAY K, REDDY A, BHANOT G, GELINAS C, DIPAOLA RS, KARANTZA-WADSWORTH V & WHITE E 2009. Autophagy suppresses tumorigenesis through elimination of p62. Cell, 137, 1062–75. [DOI] [PMC free article] [PubMed] [Google Scholar]
  95. MENARD JA, CHRISTIANSON HC, KUCHARZEWSKA P, BOURSEAU-GUILMAIN E, SVENSSON KJ, LINDQVIST E, INDIRA CHANDRAN V, KJELLEN L, WELINDER C, BENGZON J, JOHANSSON MC & BELTING M 2016. Metastasis Stimulation by Hypoxia and Acidosis-Induced Extracellular Lipid Uptake Is Mediated by Proteoglycan-Dependent Endocytosis. Cancer Res, 76, 4828–40. [DOI] [PubMed] [Google Scholar]
  96. MESZAROS B, TOMPA P, SIMON I & DOSZTANYI Z 2007. Molecular principles of the interactions of disordered proteins. J Mol Biol, 372, 549–61. [DOI] [PubMed] [Google Scholar]
  97. MIJALJICA D, PRESCOTT M & DEVENISH RJ 2011. Microautophagy in mammalian cells: revisiting a 40-year-old conundrum. Autophagy, 7, 673–82. [DOI] [PubMed] [Google Scholar]
  98. MISRA P & REDDY JK 2014. Peroxisome proliferator-activated receptor-alpha activation and excess energy burning in hepatocarcinogenesis. Biochimie, 98, 63–74. [DOI] [PubMed] [Google Scholar]
  99. MUKHOPADHYAY S, SCHLAEPFER IR, BERGMAN BC, PANDA PK, PRAHARAJ PP, NAIK PP, AGARWAL R & BHUTIA SK 2017. ATG14 facilitated lipophagy in cancer cells induce ER stress mediated mitoptosis through a ROS dependent pathway. Free Radic Biol Med, 104, 199–213. [DOI] [PubMed] [Google Scholar]
  100. MULCAHY LEVY JM & THORBURN A 2020. Autophagy in cancer: moving from understanding mechanism to improving therapy responses in patients. Cell Death Differ, 27, 843–857. [DOI] [PMC free article] [PubMed] [Google Scholar]
  101. MULCAHY LEVY JM, ZAHEDI S, GRIESINGER AM, MORIN A, DAVIES KD, AISNER DL, KLEINSCHMIDT-DEMASTERS BK, FITZWALTER BE, GOODALL ML, THORBURN J, AMANI V, DONSON AM, BIRKS DK, MIRSKY DM, HANKINSON TC, HANDLER MH, GREEN AL, VIBHAKAR R, FOREMAN NK & THORBURN A 2017. Autophagy inhibition overcomes multiple mechanisms of resistance to BRAF inhibition in brain tumors. Elife, 6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  102. NAKAHIRA K, HASPEL JA, RATHINAM VA, LEE SJ, DOLINAY T, LAM HC, ENGLERT JA, RABINOVITCH M, CERNADAS M, KIM HP, FITZGERALD KA, RYTER SW & CHOI AM 2011. Autophagy proteins regulate innate immune responses by inhibiting the release of mitochondrial DNA mediated by the NALP3 inflammasome. Nat Immunol, 12, 222–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
  103. NARENDRA D, TANAKA A, SUEN DF & YOULE RJ 2008. Parkin is recruited selectively to impaired mitochondria and promotes their autophagy. J Cell Biol, 183, 795–803. [DOI] [PMC free article] [PubMed] [Google Scholar]
  104. NARENDRA DP, JIN SM, TANAKA A, SUEN DF, GAUTIER CA, SHEN J, COOKSON MR & YOULE RJ 2010. PINK1 is selectively stabilized on impaired mitochondria to activate Parkin. PLoS Biol, 8, e1000298. [DOI] [PMC free article] [PubMed] [Google Scholar]
  105. NIEVA C, MARRO M, SANTANA-CODINA N, RAO S, PETROV D & SIERRA A 2012. The lipid phenotype of breast cancer cells characterized by Raman microspectroscopy: towards a stratification of malignancy. PLoS One, 7, e46456. [DOI] [PMC free article] [PubMed] [Google Scholar]
