Skip to main content
Cold Spring Harbor Perspectives in Biology logoLink to Cold Spring Harbor Perspectives in Biology
. 2012 Sep;4(9):a008763. doi: 10.1101/cshperspect.a008763

Metabolic Stress in Autophagy and Cell Death Pathways

Brian J Altman 1, Jeffrey C Rathmell 1
PMCID: PMC3428762  NIHMSID: NIHMS438926  PMID: 22952396

Abstract

Growth factors and oncogenic kinases play important roles in stimulating cell growth during development and transformation. These processes have significant energetic and synthetic requirements and it is apparent that a central function of growth signals is to promote glucose metabolism to support these demands. Because metabolic pathways represent a fundamental aspect of cell proliferation and survival, there is considerable interest in targeting metabolism as a means to eliminate cancer. A challenge, however, is that molecular links between metabolic stress and cell death are poorly understood. Here we review current literature on how cells cope with metabolic stress and how autophagy, apoptosis, and necrosis are tightly linked to cell metabolism. Ultimately, understanding of the interplay between nutrients, autophagy, and cell death will be a key component in development of new treatment strategies to exploit the altered metabolism of cancer cells.


Autophagy is induced by nutrient stresses and other cellular insults; it delays apoptosis. When autophagy is unable to mitigate cell stress, insufficient metabolism can induce apoptosis.


Although single-celled organisms grow and proliferate based on nutrient availability, metazoan cells rely on growth factor input to promote nutrient uptake, regulate growth and proliferation, and survive (Raff 1992; Rathmell et al. 2000). Access and competition for these signals are critical in developmental patterning and to maintain homeostasis of mature tissues. Cells that do not receive proper growth factor signals typically atrophy, lose the ability to uptake and use extracellular nutrients, and instead induce the self-digestive process of autophagy as an intracellular energy source before ultimately undergoing programmed cell death. Cancer cells, in contrast, often become independent of extracellular growth signals by gaining mutations or expressing oncogenic kinases to drive intrinsic growth signals that mimic growth factor input, which can be the source of oncogene addiction. Growth factor input or oncogenic signals often drive highly elevated glucose uptake and metabolism (Rathmell et al. 2000; DeBerardinis et al. 2008; Michalek and Rathmell 2010). First described in cancer by Warburg in the 1920s, this highly glycolytic metabolic program is termed aerobic glycolysis and is a general feature of many nontransformed proliferative cells (Warburg 1956; DeBerardinis et al. 2008).

Nutrient uptake and aerobic glycolysis induced by growth signals play key roles in cell survival (Vander Heiden et al. 2001). Manipulating cell metabolism as a means to promote the death of inappropriately dividing cells, therefore, is a promising new avenue to treat disease. Targeting the altered metabolism of cancer cells in particular is of great interest. It is still unclear at the molecular level, however, how inhibiting or modulating cell metabolism leads to apoptosis, and how these pathways may best be exploited (Dang et al. 2009; Wise and Thompson 2010).

Growth factor or oncogenic kinases promote multiple metabolic pathways that are essential to prevent metabolic stress and may be targets in efforts to link metabolism and cell death (Vander Heiden et al. 2001). Decreased glucose metabolism on loss of growth signals leads to decreased ATP generation as well as loss in generation of many biosynthetic precursor molecules, including nucleic acids, fatty acids, and acetyl-CoA for acetylation (Zhao et al. 2007; Wellen et al. 2009; Coloff et al. 2011). Glucose is also important as a precursor for the hexosamine pathway, to allow proper glycosylation and protein folding in the endoplasmic reticulum (Dennis et al. 2009; Kaufman et al. 2010). If glucose metabolism remains insufficient or disrupted, the cells can switch to rely on mitochondrial oxidation of fatty acids and amino acids, which are energy rich but do not readily support cell growth and can lead to potentially dangerous levels of reactive oxygen species (Wellen and Thompson 2010). Amino acid deficiency can directly inhibit components of the signaling pathways downstream from growth factors and activate autophagy (Lynch 2001; Beugnet et al. 2003; Byfield et al. 2005; Nobukuni et al. 2005). Finally, hypoxia induces a specific pathway to increase nutrient uptake and metabolism via the hypoxia-inducible factor (HIF1/2α) that promotes adaptation to anaerobic conditions, but may lead to apoptosis if hypoxia is severe (Saikumar et al. 1998; Suzuki et al. 2001; Fulda and Debatin 2007).

Typically a combination of metabolic stresses rather than loss of a single nutrient input occur at a given time (Degenhardt et al. 2006) and autophagy is activated to mitigate damage and provide nutrients for short-term survival (Bernales et al. 2006; Tracy et al. 2007; Altman et al. 2011; Guo et al. 2011). Autophagy is a cellular process of bulk cytoplasmic and organelle degradation common to nearly all eukaryotes. Unique double-membraned vesicles known as autophagosomes engulf cellular material and fuse with lysosomes to promote degradation of the contents (Kelekar 2005). Described in greater detail below, autophagy can reduce sources of stress, such as protein aggregates and damaged or dysfunctional intracellular organelles, and provide nutrients during times of transient and acute nutrient withdrawal.

Despite the protective effects of autophagy, cells deprived of growth signals, nutrients, or oxygen for prolonged times will eventually succumb to cell death. Apoptosis is the initial death response on metabolic stress and is regulated by Bcl-2 family proteins. In healthy cells, antiapoptotic Bcl-2 family proteins, such as Bcl-2, Bcl-xl, and Mcl-1, bind and inhibit the multidomain proapoptotic proteins Bax and Bak (van Delft and Huang 2006; Walensky 2006; Chipuk et al. 2010). In metabolic stress, proapoptotic “BH3-only” proteins of the Bcl-2 family are induced or activated and bind to and inhibit the antiapoptotic Bcl-2 family proteins to allow activation of the proapoptotic Bax and Bak (Galonek and Hardwick 2006). The BH3-only proteins Bim, Bid, and Puma can also directly bind and activate Bax and Bak (Letai et al. 2002; Ren et al. 2010). Active Bax and Bak disrupt the outer mitochondrial membrane (termed mitochondrial outer-membrane permeabilization, or MOMP) and release several proapoptotic factors including cytochrome-C that activate the apoptosome that in turn activates effector caspases to cleave a variety of cellular proteins and drive apoptosis (Schafer and Kornbluth 2006). In cases in which these apoptotic pathways are suppressed, metabolic stress can instead lead to necrotic cell death (Jin et al. 2007).

ROLE OF GROWTH FACTORS IN METABOLISM AND CELL DEATH

The phosphatidylinositol-3-kinase (PI3K)/AKT/mTOR pathway, outlined in Figure 1, is central in growth factor-stimulated control of glucose uptake and metabolism and is often altered in cancer. PI3K catalyzes the phosphorylation of phosphatidylinositol (PI) 4,5 bisphosphate (PIP2) to PI 3,4,5 triphosphate (PIP3). After growth factor signals are received, PIP3 accumulates, leading to recruitment and phosphorylation of Akt (Frech et al. 1997; Zoncu et al. 2011). Activated Akt supports metabolism in a number of ways, notably by maintaining protein translation, glucose metabolism, and inhibiting autophagy and apoptosis. Akt is essential for growth factor-stimulated glucose uptake and glycolysis by localizing Glut1 to the cell surface (Plas et al. 2001; Vander Heiden et al. 2001; Wieman et al. 2007; Wofford et al. 2008) and also increasing the activity and proper localization of hexokinases (HKs) (Gottlob et al. 2001; Robey and Hay 2006) to support the phosphorylation and downstream catabolism of glucose. Akt can also promote glucose metabolism through the pentose phosphate pathway (PPP), which generates NADPH and ribonucleotides to control cellular redox as well as lipid and nucleic acid synthesis (Rathmell et al. 2003; Duvel et al. 2010). In addition, Akt suppresses the catabolism of intracellular components such as fatty acids that would otherwise be used to support cell growth (Deberardinis et al. 2006). Thus, Akt activation promotes glucose metabolism and inhibits other metabolic pathways, leading to a glycolytic phenotype in growth factor-stimulated cells and the aerobic glycolysis characteristic of cancer cells (Elstrom et al. 2004). Importantly, Akt is highly antiapoptotic, but requires glucose to protect cells from death (Vander Heiden et al. 2001; Rathmell et al. 2003; Coloff et al. 2011), highlighting the connections between metabolism and apoptosis and enforcing a glucose addiction on stimulated or cancerous cells.

Figure 1.

Figure 1.

The PI3K-Akt-mTORC1 pathway and control of autophagy. PI3K is induced downstream from growth factor input and activates Akt and mTORC1. Akt supports the localization of nutrient transporters to the cell surface and maintains nutrient uptake. mTORC1 both supports protein translation and inhibits autophagy through phosphorylation of ULK1 and ULK2. AMPK, activated under energetic stress, inhibits mTORC1 and directly associates with ULK1/2 to activate autophagy.

mTORC1 (mechanistic target of rapamycin complex 1) is activated downstream from Akt and is directly regulated by amino acid availability (Lynch 2001; Beugnet et al. 2003; Roccio et al. 2006; Kim et al. 2008; Sancak et al. 2010) to control translation and autophagy (Zoncu et al. 2011). mTORC1 regulates cell growth by promoting protein translation through phosphorylation of two key enzymes. Protein synthesis inhibitor eIF4E binding protein (4EBP) is inactivated by mTORC1 to allow for cap-dependent translation, and S6 kinase (S6K) is phosphorylated and activated by mTORC1 to activate the S6-ribisomal protein that appears to be important for cell metabolism and growth (Hara et al. 1998; Fingar et al. 2002; Tandon et al. 2011). mTORC1 also negatively regulates autophagy through inhibitory phosphorylation of the autophagy-essential kinases Unc-51-like kinase-1 (ULK1) and ULK2 (Ganley et al. 2009; Jung et al. 2009). In addition, mTOR signaling can be suppressed and autophagy induced by the AMP-activated protein kinase (AMPK) (Hoyer-Hansen and Jaattela 2007a; Canto and Auwerx 2010).

