Abstract
The relationship between cellular metabolism and the cell cycle machinery is by no means unidirectional. The ability of a cell to enter the cell cycle critically depends on the availability of metabolites. Conversely, the cell cycle machinery commits to regulating metabolic networks in order to support cell survival and proliferation. In this review, we will give an account of how the cell cycle machinery and metabolism are interconnected. Acquiring information on how communication takes place among metabolic signaling networks and the cell cycle controllers is crucial to increase our understanding of the deregulation thereof in disease, including cancer.
Keywords: cell cycle, cyclin-dependent kinases, glycolysis, metabolism, nutrients, proliferation
Resting cells require a basal level of catabolic metabolism to ensure energy homeostasis. Cells that commit to entering the cell cycle, however, differ greatly from resting cells in terms of their metabolic profile, as they will eventually have to double their cell content, that is, their DNA, membranes, organelles and other biomass. To support the energy-consuming processes needed for this program, cells increase the uptake of glucose and glutamine and shut down oxidative metabolism. In this way, glucose and glutamine-derived metabolic intermediates can be used for the biosynthesis of macromolecules required for the cell division. Highly proliferating cells, including cancer cells but also activated lymphocytes, thymocytes and embryonic cells, preferentially use glycolysis even in the presence of oxygen.1-8 This phenomenon is called aerobic glycolysis or “the Warburg effect”.9
In unicellular organisms, cell cycle progression is dependent on the availability of nutrients, which directly couples available resources to the generation of progeny. For example, stationary-phase yeast switches to a mitotic phenotype when exposed to glucose, but becomes quiescent or sporulates when no other nutrients are provided.10 Under nutrient-steady growth conditions, cycling yeast cells display fluctuations in oxygen consumption, alternating between glycolysis and respiration. Their cell division is solely limited to the glycolytic phase, with DNA replication taking place only during that period.11 Interestingly, many genes identified in classic screens for factors regulating the cell cycle in yeast, were later shown to have a function in metabolic regulation, too.12-17 Also, transcriptome studies demonstrated that yeast genes involved in glycolysis respiration, lipids and amino acid synthesis are expressed as a function of the cell cycle.18,19 Taken together, these observations show that in unicellular organisms, intimate connections between cell cycle and metabolism must exist.
In contrast to single-cell eukaryotes, cells of multicellular organisms usually have an unlimited access to nutrients. However, they are not cell-autonomous for nutrient uptake but instead depend on proliferation-regulating pathways. Mitogen-mediated activation of signaling routes triggers nutrient uptake and represents the rate-limiting cue for cell cycle entry.20 As a consequence, growth factor-stimulated cells initiate cell division in a fashion comparable to that of yeast exposed to a nutrient-rich medium.21,22 Accordingly, in the absence of mitogens, even in a nutrient-rich environment, cells will not enter the cell cycle.23 On the other hand, even in the presence of promitogenic cues, glucose deprivation will keep cells from proliferating, which is a widely used method for synchronizing mammalian cells.24 The fact that signaling pathways coordinating cell cycle progression control, and are controlled by, changes in cellular metabolism25,26 shows that, also in multicellular organisms, there must be a crosstalk between these pathways, cell cycle and metabolism. Yet, the molecular basis that connects nutrient availability, biosynthetic intermediates and energetic balance to the core cell cycle machinery remains incompletely understood. Here, we will give an overview of how the cell cycle machinery and metabolism are interconnected.
Cell Cycle Regulation of Metabolism
Evidence is emerging in support of the coordinated temporal regulation of metabolism directly by the cell cycle modulators. A first indication for this came from the observation that in yeast, metabolites of nucleotide, protein and lipid synthesis are cyclically fluctuating, as a function of cell cycle progression.27 Indeed, it has been shown subsequently that the glycolysis-promoting enzyme 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase 3 (PFKFB3) is subjected to cell cycle dependent temporal regulation by members of the ubiquitin proteasome system (UPS; Figure 1, upper panel).28,29 Since then, a number of mechanisms have been revealed that couple the cellular metabolic state to the cell cycle (Fig. 2).