  106. NISHIMURA Y, SAMENI M & SLOANE BF 1998. Malignant transformation alters intracellular trafficking of lysosomal cathepsin D in human breast epithelial cells. Pathol Oncol Res, 4, 283–96. [DOI] [PubMed] [Google Scholar]
  107. NIXON-ABELL J, OBARA CJ, WEIGEL AV, LI D, LEGANT WR, XU CS, PASOLLI HA, HARVEY K, HESS HF, BETZIG E, BLACKSTONE C & LIPPINCOTT-SCHWARTZ J 2016. Increased spatiotemporal resolution reveals highly dynamic dense tubular matrices in the peripheral ER. Science, 354. [DOI] [PMC free article] [PubMed] [Google Scholar]
  108. NOVAK I, KIRKIN V, MCEWAN DG, ZHANG J, WILD P, ROZENKNOP A, ROGOV V, LOHR F, POPOVIC D, OCCHIPINTI A, REICHERT AS, TERZIC J, DOTSCH V, NEY PA & DIKIC I 2010. Nix is a selective autophagy receptor for mitochondrial clearance. EMBO Rep, 11, 45–51. [DOI] [PMC free article] [PubMed] [Google Scholar]
  109. NTHIGA TM, KUMAR SHRESTHA B, SJOTTEM E, BRUUN JA, BOWITZ LARSEN K, BHUJABAL Z, LAMARK T & JOHANSEN T 2020. CALCOCO1 acts with VAMP-associated proteins to mediate ER-phagy. EMBO J, 39, e103649. [DOI] [PMC free article] [PubMed] [Google Scholar]
  110. OKATSU K, KOYANO F, KIMURA M, KOSAKO H, SAEKI Y, TANAKA K & MATSUDA N 2015. Phosphorylated ubiquitin chain is the genuine Parkin receptor. J Cell Biol, 209, 111–28. [DOI] [PMC free article] [PubMed] [Google Scholar]
  111. OSTENFELD MS, FEHRENBACHER N, HOYER-HANSEN M, THOMSEN C, FARKAS T & JAATTELA M 2005. Effective tumor cell death by sigma-2 receptor ligand siramesine involves lysosomal leakage and oxidative stress. Cancer Res, 65, 8975–83. [DOI] [PubMed] [Google Scholar]
  112. PANDA PK, PATRA S, NAIK PP, PRAHARAJ PP, MUKHOPADHYAY S, MEHER BR, GUPTA PK, VERMA RS, MAITI TK & BHUTIA SK 2020. Deacetylation of LAMP1 drives lipophagy-dependent generation of free fatty acids by Abrus agglutinin to promote senescence in prostate cancer. J Cell Physiol, 235, 2776–2791. [DOI] [PubMed] [Google Scholar]
  113. PAPADOPOULOS C, KIRCHNER P, BUG M, GRUM D, KOERVER L, SCHULZE N, POEHLER R, DRESSLER A, FENGLER S, ARHZAOUY K, LUX V, EHRMANN M, WEIHL CC & MEYER H 2017. VCP/p97 cooperates with YOD1, UBXD1 and PLAA to drive clearance of ruptured lysosomes by autophagy. EMBO J, 36, 135–150. [DOI] [PMC free article] [PubMed] [Google Scholar]
  114. PENG Y, SHAPIRO SL, BANDUSEELA VC, DIETERICH IA, HEWITT KJ, BRESNICK EH, KONG G, ZHANG J, SCHUELER KL, KELLER MP, ATTIE AD, HACKER TA, SULLIVAN R, KIELAR-GREVSTAD E, ARRIOLA APELO SI, LAMMING DW, ANDERSON RM & PUGLIELLI L 2018. Increased transport of acetyl-CoA into the endoplasmic reticulum causes a progeria-like phenotype. Aging Cell, 17, e12820. [DOI] [PMC free article] [PubMed] [Google Scholar]
  115. PENGO N, SCOLARI M, OLIVA L, MILAN E, MAINOLDI F, RAIMONDI A, FAGIOLI C, MERLINI A, MARIANI E, PASQUALETTO E, ORFANELLI U, PONZONI M, SITIA R, CASOLA S & CENCI S 2013. Plasma cells require autophagy for sustainable immunoglobulin production. Nat Immunol, 14, 298–305. [DOI] [PubMed] [Google Scholar]
  116. PETRUCCELLI LA, DUPERE-RICHER D, PETTERSSON F, RETROUVEY H, SKOULIKAS S & MILLER WH JR. 2011. Vorinostat induces reactive oxygen species and DNA damage in acute myeloid leukemia cells. PLoS One, 6, e20987. [DOI] [PMC free article] [PubMed] [Google Scholar]