Many oncogenes can mimic growth signals to support metabolism in the absence of exogenous growth factor signaling (Vander Heiden et al. 2009). The PI3K/Akt/mTOR pathway plays a major role downstream from these oncogenic mutations to drive aerobic glycolysis. The BCR-Abl fusion protein, for example, promotes trafficking of Glut1 to the cell surface. Inhibition of PI3K, however, blocks this function and Glut1 is internalized thus limiting glucose uptake and glycolysis (Barnes et al. 2005). Myc can directly promote transcription of essentially all key glycolytic genes (Osthus et al. 2000; Dang et al. 2009). Indeed, recent studies have shown that c-Myc and Akt have complementary effects in promoting aerobic glycolysis (Fan et al. 2010). In addition to glucose, proliferating cells also require glutamine (Wise et al. 2008; Dang et al. 2009) and c-Myc has been shown to drive glutamine uptake and metabolism to render cells dependent on this important nutrient to support energy generation and biosynthesis (Wise et al. 2008; Gao et al. 2009).

AUTOPHAGY AND CONTROL OF METABOLIC STRESS

Decreased metabolism leads to induction of autophagy to generate nutrients from intracellular components until proper extracellular nutrient uptake is restored. Autophagy can play a prodeath role when prolonged or in certain developmental conditions, such as elimination of blastocyst inner cell mass (see review by Das et al. 2012), but in most circumstances autophagic generation of nutrients prevents or delays cell death. In autophagy, the cell packages organelles, bulk cytoplasm, and long-lived proteins in double-membraned vesicles for delivery to the lysosome and eventual degradation (Kelekar 2005). Autophagy occurs in all metazoan cells at low levels under basal conditions as a quality-control and waste-disposal mechanism (Ganley et al. 2009; Jung et al. 2009), but is induced as a protective mechanism when cells are under stress (Fig. 2).

Figure 2.

Figure 2.

Autophagy responds to a variety of stresses to prevent cell death. Autophagy can be induced by a variety of nutrient stresses and other cellular insults. Induced autophagy can provide nutrients for mitochondrial oxidation, suppress p53 pathway activation, and ultimately delay apoptosis.

Induction of autophagy can relieve a variety of cell stresses (Fig. 2). Autophagy can prevent overaccumulation of mitochondria and remove damaged mitochondria to prevent cell death (Colell et al. 2007; Pua et al. 2009) and to prevent an increase in damaging reactive oxygen species (ROS) (Mathew et al. 2009; Rouschop et al. 2009; Tal et al. 2009). Although some amounts of ROS are required for normal cell signaling (Hamanaka and Chandel 2010), excess ROS can lead to cellular damage, p53 activation, induction of proapoptotic proteins, and cell death in a wide variety of tissues (von Harsdorf et al. 1999; Sade and Sarin 2004; Karawajew et al. 2005; Liu et al. 2008a; Niizuma et al. 2009; Bodet et al. 2010). Autophagy is also important to engulf damaged endoplasmic reticulum (ER) in the unfolded protein response (Bernales et al. 2006; Hoyer-Hansen and Jaattela 2007b) and degrade protein aggregates that may otherwise lead to neurodegeneration (Bjorkoy et al. 2005; Komatsu et al. 2006). Finally, autophagy is essential to limit DNA damage and genomic instability, possibly through modulation of protein aggregates and the adaptor protein p62 (Mathew et al. 2007, 2009). As a consequence, autophagy-deficient cancer cells have increased genomic damage that can paradoxically promote tumor progression (Liang et al. 1999; Yue et al. 2003). This DNA damage may lead to a p53-dependent stress response, and p53 deficiency can promote continued proliferation despite insufficient autophagy and DNA damage (Altman et al. 2011).

As discussed above, growth factor-mediated activation of the PI3K-Akt-mTORC1 pathway suppresses autophagy in healthy cells in nutrient-replete conditions. mTORC1 inhibits autophagy by phosphorylating and inhibiting ULK1 and ULK2, key upstream regulators of autophagy (Fig. 1) (Ganley et al. 2009; Jung et al. 2009). Growth factor or nutrient withdrawal (particularly withdrawal of amino acids) inactivates mTORC1 and activates autophagy (Beugnet et al. 2003; Altman et al. 2009). The program of autophagy is performed by a complex pathway of Atg-family proteins and two ubiquitinlike conjugation systems, reviewed in detail in Das et al. (2012). In addition to amino acid-sensitive regulation of autophagy by mTORC1, decreases in ATP resulting from nutrient deprivation can activate the AMPK pathway to promote autophagy. The AMPK heterotrimer is maximally activated when both AMP (adenyl monophosphate) increases and the AMPKα subunit is phosphorylated by the tumor suppressor LKB1 (Hawley et al. 2003; Woods et al. 2003; Shaw et al. 2004; Shaw 2009). AMPK then initiates a cellular program to conserve and generate additional ATP by initiating a G1 cell-cycle arrest, increasing glucose uptake, glycolysis, fatty acid oxidation, and halting protein synthesis and glycogen synthesis (Imamura et al. 2001; Jones et al. 2005; Shaw 2009). AMPK can phosphorylate and activate TSC2 to inhibit mTORC1 and thus indirectly activate autophagy to degrade damaged organelles and mobilize intracellular nutrients (Hoyer-Hansen and Jaattela 2007a; Canto and Auwerx 2010). In addition, several recent studies in Caenorhabditis elegans and mammalian cells have shown that AMPK directly associates and phosphorylates ULK1 in response to glucose deprivation or multinutrient deprivation, and that this interaction is essential for induction of autophagy (Fig. 1) (Egan et al. 2011; Kim et al. 2011; Shang et al. 2011).

Autophagic degradation of mitochondria and other organelles, protein aggregates, cytoplasm, and long-lived proteins can yield significant energy to cells (Kelekar 2005). Autophagosomes fuse with lysosomes to digest the contents and release amino acids and free fatty acids back to the cytosol for mitochondrial oxidation (Fig. 2) (Altman et al. 2011; Guo et al. 2011). This metabolic strategy can be highly efficient to maintain bioenergetics for a potential long period, as complete catabolism of a single molecule of palmitate, a long-chain fatty acid that is often a component of the phospholipid bilayer of membranes, can yield up to 104 ATP, compared to 31 from a single molecule of glucose (Salway 2004). Likewise, metabolism of amino acids can also yield large amounts of ATP.

Although a role for autophagy as a nutrient source has been apparent in a variety of genetic experiments using yeast (Abeliovich and Klionsky 2001), direct biochemical evidence for autophagy in mammalian cell metabolism has emerged only recently. Lum et al. (2005) showed that autophagy eventually becomes critical as a nutrient source for survival after growth factor deprivation of Bax/Bak-deficient cells. Most interestingly, this cell death caused by autophagy inhibition could be rescued by addition of exogenous nutrients, strongly indicating that autophagy was functioning as a nutrient-generating process. Similarly, Boya et al. (2005) showed that autophagy was critical for survival under conditions of nutrient limitation. More recently, we examined the potential of autophagy to support metabolism by mass spectrometry measurement of metabolite levels in autophagy-deficient cells. These data highlighted the ability of autophagy to supply growth factor or nutrient-deprived cells with an intracellular source of long-chain fatty acids (Altman et al. 2009, 2011). Likewise, Singh et al. (2009) showed that autophagy is critical in regulating lipid metabolism. This ability of autophagy to produce nutrients can be critical to allow cell survival in conditions of low exogenous nutrients (Degenhardt et al. 2006; Karantza-Wadsworth et al. 2007; Mathew et al. 2007; Altman et al. 2011).

The ability of autophagy to both control cell stress and produce nutrients has led to the recent observation that some cancers may be “addicted” to autophagy. We found that autophagy may suppress cell stress and a p53-dependent pathway of cell death, and that cells transformed by the BCR-Abl oncogene were dependent on autophagy for survival and leukemogenesis (Altman et al. 2011). Similarly, Guo et al. (2011) recently showed that autophagy is necessary for survival and can support mitochondrial metabolism of Ras-transformed cells under conditions of nutrient starvation. In Ras-transformed breast epithelial cells, Lock et al. showed that autophagy increases glucose flux through glycolysis to support survival of matrix-detached cells (Lock et al. 2011). These data all suggest that autophagy can prevent death in the absence of nutrients in part by supporting metabolism from breakdown of intracellular components and may play a broad role in cellular homeostasis and control of stress.

Despite these effects to relieve stress, autophagy induction can in some cases lead to apoptosis rather than protection from cell death. In Drosophila, Scott et al. (2007) showed that enforced expression of Atg1, the homolog of ULK1/2, led to apoptosis. Autophagy contributes to cell death of several different tissue types during Drosophila development, including the salivary gland, midgut, and reproductive cells (Berry and Baehrecke 2007; Hou et al. 2008; Denton et al. 2009; Nezis et al. 2010). We observed in several mammalian hematopoietic lines that autophagy induction in the absence of growth factor initially provided nutrients but eventually led to apoptosis dependent on direct induction of the proapoptotic protein Bim (Altman et al. 2009). Similarly, Kiyono et al. (2009) recently observed that autophagy induced in hepatocellular carcinoma cells by TGFβ led to induction of Bim and the proapoptotic protein BMF and eventual apoptosis. Finally, other groups showed that autophagy was necessary for apoptosis of rat neurons treated with a glutamate receptor agonist (Wang et al. 2008a) and for apoptosis downstream from death receptor signaling mouse embryonic fibroblasts (Wang et al. 2008b). However, the molecular mechanisms governing the decision of autophagy to act as a cytoprotective process or to induce apoptosis are poorly understood.

BEYOND AUTOPHAGY: METABOLIC STRESS AND APOPTOSIS

When autophagy is unable to provide sufficient additional nutrients or mitigate cell stress, insufficient metabolism can induce apoptosis. Links between metabolism and apoptosis are now widely appreciated in many species, and glucose metabolism has been shown to play a direct role in regulation apoptosis. In Xenopus oocytes, nutrient depletion over time leads to reduced NADPH and activation of capase 2 to induce apoptosis (Nutt et al. 2005, 2009). Likewise, loss of growth signals and diminished Akt/mTOR signaling in mammalian cells leads to internalization of Glut1 that ultimately leads to apoptosis (Vander Heiden et al. 2001; Wieman et al. 2007; Wofford et al. 2008). If Glut1 is overexpressed, glucose uptake can be maintained even after growth factor deprivation, allowing growth factor-independent glucose metabolism that is sufficient to delay apoptosis (Rathmell et al. 2003; Zhao et al. 2007).