Figure 1.
Protein activities and metabolic events during the cell cycle. A schematic representation of the temporal regulation of metabolic factors (upper panel) and the cell cycle machinery (lower panel). The represented protein levels are not relative, but rather indicate their relative timing of expression.
Figure 2.
Bidirectional regulation of the cell cycle machinery and metabolic enzymes. Interactions between the cell cycle machinery and metabolic enzymes indicated by arrows may be either direct or indirect. Color codes represent proteins belonging to the cell cycle machinery or to metabolism.
Most somatic cells are differentiated and quiescent, that is, they reside in the G0 phase of the cell cycle. Following mitogenic stimulation, cells typically re-enter the cell cycle and proceed through the G1 phase, in which the stage is set for DNA replication. Upon passage through the G1/S restriction point, cells enter S phase in which they double their DNA content, move on into the G2 phase and the final mitotic (M) phase, in which cellular contents are divided over 2 daughter cells (Fig. 2). Key proteins for the tight regulation of the cell cycle are cyclin-dependent kinases (CDKs), which associate with one of different cyclins across the cell cycle to ensure accurate cell cycle progression.30-33 The kinase activity of cyclin-CDK complexes is tightly regulated by a plethora of CDK inhibitors (CKIs), which stop cell cycle progression in unfavorable circumstances.34
D-type cyclins
The role of D-type cyclins in metabolism was first demonstrated in cyclin D-deficient mice that display marked metabolic phenotypes. Cyclin D2-deficient mice show a diabetic phenotype due to impaired pancreatic β-cell expansion and function, which is further enhanced by cyclin D1 co-depletion.35 Cyclin D3-deficient mice display reduced adipocyte size and increased sensitivity to insulin, which is a consequence of the inactivation of peroxisome proliferator-activated receptor (PPAR)γ, the master regulator of adipogenesis (Fig. 2).36 Accordingly, depletion of cyclin D3 diminishes PPARγ activity and adipogenesis, while cyclin D3 overexpression has the opposing effect. Cyclin D1 is highly expressed in breast cancer cells; among other functions, it decreases the abundance and activity of the glycolytic enzyme hexokinase 2 (HK2; Fig. 2).37 Correspondingly, cyclin D1 depletion in either normal or oncogenic breast cells leads to an increase in glycolytic enzyme pyruvate kinase (PK) as well as the lipogenic enzymes acetyl-CoA carboxylase (ACC) and fatty acid synthase levels (FASN; Fig. 2).37 In hepatocytes, cyclin D1 inhibits the glucose-mediated induction of central lipogenic genes via repression of the carbohydrate response element binding protein (ChREBP) and hepatocyte nuclear factor 4α (HNF4α), which are important regulators of glucose sensing and lipid metabolism (Fig. 2).38 Hence, cyclin D1 inhibits both glycolysis and lipogenesis. Moreover, it hinders mitochondrial biogenesis and functions through inhibition of the nuclear respiratory factor 1 (NRF1), which regulates nuclear-encoded mitochondrial genes, and the mitochondrial voltage-dependent anion channel (VDAC), respectively (Fig. 2).39,40 Finally, unbiased mass-spectrometry analysis of proteins interacting with cyclin D1 using cyclin D1 knockin mice revealed interactions between cyclin D1 and numerous metabolic proteins, such as lipogenic enzymes FASN and ACC, as well as the mitochondrial electron transport chain components cytochrome c oxidase (COX) and ATP synthase (Fig. 2).41 These observations together demonstrate that cyclin D1, by hindering glycolysis, lipogenesis as well as mitochondrial activity, downregulates metabolic activity through several routes.