  117. PIFFOUX M, ERIAU E & CASSIER PA 2020. Autophagy as a therapeutic target in pancreatic cancer. Br J Cancer. [DOI] [PMC free article] [PubMed] [Google Scholar]
  118. POILLET-PEREZ L & WHITE E 2019. Role of tumor and host autophagy in cancer metabolism. Genes Dev, 33, 610–619. [DOI] [PMC free article] [PubMed] [Google Scholar]
  119. POIRIER Y, ANTONENKOV VD, GLUMOFF T & HILTUNEN JK 2006. Peroxisomal beta-oxidation--a metabolic pathway with multiple functions. Biochim Biophys Acta, 1763, 1413–26. [DOI] [PubMed] [Google Scholar]
  120. QU P, DU H, WILKES DS & YAN C 2009. Critical roles of lysosomal acid lipase in T cell development and function. Am J Pathol, 174, 944–56. [DOI] [PMC free article] [PubMed] [Google Scholar]
  121. RAK S, DE ZAN T, STEFULJ J, KOSOVIC M, GAMULIN O & OSMAK M 2014. FTIR spectroscopy reveals lipid droplets in drug resistant laryngeal carcinoma cells through detection of increased ester vibrational bands intensity. Analyst, 139, 3407–15. [DOI] [PubMed] [Google Scholar]
  122. RAO S, TORTOLA L, PERLOT T, WIRNSBERGER G, NOVATCHKOVA M, NITSCH R, SYKACEK P, FRANK L, SCHRAMEK D, KOMNENOVIC V, SIGL V, AUMAYR K, SCHMAUSS G, FELLNER N, HANDSCHUH S, GLOSMANN M, PASIERBEK P, SCHLEDERER M, RESCH GP, MA Y, YANG H, POPPER H, KENNER L, KROEMER G & PENNINGER JM 2014. A dual role for autophagy in a murine model of lung cancer. Nat Commun, 5, 3056. [DOI] [PubMed] [Google Scholar]
  123. REBECCA VW, NICASTRI MC, FENNELLY C, CHUDE CI, BARBER-ROTENBERG JS, RONGHE A, MCAFEE Q, MCLAUGHLIN NP, ZHANG G, GOLDMAN AR, OJHA R, PIAO S, NOGUERA-ORTEGA E, MARTORELLA A, ALICEA GM, LEE JJ, SCHUCHTER LM, XU X, HERLYN M, MARMORSTEIN R, GIMOTTY PA, SPEICHER DW, WINKLER JD & AMARAVADI RK 2019. PPT1 Promotes Tumor Growth and Is the Molecular Target of Chloroquine Derivatives in Cancer. Cancer Discov, 9, 220–229. [DOI] [PMC free article] [PubMed] [Google Scholar]
  124. RILEY JS & TAIT SW 2020. Mitochondrial DNA in inflammation and immunity. EMBO Rep, 21, e49799. [DOI] [PMC free article] [PubMed] [Google Scholar]
  125. ROGOV V, DOTSCH V, JOHANSEN T & KIRKIN V 2014. Interactions between autophagy receptors and ubiquitin-like proteins form the molecular basis for selective autophagy. Mol Cell, 53, 167–78. [DOI] [PubMed] [Google Scholar]
  126. ROSENFELDT MT, O’PREY J, MORTON JP, NIXON C, MACKAY G, MROWINSKA A, AU A, RAI TS, ZHENG L, RIDGWAY R, ADAMS PD, ANDERSON KI, GOTTLIEB E, SANSOM OJ & RYAN KM 2013. p53 status determines the role of autophagy in pancreatic tumour development. Nature, 504, 296–300. [DOI] [PubMed] [Google Scholar]
  127. SAHU R, KAUSHIK S, CLEMENT CC, CANNIZZO ES, SCHARF B, FOLLENZI A, POTOLICCHIO I, NIEVES E, CUERVO AM & SANTAMBROGIO L 2011. Microautophagy of cytosolic proteins by late endosomes. Dev Cell, 20, 131–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  128. SAITOH T & AKIRA S 2016. Regulation of inflammasomes by autophagy. J Allergy Clin Immunol, 138, 28–36. [DOI] [PubMed] [Google Scholar]
  129. SANTOS CR & SCHULZE A 2012. Lipid metabolism in cancer. FEBS J, 279, 2610–23. [DOI] [PubMed] [Google Scholar]
  130. SARGENT G, VAN ZUTPHEN T, SHATSEVA T, ZHANG L, DI GIOVANNI V, BANDSMA R & KIM PK 2016. PEX2 is the E3 ubiquitin ligase required for pexophagy during starvation. J Cell Biol, 214, 677–90. [DOI] [PMC free article] [PubMed] [Google Scholar]