Disruption of metabolism leads to a proapoptotic balance of Bcl-2 family proteins that is essential for apoptosis (Fig. 3). If expression of the proapoptotic Bcl-2 family proteins Puma, Bim, or Noxa is decreased, cells can persist for extended periods even in the absence of glucose (Alves et al. 2006; Coloff et al. 2011). Induction or activation of some proapoptotic BH3-only members of the Bcl-2 family in particular are sensitive to metabolic status. Puma is induced by inhibition of glucose metabolism or disruption in glucose availability in a pathway partially dependent on p53-mediated transcription (Zhao et al. 2008; Coloff et al. 2011). Glucose metabolism also regulates Puma protein stability, as loss of glucose stabilized the Puma protein to enhance apoptosis, whereas addition of exogenous nutrients to support mitochondrial metabolism caused Puma degradation and enhanced cell survival (Coloff et al. 2011). Bim expression is also increased when glucose metabolism is blocked, although this may be owing to the onset of ER stress (Puthalakath et al. 2007) and deficiency of the hexosamine pathway rather than an energetic stress.

Figure 3.

Figure 3.

Glucose withdrawal in apoptosis induction. Glucose withdrawal can lead to a shift in the balance of antiapoptotic and proapoptotic proteins to lead to cell death. Activation of GSK3β downstream can lead to loss of Mcl-1. Activation of AMPK can lead to loss of Mcl-1 and activation of p53. Activation of p53 can lead to induction of Puma. ER stress downstream from glucose withdrawal and loss of glycosylation can lead to induction of both Bim and Puma. Glucose withdrawal can also independently lead to Puma accumulation through stabilization of the protein. Finally, glucose withdrawal can lead to activation of Noxa and Caspase 8.

In contrast to Puma and Bim, the proapoptotic Bcl-2 family protein Noxa is important to promote apoptosis in metabolic stress, but Noxa activity rather than expression appears regulated by cell metabolism. Eldering and colleagues showed in two separate studies that activated T cells up-regulated Noxa, which bound to the antiapoptotic protein Mcl-1. When glucose became limiting, Mcl-1 levels decreased and Noxa promoted apoptosis (Alves et al. 2006; Wensveen et al. 2011). Lowman et al. shed some light on a possible mechanism for this effect by demonstrating that glucose deprivation led to loss of an inhibitory phosphorylation of Noxa, promoting Noxa activation (Lowman et al. 2010). Ultimately the combination of proapoptotic proteins including Puma, Noxa, and also Bim all contribute to cell death.

AMP and LKB1 can activate AMPK on metabolic stress to regulate Bcl-2 family proteins and apoptosis. AMPK and LKB1 can inhibit apoptosis by promoting up-regulation of Bcl-xL and suppressing proapoptotic Erk signaling (Cao et al. 2010; Kim et al. 2010). Indeed, LKB1-deficient thymocytes have low Bcl-xL levels and are sensitive to apoptosis (Cao et al. 2010). Pradelli et al. showed that prolonged AMPK activation caused by inhibition of glycolysis suppresses mTOR signaling and leads to decreased translation of Mcl-1, sensitizing cells to apoptosis (Pradelli et al. 2010). Additionally, AMPK can phosphorylate and activate p53 after nutrient stress, with subsequent cell-cycle arrest and apoptosis (Jones et al. 2005; Okoshi et al. 2008).

Glucose deprivation also results in decreased flux through the hexoamine pathway that can lead to diminished protein glycosylation, protein misfolding, and ER stress. Protein glycosylation depends in part on N-acetylglucosamine (GlcNAc) and is important for proper protein folding in the ER (Kaufman et al. 2010). Misfolded proteins invoke the unfolded protein response (UPR) to reduce global protein synthesis and increase production of chaperone proteins to increase ER protein folding (Hoyer-Hansen and Jaattela 2007b). Initially, lack of glycosylation can greatly reduce glycosylation and surface expression of both nutrient transporters and growth factor receptors, which diminishes cell signaling to further exacerbate metabolic stress (Wellen et al. 2010). Ultimately, however, unresolved misfolded proteins lead to apoptotic death through transcription activation of proapoptotic proteins such as Bim and Puma and activation of apoptosis by the UPR-responsive transcription factor Chop/GADD153 (Oyadomari and Mori 2004; Ishihara et al. 2007; Puthalakath et al. 2007).

In some cases loss of glucose metabolism may lead to apoptosis even in cells lacking essential proapoptotic Bcl-2 family proteins. Two studies from Muñoz-Pinedo and colleagues showed that glucose deprivation could lead to apoptosis of Bak and Bax-deficient cells (Munoz-Pinedo et al. 2003; Caro-Maldonado et al. 2010). In this case, Caspase 8 was activated to induce apoptosis. Surprisingly, however, Caspase 8 activation did not appear to be caused by death receptor signaling and was not Fas-dependent. Rather, Caspase 8 appeared to respond to metabolic stress and promote apoptosis through an unknown mechanism. Although poorly understood, this pathway may be critical as a link to promote the cell death of cancer cells that have acquired mutations to resist intrinsic pathways of apoptosis.

HYPOXIA AND CELL DEATH

Oxygen is essential to support mitochondrial metabolism, but ischemia, tissue damage, or poorly vascularized tumor microenvironments can limit oxygen distribution, leading to metabolic stress and potentially apoptosis (Melillo 2007; Rey and Semenza 2010). Under hypoxia, the normally short-lived transcription factors hypoxia-inducible factor 1/2α (HIF1α and HIF2α) are stabilized to promote an adaptive response that increases anaerobic metabolic pathways. Regulation of HIF1/2α by oxygen occurs through prolyl hydroxylation in an oxygen-dependent reaction that leads to degradation by the ubiquitin-ligase Von Hippel-Lindau (VHL) in normoxia. In hypoxia, prolyl hydroxylation is reduced and HIF1/2α are stabilized (Rey and Semenza 2010). The HIF transcription factors promote a complex transcriptional program to increase anaerobic metabolic flux and survival by recruiting new vasculature and expressing Glut1 and glycolytic enzymes (Fulda and Debatin 2007). HIF also induces the small BH3-only protein BNIP3, which is not thought to induce apoptosis but rather promotes autophagic targeting of mitochondria, or mitophagy (Tracy et al. 2007; Burton and Gibson 2009). Interestingly, BNIP3 is frequently lost in cancer cells, suggesting that mitophagy may be an impediment to cancer cell response to hypoxia (Lee and Paik 2006).

Hypoxia and HIF1α activation can also inhibit apoptosis on a transcriptional level. HIF1α can up-regulate the antiapoptotic proteins Bcl-2, Bcl-xL, and Survivin and down-regulate the proapoptotic Bid and Bax (Erler et al. 2004; Liu et al. 2008b; Chen et al. 2009a,b). HIF1α activity can also antagonize p53-mediated apoptosis, especially with combined DNA damage (Graeber et al. 1996; Hao et al. 2008; Sendoel et al. 2010). In some settings, however, HIF1α has been found to stabilize p53, leading to subsequent apoptosis (Suzuki et al. 2001; Hansson et al. 2002; Chen et al. 2003; An et al. 2004). The interaction between the HIF1α and p53 may thus be highly context and cell-type dependent. More severe hypoxia or anoxia promotes apoptosis independent of the HIF1α pathway. In these conditions, the electron transport chain collapses, expression of the antiapoptotic protein Mcl-1 is lost, and Bax and Bak are activated to induce apoptosis (Saikumar et al. 1998; Brunelle et al. 2007). Thus, changes in oxygen availability induce an adaptive response that may allow for extended survival, but severe or prolonged oxygen deprivation leads to apoptosis.

p53 IN METABOLIC STRESS

Metabolic stress also leads to p53 activation to induce apoptosis by up-regulating Puma, Noxa, and Bax (Nakano and Vousden 2001; Gottlieb et al. 2002; Rozan and El-Deiry 2007; Coloff et al. 2011). Lee et al. found that glucose deprivation caused the tricarboxylic acid (TCA)-cycle enzyme malate dehydrogenase (MDH) to bind to and activate acetylated p53, leading to cell-cycle arrest and apoptosis (Lee et al. 2009). AMPK activation on glucose withdrawal can also lead to p53 phosphorylation on serine 15 to cause cell-cycle arrest (Jones et al. 2005). This phosphorylation may also play a key role in cellular response to metabolic stress to induce Puma and apoptosis. It is unclear, however, if AMPK directly phosphorylates p53 or if an intermediate kinase is also involved.

Growth factor withdrawal also leads to p53 phosphorylation on serine 15 (18 in mouse) that is metabolically sensitive. We have observed Glut1/Hexokinase overexpressing cells that maintain growth factor-independent glucose metabolism can selectively suppress this phosphorylation and p53 activity after growth factor withdrawal (Mason et al. 2010). Importantly, changes in glucose metabolism also controlled p53 phosphorylation on treatment of BCR-Abl+ leukemic cells with imatinib mesylate, demonstrating that metabolic control of p53 activity may play a general role in the action of kinase inhibition in cancer therapy. This glucose-sensitive regulation of p53 occurred in part through the activation of the lipid-sensitive kinase PKCδ and was independent from canonical DNA damage-induced p53 activation, which led to p53 activation independent of PKCδ and regardless of cellular metabolic state. Romero Rosales et al. (2009) also observed a proapoptotic role for PKCδ downstream from cytokine withdrawal. Importantly, PKCδ can confer sensitivity to apoptosis after chemotherapy or radiation treatment, and the loss of PKCδ has been associated with several kinds of aggressive cancer (Gonelli et al. 2009). Although it does not appear that PKCδ directly phosphorylates p53, this pathway nevertheless provides a metabolically sensitive mechanism for p53 regulation.

Acetylation may also link metabolism to p53 regulation. Acetyl-CoA is required for acetylation and is derived from the metabolism of glucose, the β-oxidation of long-chain fatty acids, or from the conversion of the TCA-cycle intermediate citrate via the ATP citrate lyase (ACL) enzyme (Salway 2004). Thus, levels of acetyl-CoA are sensitive to changes in growth factor input or nutrient availability. Indeed, Wellen et al. (2009) have recently shown that ACL is an important source of acetyl-CoA to support histone acetylation, and loss of growth factor input or glucose availability leads to a global decrease in acetyl-CoA and protein acetylation. p53 acetylation (Gu and Roeder 1997; Sakaguchi et al. 1998; Pearson et al. 2000) is critical for p53 to induce cell-cycle arrest or apoptosis (Lavin and Gueven 2006; Tang et al. 2008) and may be metabolically regulated. Indeed, we have showed that p53 is acetylated downstream from growth factor withdrawal in a metabolically sensitive manner, leading to a transcriptional program distinct from DNA damage-induced activation (Mason et al. 2010).