Restraining the conversion of glucose to lipids for storage by D-type cyclins might allow cells to use glucose-derived metabolites for doubling their cell content for cell division to take place. Alternatively, cyclin D-mediated inhibition of metabolism might provide a negative feedback loop to ensure unidirectionality of cell cycle progression.42 Cyclin D activity accumulates in G1, as a function of growth factor signaling, hence when metabolic activity is abundant (Fig. 1).43 If metabolic activity increases cyclin D expression and, in turn, cyclin D inhibits metabolism, a rise in cyclin D in G1 would shut down metabolism, thereby preventing re-entry into G1 until the G1/S transition is complete.
pRB-E2F
Cyclin D-CDK4/6 complexes regulate the phosphorylation state of the retinoblastoma tumor suppressor (pRB) in G1; its phosphorylation abolishes inhibition of physically associated E2F/DP transcription factors, thereby allowing the expression of genes required for DNA synthesis.44,45 Thus, phosphorylation of pRB in late G1 by cyclin D-CDK4/6 converts E2F/DP from transcriptional repressor into activator, thereby promoting S-phase entry (Fig. 2).
Given that E2F transcription factors are involved in cell cycle progression and survival, it is not surprising that roles for increased E2F activity have been shown in cancer.45 It is well established that E2F promotes (cancer) cell proliferation through transcriptional activation of genes involved in cell cycle regulation and DNA synthesis. But a role for E2F in metabolism has also emerged. E2F1, as well as its upstream activator CDK4, has been shown to regulate adipogenesis by positively regulating PPARγ.46,47 In accordance with this, pRB represses PPARγ at the early stages of adipocyte differentiation.48 On the other hand, pRB acts positively on terminal adipocyte differentiation by binding directly to the transcription factor CCAAT/enhancer binding protein (C/EBP)β, thereby facilitating its transactivation.49
Besides adipogenesis, the E2F-pRB pathway is involved in the glucose metabolism of pancreatic β-cells. E2F1-depleted mice display a diabetic phenotype, resulting from glucose intolerance and deficient insulin secretion.50 In line with this, E2F1 regulates Kir6.2, a key component of the KATP channel controlling glucose-induced insulin secretion (Fig. 2).51 Expression of Kir6.2 is lost in the pancreas of E2F1-depleted mice, resulting in insulin secretion defects. Furthermore, E2F1 was shown to induce expression of pyruvate dehydrogenase kinase 4 (PDK4). PDK4 is one of 4 kinases that inhibit pyruvate dehydrogenase (PDH) and thereby oxidative metabolism (Fig. 2).52 Likewise, inactivation of pRB by the oncogenic E1A adenoviral protein triggers PDK4 expression (Fig. 2). Induction of PDK4 by pRB-E2F1 in myoblasts diverts pyruvate away from mitochondrial oxidation, thereby increasing lactate production and flux into biosynthetic pathways.53 Furthermore, E2F1 stimulates glycolysis by upregulating the expression of the phosphofructokinase 2 PFK2 enzyme (Fig. 2).54 Depletion of E2F1 increases the expression of regulators of mitochondrial biogenesis and function, such as mitochondrial topoisomerase I (TopImt).55 Correspondingly, E2F1 downregulates oxidative and increases glycolytic genes expression.56 In this way, the pRB-E2F1 pathway promotes a switch from oxidative to glycolytic metabolism, thereby supporting the metabolic phenotype typical of proliferating cells.
Altogether, the pRB-E2F pathway provides yet another mechanism for the coordinated regulation of metabolism throughout the cell cycle by facilitating glycolysis and repressing oxidative phosphorylation (Fig. 2). pRB-E2F-mediated stimulation of adipogenesis and inhibition of oxidative metabolism are in line with the observations made for cyclin D and suggest that the latter acts on these metabolic pathways at least to a significant extent through pRB-E2F. However, while pRB-E2F promotes glycolysis, cyclin D suppresses it. This implies that cyclin D acts on glycolytic enzymes in a pRB-E2F-independent manner.