  131. SAROSIEK KA, NI CHONGHAILE T & LETAI A 2013. Mitochondria: gatekeepers of response to chemotherapy. Trends Cell Biol, 23, 612–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  132. SCHROEDER LK, BARENTINE AES, MERTA H, SCHWEIGHOFER S, ZHANG Y, BADDELEY D, BEWERSDORF J & BAHMANYAR S 2019. Dynamic nanoscale morphology of the ER surveyed by STED microscopy. J Cell Biol, 218, 83–96. [DOI] [PMC free article] [PubMed] [Google Scholar]
  133. SECZYNSKA M & DIKIC I 2017. Removing the waste bags: how p97 drives autophagy of lysosomes. EMBO J, 36, 129–131. [DOI] [PMC free article] [PubMed] [Google Scholar]
  134. SETTEMBRE C, FRALDI A, MEDINA DL & BALLABIO A 2013. Signals from the lysosome: a control centre for cellular clearance and energy metabolism. Nat Rev Mol Cell Biol, 14, 283–96. [DOI] [PMC free article] [PubMed] [Google Scholar]
  135. SEXTON JZ, HE Q, FORSBERG LJ & BRENMAN JE 2010. High content screening for non-classical peroxisome proliferators. Int J High Throughput Screen, 2010, 127–140. [DOI] [PMC free article] [PubMed] [Google Scholar]
  136. SHIBA-FUKUSHIMA K, IMAI Y, YOSHIDA S, ISHIHAMA Y, KANAO T, SATO S & HATTORI N 2012. PINK1-mediated phosphorylation of the Parkin ubiquitin-like domain primes mitochondrial translocation of Parkin and regulates mitophagy. Sci Rep, 2, 1002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  137. SINGH R, KAUSHIK S, WANG Y, XIANG Y, NOVAK I, KOMATSU M, TANAKA K, CUERVO AM & CZAJA MJ 2009. Autophagy regulates lipid metabolism. Nature, 458, 1131–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  138. SLITER DA, MARTINEZ J, HAO L, CHEN X, SUN N, FISCHER TD, BURMAN JL, LI Y, ZHANG Z, NARENDRA DP, CAI H, BORSCHE M, KLEIN C & YOULE RJ 2018. Parkin and PINK1 mitigate STING-induced inflammation. Nature, 561, 258–262. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  139. SMITH MD, HARLEY ME, KEMP AJ, WILLS J, LEE M, ARENDS M, VON KRIEGSHEIM A, BEHRENDS C & WILKINSON S 2018. CCPG1 Is a Non-canonical Autophagy Cargo Receptor Essential for ER-Phagy and Pancreatic ER Proteostasis. Dev Cell, 44, 217–232 e11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  140. STEPHANI M, PICCHIANTI L, GAJIC A, BEVERIDGE R, SKARWAN E, SANCHEZ DE MEDINA HERNANDEZ V, MOHSENI A, CLAVEL M, ZENG Y, NAUMANN C, MATUSZKIEWICZ M, TURCO E, LOEFKE C, LI B, DURNBERGER G, SCHUTZBIER M, CHEN HT, ABDRAKHMANOV A, SAVOVA A, CHIA KS, DJAMEI A, SCHAFFNER I, ABEL S, JIANG L, MECHTLER K, IKEDA F, MARTENS S, CLAUSEN T & DAGDAS Y 2020. A cross-kingdom conserved ER-phagy receptor maintains endoplasmic reticulum homeostasis during stress. Elife, 9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  141. STROHECKER AM, GUO JY, KARSLI-UZUNBAS G, PRICE SM, CHEN GJ, MATHEW R, MCMAHON M & WHITE E 2013. Autophagy sustains mitochondrial glutamine metabolism and growth of BrafV600E-driven lung tumors. Cancer Discov, 3, 1272–85. [DOI] [PMC free article] [PubMed] [Google Scholar]