METABOLIC STRESS AND NECROSIS

Ultimately, necrosis may occur when cells do not meet their minimal bioenergetic demands (Jin et al. 2007). In these instances, a collapse of ATP levels may lead to failure of ATP-dependent sodium/potassium exchangers and osmotic stress and cell rupture. Necrosis may occur in metabolically challenging situations such as ischemia/reperfusion, in which tissue is subjected to nutrient deprivation during ischemia followed by an oxidative and ROS burst during reperfusion (Zong and Thompson 2006). Demonstrating the role of ATP in preventing necrosis, Leist et al. (1997, 1999) showed in seminal studies that T cells subjected to various forms of stress died by necrosis rather than apoptosis when ATP was depleted. To respond to metabolic stress, cells activate adaptive pathways to uptake or generate more nutrients, such as autophagy or the HIF pathway. Thus, inhibiting autophagy in the presence of metabolic stress has been shown to lead to metabolic catastrophe and necrosis (Lum et al. 2005; Degenhardt et al. 2006), and a similar effect has been observed in hypoxic cells deprived of proper HIF signaling (Tennant et al. 2009).

Some cell stresses may lead to artificial nutrient depletion that can cause necrosis. In particular, DNA repair performed by poly (ADP-ribose) polymerase (PARP) can also cause necrosis if PARP activity is excessive. PARP requires cytosolic nicotinamide adenine dinucleotide (NAD+) as a substrate to repair various kinds of DNA damage including single- and double-strand breaks and base excision repair (Sodhi et al. 2010). If DNA damage is extensive, PARP can deplete NAD+, causing a collapse of glycolysis, loss of ATP production, and subsequent necrosis (Martin et al. 2000; Cipriani et al. 2005). Consistent with this ability of PARP to lead to necrosis, caspase-dependent cleavage of PARP may be a key step to prevent energy depletion and allow apoptosis to occur (Sodhi et al. 2010). Although metabolic collapse and necrosis would seem to be an attractive strategy in which to target cancer cells, excessive necrosis has been shown to lead to inflammation and promotion of advanced tumorigenesis (Balkwill et al. 2005), so treatment strategies that instead cause apoptotic death are often pursued.

CONCLUDING REMARKS

Mammalian cells rely on extracellular nutrient uptake to maintain metabolism and provide bioenergetic precursors for macromolecular synthesis. This uptake is normally under tight control of growth factors, but oncogenes and oncogenic kinases can mimic growth factor signaling to promote cell intrinsic nutrient uptake and metabolism. In particular, activation of Akt can promote glucose, amino acid, and lipid uptake (Edinger and Thompson 2002; Wieman et al. 2007). Nutrient withdrawal, either by direct means, growth factor deprivation, or oncogenic kinase inhibition, leads to metabolic stress and a set of responses that may ultimately result in cell death, and understanding these responses may be critical in efforts to exploit cancer metabolism. It is now clear that autophagy plays a key role to reduce intracellular stress and provide nutrients to replace diminished extracellular nutrient uptake, and inhibition of autophagy can enhance the proapoptotic ability of oncogenic kinase inhibitors (Kamitsuji et al. 2008; Bellodi et al. 2009; Altman et al. 2011; Guo et al. 2011). Continued nutrient deprivation will regulate Bcl-2 family proteins to induce Puma and Bim and activate Noxa while down-regulating Mcl-1 to promote apoptosis. If cells resist apoptosis or if nutrient deprivation is severe, necrosis can occur. Ultimately, understanding links between nutrient stress, autophagy, and cell death through apoptosis or necrosis will be central to inhibiting cancer cell metabolism in novel metabolic cancer therapies.

ACKNOWLEDGMENTS

We thank Drs. Pankuri Goraksha-Hicks, Andrew Macintyre, and Nancie MacIver of the Rathmell laboratory for support and comments. This work was supported by National Institutes of Health (NIH) R01CA123350 (J.C.R.), the Gabrielle’s Angel Foundation (J.C.R.), and the Leukemia and Lymphoma Foundation (J.C.R.).

Footnotes

Editors: Eric H. Baehrecke, Douglas R. Green, Sally Kornbluth, and Guy S. Salvesen

Additional Perspectives on Cell Survival and Cell Death available at www.cshperspectives.org

REFERENCES

*Reference is also in this collection.