CKIs
As cyclin-CDK complexes regulate metabolism, and because the activities and functions of CDK-cyclin complexes are regulated by the CKIs, the latter should also be associated with the cellular metabolism. Indeed, p21Cip1 and p27Kip1 were demonstrated to modulate adipocyte differentiation, since loss of either kinase inhibitor in mice induces adipocyte hyperplasia.57 In accordance with this, combined disruption of p21Cip1 and p27Kip1 in mice induces an increase in the number of adipocytes and the development of hypercholesterolemia, glucose intolerance and insulin insensitivity, which are features of obesity. However, these observations require further investigation, as other studies have shown that p21Cip1 null mice are less prone to obesity induced by lipid-rich diet, whereas p27Kip1 null mice do not show hyperplasia of adipocytes but increased insulin secretion that prevents hyperglycemia in diabetic mice models.58-60
The UPS and glycolysis
Cyclins and CKIs are both subject to tight temporal control and degradation by the UPS. Two ubiquitin ligases are crucial in the cell cycle. The ligase anaphase-promoting complex or cyclosome (APC/C) regulates both the transition through G1 and the exit from M phase by degrading both S- and M-phase cyclins (cyclin A and B) and Securin (allowing chromosome separation). Directing APC/C to correct substrates at specific time points in the cell cycle depends on one of 2 activators, CDC20 or CDC20 homolog (CDH1).61,62 The latter is expressed and active in mitosis, until it is replaced by CDH1 in late mitosis and the G1 phase (Figs. 1 and 2).63 The ligase Skp1/cullin/F-box protein (SCF) complex controls the G1/S-phase and the G2/M transition. The F-box protein of the SCF complex regulates its substrate recognition.64,65 SCF-Skp2 mainly ubiquitinates and degrades CKIs such as p27Kip1 and p21Cip1, but also cyclin E, whereas SCF-β-TrCP positively regulates APC/CCDC20 and CDK1 to ensure G2/M transition (Fig. 2). APC/C and SCF control each other to regulate progression of the cell cycle.66
The yeast F-box protein GRR1, a component of SCF complex regulating G1/S transition, was first identified as being essential for adaptation to nutrient availability. In response to extracellular glucose, SCFGRR1 blocks exit from the cell cycle and sporulation by targeting Ime2p kinase.67 At the same time, SCFGRR1 represses genes required for the utilization of alternative carbon sources, and upregulates hexose transporters.68-71 On the other hand, when glucose is removed, SCFGRR1-mediated degradation of Ime2p is abrogated, cells exit the cell cycle and sporulation proceeds.67 Moreover, following glucose deprivation, SCFGRR1 inhibits glycolysis by degrading Pfk27, the yeast homolog of the glycolytic enzyme PFKFB3 (Fig. 2).28 Thus, SCFGRR1 in yeasts regulates both cell cycle exit and metabolism as a function of glucose availability.
Shortly after Pfk27 was identified as the target of SCFGRR1 in yeast, PFKFB3 was found to be a degraded by APC/CCDH1 in neurons (Fig. 2).29 Constitutive breakdown of PFKFB3 helps terminally differentiated cortical neurons to preserve their low glycolytic state. It also prevents oxidative damage by redirecting glucose-derived metabolites into antioxidants-providing pentose phosphate pathway (PPP). In dividing cells, PFKFB3 is directed for degradation both by APC/CCDH1 during late mitosis and G1 and subsequently by SCFβ-TrCP in late S phase (Figs. 1 and 2).72-75 Overexpression of the APC/C activator CDH1 leads to degradation of PFKFB3 and thereby restricts glycolysis. Accordingly, depletion of CDH1 promotes glycolysis in a PFKFB3-dependent manner and stimulates cells to enter S phase.73,74 Inactivation of APC/CCDH1, resulting from a phosphorylation of CDH1 that occurs in late G1, leads to the accumulation of PFKFB3 and consequently promotes both proliferation and glycolysis (Fig. 1). In late S-phase, PFKFB3 levels drop again due to an increase in activity of SCFβ-TrCP, which specifically directs PFKFB3 for degradation (Fig. 1).72,75 Restricted expression of PFKFB3 to the specific window of late G1 and early S phase generates a peak in anaerobic glycolysis during the G1/S transition and a PPP peak in S phase (Fig. 1, upper panel). Thus, the joint action of the APC/C and SCF complexes on PFKFB3 coordinates metabolic activity and cell cycle progression.