  142. SUKHAI MA, PRABHA S, HURREN R, RUTLEDGE AC, LEE AY, SRISKANTHADEVAN S, SUN H, WANG X, SKRTIC M, SENEVIRATNE A, CUSIMANO M, JHAS B, GRONDA M, MACLEAN N, CHO EE, SPAGNUOLO PA, SHARMEEN S, GEBBIA M, URBANUS M, EPPERT K, DISSANAYAKE D, JONET A, DASSONVILLE-KLIMPT A, LI X., DATTI A, OHASHI PS, WRANA J, ROGERS I, SONNET P, ELLIS WY, COREY SJ, EAVES C, MINDEN MD, WANG JC, DICK JE, NISLOW C, GIAEVER G & SCHIMMER AD 2013. Lysosomal disruption preferentially targets acute myeloid leukemia cells and progenitors. J Clin Invest, 123, 315–28. [DOI] [PMC free article] [PubMed] [Google Scholar]
  143. SUZUKI K, KIRISAKO T, KAMADA Y, MIZUSHIMA N, NODA T & OHSUMI Y 2001. The pre-autophagosomal structure organized by concerted functions of APG genes is essential for autophagosome formation. EMBO J, 20, 5971–81. [DOI] [PMC free article] [PubMed] [Google Scholar]
  144. TAKAMURA A, KOMATSU M, HARA T, SAKAMOTO A, KISHI C, WAGURI S, EISHI Y, HINO O, TANAKA K & MIZUSHIMA N 2011. Autophagy-deficient mice develop multiple liver tumors. Genes Dev, 25, 795–800. [DOI] [PMC free article] [PubMed] [Google Scholar]
  145. TAKENOKUCHI M, MIYAMOTO K, SAIGO K & TANIGUCHI T 2015. Bortezomib Causes ER Stress-related Death of Acute Promyelocytic Leukemia Cells Through Excessive Accumulation of PML-RARA. Anticancer Res, 35, 3307–16. [PubMed] [Google Scholar]
  146. TANAKA A, CLELAND MM, XU S, NARENDRA DP, SUEN DF, KARBOWSKI M & YOULE RJ 2010. Proteasome and p97 mediate mitophagy and degradation of mitofusins induced by Parkin. J Cell Biol, 191, 1367–80. [DOI] [PMC free article] [PubMed] [Google Scholar]
  147. TANG WK, CHUI CH, FATIMA S, KOK SH, PAK KC, OU TM, HUI KS, WONG MM, WONG J, LAW S, TSAO SW, LAM KY, BEH PS, SRIVASTAVA G, CHAN AS, HO KP & TANG JC 2007. Oncogenic properties of a novel gene JK-1 located in chromosome 5p and its overexpression in human esophageal squamous cell carcinoma. Int J Mol Med, 19, 915–23. [PubMed] [Google Scholar]
  148. TEJEDA-MUNOZ N, ALBRECHT LV, BUI MH & DE ROBERTIS EM 2019. Wnt canonical pathway activates macropinocytosis and lysosomal degradation of extracellular proteins. Proc Natl Acad Sci U S A, 116, 10402–10411. [DOI] [PMC free article] [PubMed] [Google Scholar]
  149. THURSTON TL, WANDEL MP, VON MUHLINEN N, FOEGLEIN A & RANDOW F 2012. Galectin 8 targets damaged vesicles for autophagy to defend cells against bacterial invasion. Nature, 482, 414–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  150. TILL A, LAKHANI R, BURNETT SF & SUBRAMANI S 2012. Pexophagy: the selective degradation of peroxisomes. Int J Cell Biol, 2012, 512721. [DOI] [PMC free article] [PubMed] [Google Scholar]
  151. TIRINATO L, LIBERALE C, DI FRANCO S, CANDELORO P, BENFANTE A, LA ROCCA R, POTZE L, MAROTTA R, RUFFILLI R, RAJAMANICKAM VP, MALERBA M, DE ANGELIS F, FALQUI A, CARBONE E, TODARO M, MEDEMA JP, STASSI G & DI FABRIZIO E 2015. Lipid droplets: a new player in colorectal cancer stem cells unveiled by spectroscopic imaging. Stem Cells, 33, 35–44. [DOI] [PMC free article] [PubMed] [Google Scholar]
  152. TOMPKINS KD & THORBURN A 2019. Regulation of Apoptosis by Autophagy to Enhance Cancer Therapy. Yale J Biol Med, 92, 707–718. [PMC free article] [PubMed] [Google Scholar]
  153. TOWERS CG, FITZWALTER BE, REGAN D, GOODSPEED A, MORGAN MJ, LIU CW, GUSTAFSON DL & THORBURN A 2019. Cancer Cells Upregulate NRF2 Signaling to Adapt to Autophagy Inhibition. Dev Cell, 50, 690–703 e6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  154. TOWERS CG & THORBURN A 2019. Circumventing autophagy inhibition. Cell Cycle, 18, 3421–3431. [DOI] [PMC free article] [PubMed] [Google Scholar]