  1. Abeliovich H, Klionsky DJ 2001. Autophagy in yeast: Mechanistic insights and physiological function. Microbiol Mol Biol Rev 65: 463–479 [DOI] [PMC free article] [PubMed] [Google Scholar]
  2. Altman BJ, Wofford JA, Zhao Y, Coloff JL, Ferguson EC, Wieman HL, Day AE, Ilkayeva O, Rathmell JC 2009. Autophagy provides nutrients but can lead to Chop-dependent induction of Bim to sensitize growth factor-deprived cells to apoptosis. Mol Biol Cell 20: 1180–1191 [DOI] [PMC free article] [PubMed] [Google Scholar]
  3. Altman BJ, Jacobs SR, Mason EF, Michalek RD, Macintyre AN, Coloff JL, Ilkayeva O, Jia W, He YW, Rathmell JC 2011. Autophagy is essential to suppress cell stress and to allow BCR-Abl-mediated leukemogenesis. Oncogene 30: 1855–1867 [DOI] [PMC free article] [PubMed] [Google Scholar]
  4. Alves NL, Derks IA, Berk E, Spijker R, van Lier RA, Eldering E 2006. The Noxa/Mcl-1 axis regulates susceptibility to apoptosis under glucose limitation in dividing T cells. Immunity 24: 703–716 [DOI] [PubMed] [Google Scholar]
  5. An J, Muoio DM, Shiota M, Fujimoto Y, Cline GW, Shulman GI, Koves TR, Stevens R, Millington D, Newgard CB 2004. Hepatic expression of malonyl-CoA decarboxylase reverses muscle, liver and whole-animal insulin resistance. Nat Med 10: 268–274 [DOI] [PubMed] [Google Scholar]
  6. Balkwill F, Charles KA, Mantovani A 2005. Smoldering and polarized inflammation in the initiation and promotion of malignant disease. Cancer Cell 7: 211–217 [DOI] [PubMed] [Google Scholar]
  7. Barnes K, McIntosh E, Whetton AD, Daley GQ, Bentley J, Baldwin SA 2005. Chronic myeloid leukaemia: An investigation into the role of Bcr-Abl-induced abnormalities in glucose transport regulation. Oncogene 24: 3257–3267 [DOI] [PubMed] [Google Scholar]
  8. Bellodi C, Lidonnici MR, Hamilton A, Helgason GV, Soliera AR, Ronchetti M, Galavotti S, Young KW, Selmi T, Yacobi R, et al. 2009. Targeting autophagy potentiates tyrosine kinase inhibitor-induced cell death in Philadelphia chromosome-positive cells, including primary CML stem cells. J Clin Invest 119: 1109–1123 [DOI] [PMC free article] [PubMed] [Google Scholar]
  9. Bernales S, McDonald KL, Walter P 2006. Autophagy counterbalances endoplasmic reticulum expansion during the unfolded protein response. PLoS Biol 4: e423. [DOI] [PMC free article] [PubMed] [Google Scholar]
  10. Berry DL, Baehrecke EH 2007. Growth arrest and autophagy are required for salivary gland cell degradation in Drosophila. Cell 131: 1137–1148 [DOI] [PMC free article] [PubMed] [Google Scholar]
  11. Beugnet A, Tee AR, Taylor PM, Proud CG 2003. Regulation of targets of mTOR (mammalian target of rapamycin) signalling by intracellular amino acid availability. Biochem J 372: 555–566 [DOI] [PMC free article] [PubMed] [Google Scholar]
  12. Bjorkoy G, Lamark T, Brech A, Outzen H, Perander M, Overvatn A, Stenmark H, Johansen T 2005. p62/SQSTM1 forms protein aggregates degraded by autophagy and has a protective effect on huntingtin-induced cell death. J Cell Biol 171: 603–614 [DOI] [PMC free article] [PubMed] [Google Scholar]
  13. Bodet L, Menoret E, Descamps G, Pellat-Deceunynck C, Bataille R, Le Gouill S, Moreau P, Amiot M, Gomez-Bougie P 2010. BH3-only protein Bik is involved in both apoptosis induction and sensitivity to oxidative stress in multiple myeloma. Br J Cancer 103: 1808–1814 [DOI] [PMC free article] [PubMed] [Google Scholar]
  14. Boya P, Gonzalez-Polo RA, Casares N, Perfettini JL, Dessen P, Larochette N, Metivier D, Meley D, Souquere S, Yoshimori T, et al. 2005. Inhibition of macroautophagy triggers apoptosis. Mol Cell Biol 25: 1025–1040 [DOI] [PMC free article] [PubMed] [Google Scholar]
  15. Brunelle JK, Shroff EH, Perlman H, Strasser A, Moraes CT, Flavell RA, Danial NN, Keith B, Thompson CB, Chandel NS 2007. Loss of Mcl-1 protein and inhibition of electron transport chain together induce anoxic cell death. Mol Cell Biol 27: 1222–1235 [DOI] [PMC free article] [PubMed] [Google Scholar]
  16. Burton TR, Gibson SB 2009. The role of Bcl-2 family member BNIP3 in cell death and disease: NIPping at the heels of cell death. Cell Death Differ 16: 515–523 [DOI] [PMC free article] [PubMed] [Google Scholar]
  17. Byfield MP, Murray JT, Backer JM 2005. hVps34 is a nutrient-regulated lipid kinase required for activation of p70 S6 kinase. J Biol Chem 280: 33076–33082 [DOI] [PubMed] [Google Scholar]
  18. Canto C, Auwerx J 2010. AMP-activated protein kinase and its downstream transcriptional pathways. Cell Mol Life Sci 67: 3407–3423 [DOI] [PMC free article] [PubMed] [Google Scholar]
  19. Cao Y, Li H, Liu H, Zheng C, Ji H, Liu X 2010. The serine/threonine kinase LKB1 controls thymocyte survival through regulation of AMPK activation and Bcl-XL expression. Cell Res 20: 99–108 [DOI] [PubMed] [Google Scholar]
  20. Caro-Maldonado A, Tait SW, Ramirez-Peinado S, Ricci JE, Fabregat I, Green DR, Munoz-Pinedo C 2010. Glucose deprivation induces an atypical form of apoptosis mediated by caspase-8 in Bax-, Bak-deficient cells. Cell Death Differ 17: 1335–1344 [DOI] [PubMed] [Google Scholar]
  21. Chen D, Li M, Luo J, Gu W 2003. Direct interactions between HIF-1α and Mdm2 modulate p53 function. J Biol Chem 278: 13595–13598 [DOI] [PubMed] [Google Scholar]
  22. Chen N, Chen X, Huang R, Zeng H, Gong J, Meng W, Lu Y, Zhao F, Wang L, Zhou Q 2009a. BCL-xL is a target gene regulated by hypoxia-inducible factor-1α. J Biol Chem 284: 10004–10012 [DOI] [PMC free article] [PubMed] [Google Scholar]
  23. Chen YQ, Zhao CL, Li W 2009b. Effect of hypoxia-inducible factor-1α on transcription of survivin in non-small cell lung cancer. J Exp Clin Cancer Res 28: 29. [DOI] [PMC free article] [PubMed] [Google Scholar]
  24. Chipuk JE, Moldoveanu T, Llambi F, Parsons MJ, Green DR 2010. The BCL-2 family reunion. Mol Cell 37: 299–310 [DOI] [PMC free article] [PubMed] [Google Scholar]
  25. Cipriani G, Rapizzi E, Vannacci A, Rizzuto R, Moroni F, Chiarugi A 2005. Nuclear poly(ADP-ribose) polymerase-1 rapidly triggers mitochondrial dysfunction. J Biol Chem 280: 17227–17234 [DOI] [PubMed] [Google Scholar]
  26. Colell A, Ricci JE, Tait S, Milasta S, Maurer U, Bouchier-Hayes L, Fitzgerald P, Guio-Carrion A, Waterhouse NJ, Li CW, et al. 2007. GAPDH and autophagy preserve survival after apoptotic cytochrome c release in the absence of caspase activation. Cell 129: 983–997 [DOI] [PubMed] [Google Scholar]
  27. Coloff JL, Mason EF, Altman BJ, Gerriets VA, Liu T, Nichols AN, Zhao Y, Wofford JA, Jacobs SR, Ilkayeva O, et al. 2011. Akt requires glucose metabolism to suppress puma expression and prevent apoptosis of leukemic T cells. J Biol Chem 286: 5921–5933 [DOI] [PMC free article] [PubMed] [Google Scholar]
  28. Dang CV, Le A, Gao P 2009. MYC-induced cancer cell energy metabolism and therapeutic opportunities. Clin Cancer Res 15: 6479–6483 [DOI] [PMC free article] [PubMed] [Google Scholar]
  29. *.Das G, Shravage BV, Baehrecke EH 2012. Regulation and function of autophagy during cell survival and cell death. Cold Spring Harb Perspect Biol 10.1101/cshperspect.a008813 [DOI] [PMC free article] [PubMed] [Google Scholar]
  30. DeBerardinis RJ, Lum JJ, Thompson CB 2006. Phosphatidylinositol 3-kinase-dependent modulation of carnitine palmitoyltransferase 1A expression regulates lipid metabolism during hematopoietic cell growth. J Biol Chem 281: 37372–37380 [DOI] [PubMed] [Google Scholar]
  31. DeBerardinis RJ, Lum JJ, Hatzivassiliou G, Thompson CB 2008. The biology of cancer: Metabolic reprogramming fuels cell growth and proliferation. Cell Metab 7: 11–20 [DOI] [PubMed] [Google Scholar]
  32. Degenhardt K, Mathew R, Beaudoin B, Bray K, Anderson D, Chen G, Mukherjee C, Shi Y, Gelinas C, Fan Y, et al. 2006. Autophagy promotes tumor cell survival and restricts necrosis, inflammation, and tumorigenesis. Cancer Cell 10: 51–64 [DOI] [PMC free article] [PubMed] [Google Scholar]
  33. Dennis JW, Nabi IR, Demetriou M 2009. Metabolism, cell surface organization, and disease. Cell 139: 1229–1241 [DOI] [PMC free article] [PubMed] [Google Scholar]
  34. Denton D, Shravage B, Simin R, Mills K, Berry DL, Baehrecke EH, Kumar S 2009. Autophagy, not apoptosis, is essential for midgut cell death in Drosophila. Curr Biol 19: 1741–1746 [DOI] [PMC free article] [PubMed] [Google Scholar]
  35. Duvel K, Yecies JL, Menon S, Raman P, Lipovsky AI, Souza AL, Triantafellow E, Ma Q, Gorski R, Cleaver S, et al. 2010. Activation of a metabolic gene regulatory network downstream of mTOR complex 1. Mol Cell 39: 171–183 [DOI] [PMC free article] [PubMed] [Google Scholar]
  36. Edinger AL, Thompson CB 2002. Akt maintains cell size and survival by increasing mTOR-dependent nutrient uptake. Mol Biol Cell 13: 2276–2288 [DOI] [PMC free article] [PubMed] [Google Scholar]
  37. Egan DF, Shackelford DB, Mihaylova MM, Gelino S, Kohnz RA, Mair W, Vasquez DS, Joshi A, Gwinn DM, Taylor R, et al. 2011. Phosphorylation of ULK1 (hATG1) by AMP-activated protein kinase connects energy sensing to mitophagy. Science 331: 456–461 [DOI] [PMC free article] [PubMed] [Google Scholar]
  38. Elstrom RL, Bauer DE, Buzzai M, Karnauskas R, Harris MH, Plas DR, Zhuang H, Cinalli RM, Alavi A, Rudin CM, et al. 2004. Akt stimulates aerobic glycolysis in cancer cells. Cancer Res 64: 3892–3899 [DOI] [PubMed] [Google Scholar]