APC/CCDH1 was recently shown to also regulate phosphatase and transactivator EYA1.76 The level of EYA1 protein oscillates in the cell cycle, peaking during mitosis and dropping radically as cells enter G1, when APC/CCDH1 reaches its peak level (Figs. 1 and 2). While depletion of CDH1 stabilizes the EYA1 protein, overexpression of CDH1 reduces its levels. Thus, APC/CCDH1 degrades EYA1 precisely during M/G1 transition. EYA1 is required for proliferation during embryogenesis77-79 and its level is elevated in several cancers.80-83 Interestingly, EYA1 is also known to reprogram the aerobic metabolism of slow-twitch muscle fibers, which depend on lipid oxidation, into the glycolytic phenotype of fast-twitch muscle fibers, which depend mostly on glycogen as an energy source. This is achieved through the upregulation of the glycolytic enzymes aldolase A and β-enolase (Fig. 2).84 Although only correlative, this might provide another mechanism by which UPS regulates glycolysis.
The UPS and glutaminolysis
Besides PFKFB3, the APC/CCDH1 also directs the glutaminolytic enzyme glutaminase 1 (GLS1) for degradation during mitotic exit and G1 (Figs. 1 and 2).74,75 Glutamine is important for proliferation as it replenishes tricarboxylic acid (TCA) cycle intermediates used for macrosyntheses of amino acids, lipids and nucleotides. GLS1 levels and the glutaminolysis rate rise after APC/CCDH1 activity declines in late G1. Accordingly, depletion of APC/C activator CDH1 leads to an increase in cellular GLS1 concentration and consequently glutamine metabolism. In contrast to PFKFB3, GLS1 is not a substrate for SCF and therefore it is not degraded through the S and G2 phases, but only when the cells progress to the G2/M transition (Figs. 1 and 2). The distinct regulation of PFKFB3 and GLS1 proteins and therefore glycolysis and glutaminolysis, suggests the different functions of glucose and glutamine at particular phases of the cell cycle. Indeed, studies in synchronized cells show that both glucose and glutamine are required throughout G1, whereas only glutamine is needed to progress through S phase into the cell division phase.75 The importance of regulating GLS1 for cell proliferation is further supported by the notion that GLS1 C, an isoform that is not targeted by the APC/CCDH1, is overexpressed in several tumors.85
Taken together, while ubiquitination by the UPS was originally recognized for its major role in regulating the cell cycle machinery, it is now also acknowledged for its integrated regulation of metabolism and proliferation. The dual regulation by these 2 ubiquitin ligases, APC/C and the SCF complex, delineates a differential regulation of both aerobic glycolysis and glutaminolysis during distinct phases of the cell cycle. In this sense, the UPS plays a major role in the provision of a specific metabolic profile reminiscent of proliferating (cancer) cells, including the Warburg effect, upregulation of the PPP and increased glutamine utilization.
p53 family members
p53 is a major tumor suppressor protein, which is mutated in many types of cancer. The activation of p53 in response to a plethora of stress signals provides the cell with 2 options: inhibition of the cell cycle at G1/S by inducing transcription of the CKI p21Cip1, or induction of pro-apoptotic signals.86 This allows cells to either repair the damage before engaging in cellular division, or, if the damage is beyond repair, to prevent cells from proliferating ever again. But recent research shows that p53 acts also as a major regulator of cellular metabolism.