  155. VILLA E, MARCHETTI S & RICCI JE 2018. No Parkin Zone: Mitophagy without Parkin. Trends Cell Biol, 28, 882–895. [DOI] [PubMed] [Google Scholar]
  156. VILLA E, PROICS E, RUBIO-PATINO C, OBBA S, ZUNINO B, BOSSOWSKI JP, ROZIER RM, CHICHE J, MONDRAGON L, RILEY JS, MARCHETTI S, VERHOEYEN E, TAIT SWG & RICCI JE 2017. Parkin-Independent Mitophagy Controls Chemotherapeutic Response in Cancer Cells. Cell Rep, 20, 2846–2859. [DOI] [PubMed] [Google Scholar]
  157. VOELTZ GK, PRINZ WA, SHIBATA Y, RIST JM & RAPOPORT TA 2006. A class of membrane proteins shaping the tubular endoplasmic reticulum. Cell, 124, 573–86. [DOI] [PubMed] [Google Scholar]
  158. WANG C & YOULE RJ 2009. The role of mitochondria in apoptosis*. Annu Rev Genet, 43, 95–118. [DOI] [PMC free article] [PubMed] [Google Scholar]
  159. WANG J, FANG Y, YAN L, YUAN N, ZHANG S, XU L, NIE M, ZHANG X & WANG J 2016. Erythroleukemia cells acquire an alternative mitophagy capability. Sci Rep, 6, 24641. [DOI] [PMC free article] [PubMed] [Google Scholar]
  160. WANG M & KAUFMAN RJ 2014. The impact of the endoplasmic reticulum protein-folding environment on cancer development. Nat Rev Cancer, 14, 581–97. [DOI] [PubMed] [Google Scholar]
  161. WARBURG O 1956. On the origin of cancer cells. Science, 123, 309–14. [DOI] [PubMed] [Google Scholar]
  162. WARBURG O, WIND F & NEGELEIN E 1927. The Metabolism of Tumors in the Body. J Gen Physiol, 8, 519–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
  163. WEI H, WANG C, CROCE CM & GUAN JL 2014. p62/SQSTM1 synergizes with autophagy for tumor growth in vivo. Genes Dev, 28, 1204–16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  164. WEI R, CAO J & YAO S 2018. Matrine promotes liver cancer cell apoptosis by inhibiting mitophagy and PINK1/Parkin pathways. Cell Stress Chaperones, 23, 1295–1309. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  165. WEINBERG F, HAMANAKA R, WHEATON WW, WEINBERG S, JOSEPH J, LOPEZ M, KALYANARAMAN B, MUTLU GM, BUDINGER GR & CHANDEL NS 2010. Mitochondrial metabolism and ROS generation are essential for Kras-mediated tumorigenicity. Proc Natl Acad Sci U S A, 107, 8788–93. [DOI] [PMC free article] [PubMed] [Google Scholar]
  166. WEMMERT S, LINDNER Y, LINXWEILER J, WAGENPFEIL S, BOHLE R, NIEWALD M & SCHICK B 2016. Initial evidence for Sec62 as a prognostic marker in advanced head and neck squamous cell carcinoma. Oncol Lett, 11, 1661–1670. [DOI] [PMC free article] [PubMed] [Google Scholar]
  167. WENG L, DU J, ZHOU Q, CHENG B, LI J, ZHANG D & LING C 2012. Identification of cyclin B1 and Sec62 as biomarkers for recurrence in patients with HBV-related hepatocellular carcinoma after surgical resection. Mol Cancer, 11, 39. [DOI] [PMC free article] [PubMed] [Google Scholar]
  168. WENT PT, SAUTER G, OBERHOLZER M & BUBENDORF L 2006. Abundant expression of AMACR in many distinct tumour types. Pathology, 38, 426–32. [DOI] [PubMed] [Google Scholar]
  169. WERNEBURG NW, GUICCIARDI ME, BRONK SF, KAUFMANN SH & GORES GJ 2007. Tumor necrosis factor-related apoptosis-inducing ligand activates a lysosomal pathway of apoptosis that is regulated by Bcl-2 proteins. J Biol Chem, 282, 28960–70. [DOI] [PubMed] [Google Scholar]