  39. Erler JT, Cawthorne CJ, Williams KJ, Koritzinsky M, Wouters BG, Wilson C, Miller C, Demonacos C, Stratford IJ, Dive C 2004. Hypoxia-mediated down-regulation of Bid and Bax in tumors occurs via hypoxia-inducible factor 1-dependent and -independent mechanisms and contributes to drug resistance. Mol Cell Biol 24: 2875–2889 [DOI] [PMC free article] [PubMed] [Google Scholar]
  40. Fan Y, Dickman KG, Zong WX 2010. Akt and c-Myc differentially activate cellular metabolic programs and prime cells to bioenergetic inhibition. J Biol Chem 285: 7324–7333 [DOI] [PMC free article] [PubMed] [Google Scholar]
  41. Fingar DC, Salama S, Tsou C, Harlow E, Blenis J 2002. Mammalian cell size is controlled by mTOR and its downstream targets S6K1 and 4EBP1/eIF4E. Genes Dev 16: 1472–1487 [DOI] [PMC free article] [PubMed] [Google Scholar]
  42. Frech M, Andjelkovic M, Ingley E, Reddy KK, Falck JR, Hemmings BA 1997. High affinity binding of inositol phosphates and phosphoinositides to the pleckstrin homology domain of RAC/protein kinase B and their influence on kinase activity. J Biol Chem 272: 8474–8481 [DOI] [PubMed] [Google Scholar]
  43. Fulda S, Debatin KM 2007. HIF-1-regulated glucose metabolism: A key to apoptosis resistance? Cell Cycle 6: 790–792 [DOI] [PubMed] [Google Scholar]
  44. Galonek HL, Hardwick JM 2006. Upgrading the BCL-2 network. Nat Cell Biol 8: 1317–1319 [DOI] [PubMed] [Google Scholar]
  45. Ganley IG, Lam du H, Wang J, Ding X, Chen S, Jiang X 2009. ULK1.ATG13.FIP200 complex mediates mTOR signaling and is essential for autophagy. J Biol Chem 284: 12297–12305 [DOI] [PMC free article] [PubMed] [Google Scholar]
  46. Gao P, Tchernyshyov I, Chang TC, Lee YS, Kita K, Ochi T, Zeller KI, De Marzo AM, Van Eyk JE, Mendell JT, et al. 2009. c-Myc suppression of miR-23a/b enhances mitochondrial glutaminase expression and glutamine metabolism. Nature 458: 762–765 [DOI] [PMC free article] [PubMed] [Google Scholar]
  47. Gonelli A, Mischiati C, Guerrini R, Voltan R, Salvadori S, Zauli G 2009. Perspectives of protein kinase C (PKC) inhibitors as anti-cancer agents. Mini Rev Med Chem 9: 498–509 [DOI] [PubMed] [Google Scholar]
  48. Gottlieb TM, Leal JF, Seger R, Taya Y, Oren M 2002. Cross-talk between Akt, p53 and Mdm2: Possible implications for the regulation of apoptosis. Oncogene 21: 1299–1303 [DOI] [PubMed] [Google Scholar]
  49. Gottlob K, Majewski N, Kennedy S, Kandel E, Robey RB, Hay N 2001. Inhibition of early apoptotic events by Akt/PKB is dependent on the first committed step of glycolysis and mitochondrial hexokinase. Genes Dev 15: 1406–1418 [DOI] [PMC free article] [PubMed] [Google Scholar]
  50. Graeber TG, Osmanian C, Jacks T, Housman DE, Koch CJ, Lowe SW, Giaccia AJ 1996. Hypoxia-mediated selection of cells with diminished apoptotic potential in solid tumours. Nature 379: 88–91 [DOI] [PubMed] [Google Scholar]
  51. Gu W, Roeder RG 1997. Activation of p53 sequence-specific DNA binding by acetylation of the p53 C-terminal domain. Cell 90: 595–606 [DOI] [PubMed] [Google Scholar]
  52. Guo JY, Chen HY, Mathew R, Fan J, Strohecker AM, Karsli-Uzunbas G, Kamphorst JJ, Chen G, Lemmons JM, Karantza V, et al. 2011. Activated Ras requires autophagy to maintain oxidative metabolism and tumorigenesis. Genes Dev 25: 460–470 [DOI] [PMC free article] [PubMed] [Google Scholar]
  53. Hamanaka RB, Chandel NS 2010. Mitochondrial reactive oxygen species regulate cellular signaling and dictate biological outcomes. Trends Biochem Sci 35: 505–513 [DOI] [PMC free article] [PubMed] [Google Scholar]
  54. Hansson LO, Friedler A, Freund S, Rudiger S, Fersht AR 2002. Two sequence motifs from HIF-1α bind to the DNA-binding site of p53. Proc Natl Acad Sci 99: 10305–10309 [DOI] [PMC free article] [PubMed] [Google Scholar]
  55. Hao J, Song X, Song B, Liu Y, Wei L, Wang X, Yu J 2008. Effects of lentivirus-mediated HIF-1α knockdown on hypoxia-related cisplatin resistance and their dependence on p53 status in fibrosarcoma cells. Cancer Gene Ther 15: 449–455 [DOI] [PubMed] [Google Scholar]
  56. Hara K, Yonezawa K, Weng QP, Kozlowski MT, Belham C, Avruch J 1998. Amino acid sufficiency and mTOR regulate p70 S6 kinase and eIF-4E BP1 through a common effector mechanism. J Biol Chem 273: 14484–14494 [DOI] [PubMed] [Google Scholar]
  57. Hawley SA, Boudeau J, Reid JL, Mustard KJ, Udd L, Makela TP, Alessi DR, Hardie DG 2003. Complexes between the LKB1 tumor suppressor, STRAD α/β and MO25 α/β are upstream kinases in the AMP-activated protein kinase cascade. J Biol 2: 28. [DOI] [PMC free article] [PubMed] [Google Scholar]
  58. Hou YC, Chittaranjan S, Barbosa SG, McCall K, Gorski SM 2008. Effector caspase Dcp-1 and IAP protein Bruce regulate starvation-induced autophagy during Drosophila melanogaster oogenesis. J Cell Biol 182: 1127–1139 [DOI] [PMC free article] [PubMed] [Google Scholar]
  59. Hoyer-Hansen M, Jaattela M 2007a. AMP-activated protein kinase: A universal regulator of autophagy? Autophagy 3: 381–383 [DOI] [PubMed] [Google Scholar]
  60. Hoyer-Hansen M, Jaattela M 2007b. Connecting endoplasmic reticulum stress to autophagy by unfolded protein response and calcium. Cell Death Differ 14: 1576–1582 [DOI] [PubMed] [Google Scholar]
  61. Imamura K, Ogura T, Kishimoto A, Kaminishi M, Esumi H 2001. Cell cycle regulation via p53 phosphorylation by a 5′-AMP activated protein kinase activator, 5-aminoimidazole- 4-carboxamide-1-β-D-ribofuranoside, in a human hepatocellular carcinoma cell line. Biochem Biophys Res Commun 287: 562–567 [DOI] [PubMed] [Google Scholar]
  62. Ishihara T, Hoshino T, Namba T, Tanaka K, Mizushima T 2007. Involvement of up-regulation of PUMA in non-steroidal anti-inflammatory drug-induced apoptosis. Biochem Biophys Res Commun 356: 711–717 [DOI] [PubMed] [Google Scholar]
  63. Jin S, DiPaola RS, Mathew R, White E 2007. Metabolic catastrophe as a means to cancer cell death. J Cell Sci 120: 379–383 [DOI] [PMC free article] [PubMed] [Google Scholar]
  64. Jones RG, Plas DR, Kubek S, Buzzai M, Mu J, Xu Y, Birnbaum MJ, Thompson CB 2005. AMP-activated protein kinase induces a p53-dependent metabolic checkpoint. Mol Cell 18: 283–293 [DOI] [PubMed] [Google Scholar]
  65. Jung CH, Jun CB, Ro SH, Kim YM, Otto NM, Cao J, Kundu M, Kim DH 2009. ULK-Atg13-FIP200 complexes mediate mTOR signaling to the autophagy machinery. Mol Biol Cell 20: 1992–2003 [DOI] [PMC free article] [PubMed] [Google Scholar]
  66. Kamitsuji Y, Kuroda J, Kimura S, Toyokuni S, Watanabe K, Ashihara E, Tanaka H, Yui Y, Watanabe M, Matsubara H, et al. 2008. The Bcr-Abl kinase inhibitor INNO-406 induces autophagy and different modes of cell death execution in Bcr-Abl-positive leukemias. Cell Death Differ 15: 1712–1722 [DOI] [PubMed] [Google Scholar]
  67. Karantza-Wadsworth V, Patel S, Kravchuk O, Chen G, Mathew R, Jin S, White E 2007. Autophagy mitigates metabolic stress and genome damage in mammary tumorigenesis. Genes Dev 21: 1621–1635 [DOI] [PMC free article] [PubMed] [Google Scholar]
  68. Karawajew L, Rhein P, Czerwony G, Ludwig WD 2005. Stress-induced activation of the p53 tumor suppressor in leukemia cells and normal lymphocytes requires mitochondrial activity and reactive oxygen species. Blood 105: 4767–4775 [DOI] [PubMed] [Google Scholar]
  69. Kaufman RJ, Back SH, Song B, Han J, Hassler J 2010. The unfolded protein response is required to maintain the integrity of the endoplasmic reticulum, prevent oxidative stress and preserve differentiation in β-cells. Diabetes Obes Metab 12: 99–107 [DOI] [PMC free article] [PubMed] [Google Scholar]
  70. Kelekar A 2005. Autophagy. Ann NY Acad Sci 1066: 259–271 [DOI] [PubMed] [Google Scholar]
  71. Kim E, Goraksha-Hicks P, Li L, Neufeld TP, Guan KL 2008. Regulation of TORC1 by Rag GTPases in nutrient response. Nat Cell Biol 10: 935–945 [DOI] [PMC free article] [PubMed] [Google Scholar]
  72. Kim MJ, Park IJ, Yun H, Kang I, Choe W, Kim SS, Ha J 2010. AMP-activated protein kinase antagonizes pro-apoptotic extracellular signal-regulated kinase activation by inducing dual-specificity protein phosphatases in response to glucose deprivation in HCT116 carcinoma. J Biol Chem 285: 14617–14627 [DOI] [PMC free article] [PubMed] [Google Scholar]
  73. Kim J, Kundu M, Viollet B, Guan KL 2011. AMPK and mTOR regulate autophagy through direct phosphorylation of Ulk1. Nat Cell Biol 13: 132–141 [DOI] [PMC free article] [PubMed] [Google Scholar]
  74. Kiyono K, Suzuki HI, Matsuyama H, Morishita Y, Komuro A, Kano MR, Sugimoto K, Miyazono K 2009. Autophagy is activated by TGF-β and potentiates TGF-β-mediated growth inhibition in human hepatocellular carcinoma cells. Cancer Res 69: 8844–8852 [DOI] [PubMed] [Google Scholar]
  75. Komatsu M, Waguri S, Chiba T, Murata S, Iwata J, Tanida I, Ueno T, Koike M, Uchiyama Y, Kominami E, et al. 2006. Loss of autophagy in the central nervous system causes neurodegeneration in mice. Nature 441: 880–884 [DOI] [PubMed] [Google Scholar]
  76. Lavin MF, Gueven N 2006. The complexity of p53 stabilization and activation. Cell Death Differ 13: 941–950 [DOI] [PubMed] [Google Scholar]
  77. Lee H, Paik SG 2006. Regulation of BNIP3 in normal and cancer cells. Mol Cells 21: 1–6 [PubMed] [Google Scholar]
  78. Lee SM, Kim JH, Cho EJ, Youn HD 2009. A nucleocytoplasmic malate dehydrogenase regulates p53 transcriptional activity in response to metabolic stress. Cell Death Differ 16: 738–748 [DOI] [PubMed] [Google Scholar]