Several functions of p53 have been demonstrated to silence glycolysis and promote oxidative metabolism. It hampers glycolytic flux through the various steps of glycolytic pathway by downregulating the expression of glucose transporters GLUT 1 and GLUT4, decreasing levels of the glycolytic enzyme phosphoglycerate mutase (PGM).87,88 It also induces TIGAR, an enzyme that lowers the level of glycolytic activator fructose-2,6-bisphosphate (F-2,6-BP) and thereby inhibits glycolysis (Fig. 2).89 Moreover, p53-mediated repression of monocarboxylate transporter 1 (MCT1) expression prevents the secretion of lactate under anaerobic conditions, which also reduces glycolysis.90 Inhibition of the glycolytic pathway would be expected to be associated with redirecting glucose-derived metabolites into PPP pathway. However, p53 evades this by binding and inhibiting the rate-limiting enzyme of PPP, glucose-6-phosphate dehydrogenase (G6PDH; Fig. 2).91 p53 also downregulates the expression of the ribonucleotide reductase subunit M2 (RRM2), an enzyme controlling dNTP synthesis (Fig. 2).92 Inhibition of glycolysis and PPP is paralleled by a role of p53 in promoting oxidative metabolism. Next to maintaining mitochondrial balance,93 p53 stimulates oxidative phosphorylation by upregulating cytochrome c oxidase (mammalian COX/yeast SCO2), a component of the mitochondrial electron transport chain (Fig. 2).94,95 Moreover, p53 increases the TCA cycle rate by downregulating PDH-inhibitory kinase PDK2 and TCA cycle - associated malic enzymes (MEs; Fig. 2).96,97 Additionally, p53 increases the expression of glutaminolytic enzyme glutaminase 2 (GLS2; Fig. 2).98 GLS2 converts glutamine to glutamate, which can feed the TCA cycle, but can also participate in glutathione synthesis to control the redox state of the cell. Thus, by upregulating GLS2, p53 influences both glutamine metabolism and redox status. p53 also regulates the cellular redox state upon serine withdrawal. It does so by allowing de novo serine to be channeled to glutathione synthesis, which occurs at the cost of nucleotide synthesis.99 Finally, p53 was shown to negatively regulate lipogenesis (Fig. 2). p53 inhibits fatty acid (FA) synthesis in mouse adipose tissue by suppressing the expression levels of transcription factor sterol regulatory element-binding protein (SREBP1c) and FASN and ATP citrate lyase (ACLY) (Fig. 2).100 At the same time, p53 stimulates FA oxidation by inducing the expression of carnitine palmitoyltransferase (CPT), which is responsible for the transport of FA into the mitochondria, and guanidinoacetate methyltransferase (GAMT), an enzyme involved in creatine synthesis (Fig. 2).101,102 Hence, p53 promotes cell cycle arrest not only by acting on the cell cycle machinery, but also by counteracting the general metabolic profiles favorable for proliferation and by supporting oxidative metabolic reactions characteristic of resting cells.
The other p53 family members p63 and p73, too, have metabolic functions, broadening the impact of the p53 family on cell metabolism. The Tp63 and Tp73 genes are transcribed from 2 separate promoters, encoding either full- length proteins that retain a full transactivation (TA) domain (TAp63 and TAp73) or N-terminally truncated isoforms (ΔNp63 and ΔNp73). Both TAp63 and TAp73 upregulate the expression of GLS2 and thereby increase glutaminolysis.103,104 Moreover, TAp73 stimulates oxidative metabolism by upregulating COX subunit 4105. Correspondingly, the depletion of TAp73 results in a decrease in both respiration and ATP production. In contrast to p53, TAp73 activates the expression G6PDH and the flux to PPP, therefore redirecting glucose for the synthesis of nucleotides and antioxidants.106 TAp73 also regulates amino acid metabolism by increasing the levels of serine, glycine, and glutathione.107 Similarly to p53, p63 was implemented in the regulation of lipid metabolism. Loss of TAp63 disrupts lipogenesis, FA synthesis and oxidation, and protects against insulin resistance in mice.108 Likewise, ΔNp63 transcriptionally activates FASN.109 Altogether, the p53 family provides yet another element contributing to direct crosstalk between the cell cycle machinery and cellular metabolism.