  170. WHITE E 2015. The role for autophagy in cancer. J Clin Invest, 125, 42–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  171. WHITE E, KARP C, STROHECKER AM, GUO Y & MATHEW R 2010. Role of autophagy in suppression of inflammation and cancer. Curr Opin Cell Biol, 22, 212–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  172. WIBLE DJ, CHAO HP, TANG DG & BRATTON SB 2019. ATG5 cancer mutations and alternative mRNA splicing reveal a conjugation switch that regulates ATG12-ATG5-ATG16L1 complex assembly and autophagy. Cell Discov, 5, 42. [DOI] [PMC free article] [PubMed] [Google Scholar]
  173. WITKIEWICZ AK, VARAMBALLY S, SHEN R, MEHRA R, SABEL MS, GHOSH D, CHINNAIYAN AM, RUBIN MA & KLEER CG 2005. Alpha-methylacyl-CoA racemase protein expression is associated with the degree of differentiation in breast cancer using quantitative image analysis. Cancer Epidemiol Biomarkers Prev, 14, 1418–23. [DOI] [PubMed] [Google Scholar]
  174. WU J, CHEN S, LIU H, ZHANG Z, NI Z, CHEN J, YANG Z, NIE Y & FAN D 2018. Tunicamycin specifically aggravates ER stress and overcomes chemoresistance in multidrug-resistant gastric cancer cells by inhibiting N-glycosylation. J Exp Clin Cancer Res, 37, 272. [DOI] [PMC free article] [PubMed] [Google Scholar]
  175. YAMAZAKI T, KIRCHMAIR A, SATO A, BUQUE A, RYBSTEIN M, PETRONI G, BLOY N, FINOTELLO F, STAFFORD L, NAVARRO MANZANO E, AYALA DE LA PENA F, GARCIA-MARTINEZ E, FORMENTI SC, TRAJANOSKI Z & GALLUZZI L 2020. Mitochondrial DNA drives abscopal responses to radiation that are inhibited by autophagy. Nat Immunol, 21, 1160–1171. [DOI] [PubMed] [Google Scholar]
  176. YAN C, LUO L, GUO CY, GOTO S, URATA Y, SHAO JH & LI TS 2017. Doxorubicin-induced mitophagy contributes to drug resistance in cancer stem cells from HCT8 human colorectal cancer cells. Cancer Lett, 388, 34–42. [DOI] [PubMed] [Google Scholar]
  177. YANG A, HERTER-SPRIE G, ZHANG H, LIN EY, BIANCUR D, WANG X, DENG J, HAI J, YANG S, WONG KK & KIMMELMAN AC 2018. Autophagy Sustains Pancreatic Cancer Growth through Both Cell-Autonomous and Nonautonomous Mechanisms. Cancer Discov, 8, 276–287. [DOI] [PMC free article] [PubMed] [Google Scholar]
  178. YANG A, RAJESHKUMAR NV, WANG X, YABUUCHI S, ALEXANDER BM, CHU GC, VON HOFF DD, MAITRA A & KIMMELMAN AC 2014. Autophagy is critical for pancreatic tumor growth and progression in tumors with p53 alterations. Cancer Discov, 4, 905–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  179. YANG S, WANG X, CONTINO G, LIESA M, SAHIN E, YING H, BAUSE A, LI Y, STOMMEL JM, DELL’ANTONIO G, MAUTNER J, TONON G, HAIGIS M, SHIRIHAI OS, DOGLIONI C, BARDEESY N & KIMMELMAN AC 2011. Pancreatic cancers require autophagy for tumor growth. Genes Dev, 25, 717–29. [DOI] [PMC free article] [PubMed] [Google Scholar]
  180. YAO N, WANG C, HU N, LI Y, LIU M, LEI Y, CHEN M, CHEN L, CHEN C, LAN P, CHEN W, CHEN Z, FU D, YE W & ZHANG D 2019. Inhibition of PINK1/Parkin-dependent mitophagy sensitizes multidrug-resistant cancer cells to B5G1, a new betulinic acid analog. Cell Death Dis, 10, 232. [DOI] [PMC free article] [PubMed] [Google Scholar]