  79. Leist M, Single B, Castoldi AF, Kuhnle S, Nicotera P 1997. Intracellular adenosine triphosphate (ATP) concentration: A switch in the decision between apoptosis and necrosis. J Exp Med 185: 1481–1486 [DOI] [PMC free article] [PubMed] [Google Scholar]
  80. Leist M, Single B, Naumann H, Fava E, Simon B, Kuhnle S, Nicotera P 1999. Inhibition of mitochondrial ATP generation by nitric oxide switches apoptosis to necrosis. Exp Cell Res 249: 396–403 [DOI] [PubMed] [Google Scholar]
  81. Letai A, Bassik MC, Walensky LD, Sorcinelli MD, Weiler S, Korsmeyer SJ 2002. Distinct BH3 domains either sensitize or activate mitochondrial apoptosis, serving as prototype cancer therapeutics. Cancer Cell 2: 183–192 [DOI] [PubMed] [Google Scholar]
  82. Liang XH, Jackson S, Seaman M, Brown K, Kempkes B, Hibshoosh H, Levine B 1999. Induction of autophagy and inhibition of tumorigenesis by beclin 1. Nature 402: 672–676 [DOI] [PubMed] [Google Scholar]
  83. Liu B, Chen Y, St Clair DK 2008a. ROS and p53: A versatile partnership. Free Radic Biol Med 44: 1529–1535 [DOI] [PMC free article] [PubMed] [Google Scholar]
  84. Liu L, Ning X, Sun L, Zhang H, Shi Y, Guo C, Han S, Liu J, Sun S, Han Z, et al. 2008b. Hypoxia-inducible factor-1α contributes to hypoxia-induced chemoresistance in gastric cancer. Cancer Sci 99: 121–128 [DOI] [PMC free article] [PubMed] [Google Scholar]
  85. Lock R, Roy S, Kenific CM, Su JS, Salas E, Ronen SM, Debnath J 2011. Autophagy facilitates glycolysis during Ras-mediated oncogenic transformation. Mol Biol Cell 22: 165–178 [DOI] [PMC free article] [PubMed] [Google Scholar]
  86. Lowman XH, McDonnell MA, Kosloske A, Odumade OA, Jenness C, Karim CB, Jemmerson R, Kelekar A 2010. The proapoptotic function of Noxa in human leukemia cells is regulated by the kinase Cdk5 and by glucose. Mol Cell 40: 823–833 [DOI] [PubMed] [Google Scholar]
  87. Lum JJ, Bauer DE, Kong M, Harris MH, Li C, Lindsten T, Thompson CB 2005. Growth factor regulation of autophagy and cell survival in the absence of apoptosis. Cell 120: 237–248 [DOI] [PubMed] [Google Scholar]
  88. Lynch CJ 2001. Role of leucine in the regulation of mTOR by amino acids: Revelations from structure-activity studies. J Nutr 131: 861S–865S [DOI] [PubMed] [Google Scholar]
  89. Martin DR, Lewington AJ, Hammerman MR, Padanilam BJ 2000. Inhibition of poly(ADP-ribose) polymerase attenuates ischemic renal injury in rats. Am J Physiol Regul Integr Comp Physiol 279: R1834–R1840 [DOI] [PubMed] [Google Scholar]
  90. Mason EF, Zhao Y, Goraksha-Hicks P, Coloff JL, Gannon H, Jones SN, Rathmell JC 2010. Aerobic glycolysis suppresses p53 activity to provide selective protection from apoptosis upon loss of growth signals or inhibition of BCR-Abl. Cancer Res 70: 8066–8076 [DOI] [PMC free article] [PubMed] [Google Scholar]
  91. Mathew R, Kongara S, Beaudoin B, Karp CM, Bray K, Degenhardt K, Chen G, Jin S, White E 2007. Autophagy suppresses tumor progression by limiting chromosomal instability. Genes Dev 21: 1367–1381 [DOI] [PMC free article] [PubMed] [Google Scholar]
  92. Mathew R, Karp CM, Beaudoin B, Vuong N, Chen G, Chen HY, Bray K, Reddy A, Bhanot G, Gelinas C, et al. 2009. Autophagy suppresses tumorigenesis through elimination of p62. Cell 137: 1062–1075 [DOI] [PMC free article] [PubMed] [Google Scholar]
  93. Melillo G 2007. Targeting hypoxia cell signaling for cancer therapy. Cancer Metastasis Rev 26: 341–352 [DOI] [PubMed] [Google Scholar]
  94. Michalek RD, Rathmell JC 2010. The metabolic life and times of a T-cell. Immunol Rev 236: 190–202 [DOI] [PMC free article] [PubMed] [Google Scholar]
  95. Munoz-Pinedo C, Ruiz-Ruiz C, Ruiz de Almodovar C, Palacios C, Lopez-Rivas A 2003. Inhibition of glucose metabolism sensitizes tumor cells to death receptor-triggered apoptosis through enhancement of death-inducing signaling complex formation and apical procaspase-8 processing. J Biol Chem 278: 12759–12768 [DOI] [PubMed] [Google Scholar]
  96. Nakano K, Vousden KH 2001. PUMA, a novel proapoptotic gene, is induced by p53. Mol Cell 7: 683–694 [DOI] [PubMed] [Google Scholar]
  97. Nezis IP, Shravage BV, Sagona AP, Lamark T, Bjorkoy G, Johansen T, Rusten TE, Brech A, Baehrecke EH, Stenmark H 2010. Autophagic degradation of dBruce controls DNA fragmentation in nurse cells during late Drosophila melanogaster oogenesis. J Cell Biol 190: 523–531 [DOI] [PMC free article] [PubMed] [Google Scholar]
  98. Niizuma K, Endo H, Chan PH 2009. Oxidative stress and mitochondrial dysfunction as determinants of ischemic neuronal death and survival. J Neurochem 109: 133–138 [DOI] [PMC free article] [PubMed] [Google Scholar]
  99. Nobukuni T, Joaquin M, Roccio M, Dann SG, Kim SY, Gulati P, Byfield MP, Backer JM, Natt F, Bos JL, et al. 2005. Amino acids mediate mTOR/raptor signaling through activation of class 3 phosphatidylinositol 3OH-kinase. Proc Natl Acad Sci 102: 14238–14243 [DOI] [PMC free article] [PubMed] [Google Scholar]
  100. Nutt LK, Margolis SS, Jensen M, Herman CE, Dunphy WG, Rathmell JC, Kornbluth S 2005. Metabolic regulation of oocyte cell death through the CaMKII-mediated phosphorylation of caspase-2. Cell 123: 89–103 [DOI] [PMC free article] [PubMed] [Google Scholar]
  101. Nutt LK, Buchakjian MR, Gan E, Darbandi R, Yoon SY, Wu JQ, Miyamoto YJ, Gibbons JA, Andersen JL, Freel CD, et al. 2009. Metabolic control of oocyte apoptosis mediated by 14-3-3ζ-regulated dephosphorylation of caspase-2. Dev Cell 16: 856–866 [DOI] [PMC free article] [PubMed] [Google Scholar]
  102. Okoshi R, Ozaki T, Yamamoto H, Ando K, Koida N, Ono S, Koda T, Kamijo T, Nakagawara A, Kizaki H 2008. Activation of AMP-activated protein kinase induces p53-dependent apoptotic cell death in response to energetic stress. J Biol Chem 283: 3979–3987 [DOI] [PubMed] [Google Scholar]
  103. Osthus RC, Shim H, Kim S, Li Q, Reddy R, Mukherjee M, Xu Y, Wonsey D, Lee LA, Dang CV 2000. Deregulation of glucose transporter 1 and glycolytic gene expression by c-Myc. J Biol Chem 275: 21797–21800 [DOI] [PubMed] [Google Scholar]
  104. Oyadomari S, Mori M 2004. Roles of CHOP/GADD153 in endoplasmic reticulum stress. Cell Death Differ 11: 381–389 [DOI] [PubMed] [Google Scholar]
  105. Pearson M, Carbone R, Sebastiani C, Cioce M, Fagioli M, Saito S, Higashimoto Y, Appella E, Minucci S, Pandolfi PP, et al. 2000. PML regulates p53 acetylation and premature senescence induced by oncogenic Ras. Nature 406: 207–210 [DOI] [PubMed] [Google Scholar]
  106. Plas DR, Talapatra S, Edinger AL, Rathmell JC, Thompson CB 2001. Akt and Bcl-xL promote growth factor-independent survival through distinct effects on mitochondrial physiology. J Biol Chem 276: 12041–12048 [DOI] [PubMed] [Google Scholar]
  107. Pradelli LA, Beneteau M, Chauvin C, Jacquin MA, Marchetti S, Munoz-Pinedo C, Auberger P, Pende M, Ricci JE 2010. Glycolysis inhibition sensitizes tumor cells to death receptors-induced apoptosis by AMP kinase activation leading to Mcl-1 block in translation. Oncogene 29: 1641–1652 [DOI] [PubMed] [Google Scholar]
  108. Pua HH, Guo J, Komatsu M, He YW 2009. Autophagy is essential for mitochondrial clearance in mature T lymphocytes. J Immunol 182: 4046–4055 [DOI] [PubMed] [Google Scholar]
  109. Puthalakath H, O'Reilly LA, Gunn P, Lee L, Kelly PN, Huntington ND, Hughes PD, Michalak EM, McKimm-Breschkin J, Motoyama N, et al. 2007. ER stress triggers apoptosis by activating BH3-only protein Bim. Cell 129: 1337–1349 [DOI] [PubMed] [Google Scholar]
  110. Raff MC 1992. Social controls on cell survival and cell death. Nature 356: 397–400 [DOI] [PubMed] [Google Scholar]
  111. Rathmell JC, Vander Heiden MG, Harris MH, Frauwirth KA, Thompson CB 2000. In the absence of extrinsic signals, nutrient utilization by lymphocytes is insufficient to maintain either cell size or viability. Mol Cell 6: 683–692 [DOI] [PubMed] [Google Scholar]
  112. Rathmell JC, Fox CJ, Plas DR, Hammerman PS, Cinalli RM, Thompson CB 2003. Akt-directed glucose metabolism can prevent Bax conformation change and promote growth factor-independent survival. Mol Cell Biol 23: 7315–7328 [DOI] [PMC free article] [PubMed] [Google Scholar]
  113. Ren D, Tu HC, Kim H, Wang GX, Bean GR, Takeuchi O, Jeffers JR, Zambetti GP, Hsieh JJ, Cheng EH 2010. BID, BIM, and PUMA are essential for activation of the BAX- and BAK-dependent cell death program. Science 330: 1390–1393 [DOI] [PMC free article] [PubMed] [Google Scholar]
  114. Rey S, Semenza GL 2010. Hypoxia-inducible factor-1-dependent mechanisms of vascularization and vascular remodelling. Cardiovasc Res 86: 236–242 [DOI] [PMC free article] [PubMed] [Google Scholar]
  115. Robey RB, Hay N 2006. Mitochondrial hexokinases, novel mediators of the antiapoptotic effects of growth factors and Akt. Oncogene 25: 4683–4696 [DOI] [PubMed] [Google Scholar]
  116. Roccio M, Bos JL, Zwartkruis FJ 2006. Regulation of the small GTPase Rheb by amino acids. Oncogene 25: 657–664 [DOI] [PubMed] [Google Scholar]
  117. Romero Rosales K, Peralta ER, Guenther GG, Wong SY, Edinger AL 2009. Rab7 activation by growth factor withdrawal contributes to the induction of apoptosis. Mol Biol Cell 20: 2831–2840 [DOI] [PMC free article] [PubMed] [Google Scholar]
  118. Rouschop KM, Ramaekers CH, Schaaf MB, Keulers TG, Savelkouls KG, Lambin P, Koritzinsky M, Wouters BG 2009. Autophagy is required during cycling hypoxia to lower production of reactive oxygen species. Radiother Oncol 92: 411–416 [DOI] [PubMed] [Google Scholar]