Metabolic Regulation of the Cell Cycle Machinery
While it is important for a cell to provide sufficient building blocks to enable cell division, this mechanism also functions vice versa: it is equally important for a cell to adapt its cell cycle to the environment (i.e., changes in nutrient availability) and the state of metabolism. As early as in 1974 it was shown that cells in the absence of glucose arrest at the G1/S restriction point110. This demonstrated that glucose availability governs as a metabolic cell cycle checkpoint. Since then, various mechanism through which cells synchronize their cell cycle with their metabolic state have been discovered, several of which are described below.
PFKFB3
Beyond its metabolic activity, PFKFB3 has been described to localize in the nucleus and regulate the cell cycle machinery.111 Overexpression of nuclear PFKFB3 is accompanied by increased expression of G1-promoting cyclin D3 (Fig. 2). Moreover, nuclear PFKFB3 stimulates proliferation by increasing the expression of mitotic kinase CDK1 and M phase-promoting phosphatase Cdc25C and by decreasing the expression of the CDK1 inhibitor p27Kip1 (Fig. 2). These effects are completely abrogated by mutating either the active site or nuclear localization residues of PFKFB3, demonstrating a requirement for nuclear delivery of F-2,6-BP in this setting. Consequently, addition of F-2,6-BP to cell lysates promotes CDK1‐mediated p27Kip1 phosphorylation, which is a cue for p27Kip1 degradation. Along these lines, suppression of PFKFB3 induces cell cycle delay.72 These data show that, while PFKFB3 is subject to a tight regulation by the UPS, it can return this action by regulating the cell cycle machinery.
PKM2
PKs catalyze the final rate-limiting step of glycolysis, generating ATP and pyruvate. PK isoform M2 (PKM2) is specifically enriched in highly proliferating cells, including cancer cells, where it regulates aerobic glycolysis.112 Apart from its important metabolic function, PKM2 also has a non-metabolic role in the control of the cell cycle progression. In response to epidermal growth factor (EGF) stimulation, PKM2 translocates to the nucleus where it binds to β-catenin and promotes its transcriptional activity.113,114 PKM2-β-catenin complex subsequently localizes to the cyclin D1 (CCND1) and c-Myc promoters and enhances their expression through detachment of a histone deacetylase, HDAC3.115 Thus, by inducing cyclin D1 expression, PKM2 regulates the G1-S phase transition (Fig. 2). Besides PKM2s role in regulating the expression of cyclin D1, its direct involvement in the regulation of cell cycle progression has also been demonstrated. During mitosis, PKM2 binds to and phosphorylates the spindle assembly checkpoint (SAC) protein Bub3 (Fig. 2).116 This phosphorylation is required for the formation of Bub3-Bub1-Blinkin complex and correct kinetochore microtubules attachment, allowing for proper chromosomal segregation. In this way, PKM2 regulates not only G1/S transition but also progression through mitosis.
Acetyl-CoA
Cytosolic and nuclear acetyl-CoA is not only an important intermediate for macrosyntheses, but also a precursor for the posttranslational modification of proteins by acetylation. For example, acetylation of histones is dependent on acetyl-CoA-producing ACLY. In various mammalian cell types, depletion of ACLY, and therefore a drop in acetyl-CoA levels, decreases histone acetylation.117 On the other hand, in yeast, a rise in acetyl-CoA upon depletion of ACC increases the acetylation of histones.118 Notably, acetylation of histones is an essential process for the release of DNA for replication and therefore for cell cycle progression.119,120 Thus, by regulating the acetylation of histones, acetyl-CoA controls the cell cycle (Fig. 2).