  181. YELDANDI AV, RAO MS & REDDY JK 2000. Hydrogen peroxide generation in peroxisome proliferator-induced oncogenesis. Mutat Res, 448, 159–77. [DOI] [PubMed] [Google Scholar]
  182. YOKOTA S & DARIUSH FAHIMI H 2009. Degradation of excess peroxisomes in mammalian liver cells by autophagy and other mechanisms. Histochem Cell Biol, 131, 455–8. [DOI] [PubMed] [Google Scholar]
  183. YU SM & KIM SJ 2010. Endoplasmic reticulum stress (ER-stress) by 2-deoxy-D-glucose (2DG) reduces cyclooxygenase-2 (COX-2) expression and N-glycosylation and induces a loss of COX-2 activity via a Src kinase-dependent pathway in rabbit articular chondrocytes. Exp Mol Med, 42, 777–86. [DOI] [PMC free article] [PubMed] [Google Scholar]
  184. ZECHNER R, MADEO F & KRATKY D 2017. Cytosolic lipolysis and lipophagy: two sides of the same coin. Nat Rev Mol Cell Biol, 18, 671–684. [DOI] [PubMed] [Google Scholar]
  185. ZHANG C, LIN M, WU R, WANG X, YANG B, LEVINE AJ, HU W & FENG Z 2011. Parkin, a p53 target gene, mediates the role of p53 in glucose metabolism and the Warburg effect. Proc Natl Acad Sci U S A, 108, 16259–64. [DOI] [PMC free article] [PubMed] [Google Scholar]
  186. ZHANG J, TRIPATHI DN, JING J, ALEXANDER A, KIM J, POWELL RT, DERE R, TAIT-MULDER J, LEE JH, PAULL TT, PANDITA RK, CHARAKA VK, PANDITA TK, KASTAN MB & WALKER CL 2015. ATM functions at the peroxisome to induce pexophagy in response to ROS. Nat Cell Biol, 17, 1259–1269. [DOI] [PMC free article] [PubMed] [Google Scholar]
  187. ZHANG N, SHENG M, WU M, ZHANG X, DING Y, LIN Y, YU W, WANG S & DU H 2019a. Berberine protects steatotic donor undergoing liver transplantation via inhibiting endoplasmic reticulum stress-mediated reticulophagy. Exp Biol Med (Maywood), 244, 1695–1704. [DOI] [PMC free article] [PubMed] [Google Scholar]
  188. ZHANG S, LIU X, BAWA-KHALFE T, LU LS, LYU YL, LIU LF & YEH ET 2012. Identification of the molecular basis of doxorubicin-induced cardiotoxicity. Nat Med, 18, 1639–42. [DOI] [PubMed] [Google Scholar]
  189. ZHANG Z, GAO W, ZHOU L, CHEN Y, QIN S, ZHANG L, LIU J, HE Y, LEI Y, CHEN HN, HAN J, ZHOU ZG, NICE EC, LI C, HUANG C & WEI X 2019b. Repurposing Brigatinib for the Treatment of Colorectal Cancer Based on Inhibition of ER-phagy. Theranostics, 9, 4878–4892. [DOI] [PMC free article] [PubMed] [Google Scholar]
  190. ZHANG ZL, WANG NN, MA QL, CHEN Y, YAO L, ZHANG L, LI QS, SHI MH, WANG HF & YING Z 2020. Somatic and germline mutations in the tumor suppressor gene PARK2 impair PINK1/Parkin-mediated mitophagy in lung cancer cells. Acta Pharmacol Sin, 41, 93–100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  191. ZHAO T, DING X, DU H & YAN C 2016. Lung Epithelial Cell-Specific Expression of Human Lysosomal Acid Lipase Ameliorates Lung Inflammation and Tumor Metastasis in Lipa(−/−) Mice. Am J Pathol, 186, 2183–2192. [DOI] [PMC free article] [PubMed] [Google Scholar]
  192. ZHAO T, DU H, DING X, WALLS K & YAN C 2015. Activation of mTOR pathway in myeloid-derived suppressor cells stimulates cancer cell proliferation and metastasis in lal(−/−) mice. Oncogene, 34, 1938–48. [DOI] [PMC free article] [PubMed] [Google Scholar]
  193. ZHITOMIRSKY B & ASSARAF YG 2016. Lysosomes as mediators of drug resistance in cancer. Drug Resist Updat, 24, 23–33. [DOI] [PubMed] [Google Scholar]

RESOURCES