  119. Rozan LM, El-Deiry WS 2007. p53 downstream target genes and tumor suppression: A classical view in evolution. Cell Death Differ 14: 3–9 [DOI] [PubMed] [Google Scholar]
  120. Sade H, Sarin A 2004. Reactive oxygen species regulate quiescent T-cell apoptosis via the BH3-only proapoptotic protein BIM. Cell Death Differ 11: 416–423 [DOI] [PubMed] [Google Scholar]
  121. Saikumar P, Dong Z, Patel Y, Hall K, Hopfer U, Weinberg JM, Venkatachalam MA 1998. Role of hypoxia-induced Bax translocation and cytochrome c release in reoxygenation injury. Oncogene 17: 3401–3415 [DOI] [PubMed] [Google Scholar]
  122. Sakaguchi K, Herrera JE, Saito S, Miki T, Bustin M, Vassilev A, Anderson CW, Appella E 1998. DNA damage activates p53 through a phosphorylation-acetylation cascade. Genes Dev 12: 2831–2841 [DOI] [PMC free article] [PubMed] [Google Scholar]
  123. Salway JG 2004. Metabolism at a glance. Blackwell, Malden, MA [Google Scholar]
  124. Sancak Y, Bar-Peled L, Zoncu R, Markhard AL, Nada S, Sabatini DM 2010. Ragulator-Rag complex targets mTORC1 to the lysosomal surface and is necessary for its activation by amino acids. Cell 141: 290–303 [DOI] [PMC free article] [PubMed] [Google Scholar]
  125. Schafer ZT, Kornbluth S 2006. The apoptosome: Physiological, developmental, and pathological modes of regulation. Dev Cell 10: 549–561 [DOI] [PubMed] [Google Scholar]
  126. Scott RC, Juhasz G, Neufeld TP 2007. Direct induction of autophagy by Atg1 inhibits cell growth and induces apoptotic cell death. Curr Biol 17: 1–11 [DOI] [PMC free article] [PubMed] [Google Scholar]
  127. Sendoel A, Kohler I, Fellmann C, Lowe SW, Hengartner MO 2010. HIF-1 antagonizes p53-mediated apoptosis through a secreted neuronal tyrosinase. Nature 465: 577–583 [DOI] [PMC free article] [PubMed] [Google Scholar]
  128. Shang L, Chen S, Du F, Li S, Zhao L, Wang X 2011. Nutrient starvation elicits an acute autophagic response mediated by Ulk1 dephosphorylation and its subsequent dissociation from AMPK. Proc Natl Acad Sci 108: 4788–4793 [DOI] [PMC free article] [PubMed] [Google Scholar]
  129. Shaw RJ 2009. LKB1 and AMP-activated protein kinase control of mTOR signalling and growth. Acta Physiol (Oxf) 196: 65–80 [DOI] [PMC free article] [PubMed] [Google Scholar]
  130. Shaw RJ, Kosmatka M, Bardeesy N, Hurley RL, Witters LA, DePinho RA, Cantley LC 2004. The tumor suppressor LKB1 kinase directly activates AMP-activated kinase and regulates apoptosis in response to energy stress. Proc Natl Acad Sci 101: 3329–3335 [DOI] [PMC free article] [PubMed] [Google Scholar]
  131. 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: 1131–1135 [DOI] [PMC free article] [PubMed] [Google Scholar]
  132. Sodhi RK, Singh N, Jaggi AS 2010. Poly(ADP-ribose) polymerase-1 (PARP-1) and its therapeutic implications. Vascul Pharmacol 53: 77–87 [DOI] [PubMed] [Google Scholar]
  133. Suzuki H, Tomida A, Tsuruo T 2001. Dephosphorylated hypoxia-inducible factor 1α as a mediator of p53-dependent apoptosis during hypoxia. Oncogene 20: 5779–5788 [DOI] [PubMed] [Google Scholar]
  134. Tal MC, Sasai M, Lee HK, Yordy B, Shadel GS, Iwasaki A 2009. Absence of autophagy results in reactive oxygen species-dependent amplification of RLR signaling. Proc Natl Acad Sci 106: 2770–2775 [DOI] [PMC free article] [PubMed] [Google Scholar]
  135. Tandon P, Gallo CA, Khatri S, Barger JF, Yepiskoposyan H, Plas DR 2011. Requirement for ribosomal protein S6 kinase 1 to mediate glycolysis and apoptosis resistance induced by Pten deficiency. Proc Natl Acad Sci 108: 2361–2365 [DOI] [PMC free article] [PubMed] [Google Scholar]
  136. Tang Y, Zhao W, Chen Y, Zhao Y, Gu W 2008. Acetylation is indispensable for p53 activation. Cell 133: 612–626 [DOI] [PMC free article] [PubMed] [Google Scholar]
  137. Tennant DA, Frezza C, MacKenzie ED, Nguyen QD, Zheng L, Selak MA, Roberts DL, Dive C, Watson DG, Aboagye EO, et al. 2009. Reactivating HIF prolyl hydroxylases under hypoxia results in metabolic catastrophe and cell death. Oncogene 28: 4009–4021 [DOI] [PubMed] [Google Scholar]
  138. Tracy K, Dibling BC, Spike BT, Knabb JR, Schumacker P, Macleod KF 2007. BNIP3 is an RB/E2F target gene required for hypoxia-induced autophagy. Mol Cell Biol 27: 6229–6242 [DOI] [PMC free article] [PubMed] [Google Scholar]
  139. van Delft MF, Huang DC 2006. How the Bcl-2 family of proteins interact to regulate apoptosis. Cell Res 16: 203–213 [DOI] [PubMed] [Google Scholar]
  140. Vander Heiden MG, Plas DR, Rathmell JC, Fox CJ, Harris MH, Thompson CB 2001. Growth factors can influence cell growth and survival through effects on glucose metabolism. Mol Cell Biol 21: 5899–5912 [DOI] [PMC free article] [PubMed] [Google Scholar]
  141. Vander Heiden MG, Cantley LC, Thompson CB 2009. Understanding the Warburg effect: The metabolic requirements of cell proliferation. Science 324: 1029–1033 [DOI] [PMC free article] [PubMed] [Google Scholar]
  142. von Harsdorf R, Li PF, Dietz R 1999. Signaling pathways in reactive oxygen species-induced cardiomyocyte apoptosis. Circulation 99: 2934–2941 [DOI] [PubMed] [Google Scholar]
  143. Walensky LD 2006. BCL-2 in the crosshairs: Tipping the balance of life and death. Cell Death Differ 13: 1339–1350 [DOI] [PubMed] [Google Scholar]
  144. Wang Y, Han R, Liang ZQ, Wu JC, Zhang XD, Gu ZL, Qin ZH 2008a. An autophagic mechanism is involved in apoptotic death of rat striatal neurons induced by the non-N-methyl-D-aspartate receptor agonist kainic acid. Autophagy 4: 214–226 [DOI] [PubMed] [Google Scholar]
  145. Wang Y, Singh R, Massey AC, Kane SS, Kaushik S, Grant T, Xiang Y, Cuervo AM, Czaja MJ 2008b. Loss of macroautophagy promotes or prevents fibroblast apoptosis depending on the death stimulus. J Biol Chem 283: 4766–4777 [DOI] [PMC free article] [PubMed] [Google Scholar]
  146. Warburg O 1956. On the origin of cancer cells. Science 123: 309–314 [DOI] [PubMed] [Google Scholar]
  147. Wellen KE, Thompson CB 2010. Cellular metabolic stress: Considering how cells respond to nutrient excess. Mol Cell 40: 323–332 [DOI] [PMC free article] [PubMed] [Google Scholar]
  148. Wellen KE, Hatzivassiliou G, Sachdeva UM, Bui TV, Cross JR, Thompson CB 2009. ATP-citrate lyase links cellular metabolism to histone acetylation. Science 324: 1076–1080 [DOI] [PMC free article] [PubMed] [Google Scholar]
  149. Wellen KE, Lu C, Mancuso A, Lemons JM, Ryczko M, Dennis JW, Rabinowitz JD, Coller HA, Thompson CB 2010. The hexosamine biosynthetic pathway couples growth factor-induced glutamine uptake to glucose metabolism. Genes Dev 24: 2784–2799 [DOI] [PMC free article] [PubMed] [Google Scholar]
  150. Wensveen FM, Alves NL, Derks IA, Reedquist KA, Eldering E 2011. Apoptosis induced by overall metabolic stress converges on the Bcl-2 family proteins Noxa and Mcl-1. Apoptosis 16: 708–721 [DOI] [PMC free article] [PubMed] [Google Scholar]
  151. Wieman HL, Wofford JA, Rathmell JC 2007. Cytokine stimulation promotes glucose uptake via phosphatidylinositol-3 kinase/Akt regulation of Glut1 activity and trafficking. Mol Biol Cell 18: 1437–1446 [DOI] [PMC free article] [PubMed] [Google Scholar]
  152. Wise DR, Thompson CB 2010. Glutamine addiction: A new therapeutic target in cancer. Trends Biochem Sci 35: 427–433 [DOI] [PMC free article] [PubMed] [Google Scholar]
  153. Wise DR, DeBerardinis RJ, Mancuso A, Sayed N, Zhang XY, Pfeiffer HK, Nissim I, Daikhin E, Yudkoff M, McMahon SB, et al. 2008. Myc regulates a transcriptional program that stimulates mitochondrial glutaminolysis and leads to glutamine addiction. Proc Natl Acad Sci 105: 18782–18787 [DOI] [PMC free article] [PubMed] [Google Scholar]
  154. Wofford JA, Wieman HL, Jacobs SR, Zhao Y, Rathmell JC 2008. IL-7 promotes Glut1 trafficking and glucose uptake via STAT5-mediated activation of Akt to support T-cell survival. Blood 111: 2101–2111 [DOI] [PMC free article] [PubMed] [Google Scholar]
  155. Woods A, Johnstone SR, Dickerson K, Leiper FC, Fryer LG, Neumann D, Schlattner U, Wallimann T, Carlson M, Carling D 2003. LKB1 is the upstream kinase in the AMP-activated protein kinase cascade. Curr Biol 13: 2004–2008 [DOI] [PubMed] [Google Scholar]
  156. Yue Z, Jin S, Yang C, Levine AJ, Heintz N 2003. Beclin 1, an autophagy gene essential for early embryonic development, is a haploinsufficient tumor suppressor. Proc Natl Acad Sci 100: 15077–15082 [DOI] [PMC free article] [PubMed] [Google Scholar]
  157. Zhao Y, Altman BJ, Coloff JL, Herman CE, Jacobs SR, Wieman HL, Wofford JA, Dimascio LN, Ilkayeva O, Kelekar A, et al. 2007. Glycogen synthase kinase 3α and 3β mediate a glucose-sensitive antiapoptotic signaling pathway to stabilize Mcl-1. Mol Cell Biol 27: 4328–4339 [DOI] [PMC free article] [PubMed] [Google Scholar]
  158. Zhao Y, Coloff JL, Ferguson EC, Jacobs SR, Cui K, Rathmell JC 2008. Glucose metabolism attenuates p53 and Puma-dependent cell death upon growth factor deprivation. J Biol Chem 283: 36344–36353 [DOI] [PMC free article] [PubMed] [Google Scholar]
  159. Zoncu R, Efeyan A, Sabatini DM 2011. mTOR: From growth signal integration to cancer, diabetes and ageing. Nat Rev Mol Cell Biol 12: 21–35 [DOI] [PMC free article] [PubMed] [Google Scholar]
  160. Zong WX, Thompson CB 2006. Necrotic death as a cell fate. Genes Dev 20: 1–15 [DOI] [PubMed] [Google Scholar]

Articles from Cold Spring Harbor Perspectives in Biology are provided here courtesy of Cold Spring Harbor Laboratory Press

RESOURCES