In addition to acetylation, availability of acetyl-CoA regulates the glycosylation of proteins (Fig. 2). O-linked N-acetylglucosamine (O-GlcNAc) modifications depend on UDP-GlcNAc, production of which is controlled by the availability of glucose, glutamine and acetyl-CoA.121 O-GlcNAc-transferases (OGTs) transfer UDP-GlcNAc onto a target protein, while O-GlcNAcases (OGAs) remove it, permitting a dynamic regulation of O-GlcNAcylation levels. In recent years, numerous proteins have been identified as substrates for OGlcNAc modification, including regulators of cell cycle progression like p53.122 Moreover, O-GlcNAcylation levels have been found to vary along the cell cycle (Fig. 1). Serum addition triggers G0/G1 transition by activating PI3K/AKT and MAPK pathways and transcription of cyclin D1. OGT levels are significantly increased following serum stimulation,123 while OGT depletion delays G1 entry and prevents serum-induced cyclin D1 synthesis (Fig. 2).124 Hence, OGT and therefore increased O-GlcNAcylation, are vital for entry into the cell cycle. Contrary to that, at the G1/S transition, global O-GlcNAcylation is decreased due to elevated OGA activity.123 Interestingly, this leads to lower O-GlcNAcylation of histones, which, similarly to the histone acetylation mentioned above, promotes the relaxation of DNA, necessary for replication and cell cycle progression.125,126 At the G2/M checkpoint, O-GlcNAcylation reaches its peak.127-130 Accordingly, depletion of OGT decreases cyclin B1 expression and therefore impairs G2/M transition (Fig. 2).127 Although we are only beginning to understand the importance of O-GlcNAcylation in regulating cell proliferation, the evidence discussed provides additional indications that the cell cycle machinery and metabolism are tightly intertwined.
ATP/AMP ratio
The ATP/AMP ratio reflects the energy status of the cell. The protein complex that plays a crucial role in regulating cellular energy is AMP-activated protein kinase (AMPK).131 Under conditions of energetic stress such as glucose deprivation and low ATP levels, activated AMPK negatively regulates energy-consuming metabolic processes such as protein and lipid synthesis, while at the same time it promotes energy-producing oxidative phosphorylation and FA oxidation (Fig. 2).132 It has been recently suggested that AMPK can regulate energy levels also by direct control of the cell cycle machinery. Along these lines, activation of AMPK upon glucose deprivation or treatment with AMP analog AICAR causes an arrest in the G1 phase.133,134 This cessation of proliferation is associated with phosphorylation of p53 at Ser15 and upregulation of CKI p21Cip1 expression, a target of activated p53 (Fig. 2). AMPK phosphorylates also the C-terminal residue of another CKI, p27Kip1, causing its stabilization (Fig. 2).135 Likewise, methylene blue (MB)-mediated activation of AMPK reduces expression of cyclins A2, B1 and D1, leading to proliferative arrest.136 Moreover, activation of AMPK due to mitochondrial dysfunction promotes p53-dependent transcription of the F‐box protein archipelago. Archipelago then recruits cyclin E to the SCF complex, resulting in cyclin E degradation and G1-S cell cycle arrest (Fig. 2).137,138 As such, AMPK plays a pivotal role as a metabolic cell cycle checkpoint, preventing cell cycle entry in conditions of low nutrient availability.
NAD+/NADH ratio
The NAD+/ NADH ratio has an important role in the regulation of redox homeostasis stress and is often considered to be a readout of the metabolic status. NAD+ is converted to NADH in catabolic reactions including glycolysis and the TCA cycle. To maintain a proper redox state, NADH is regenerated constantly via several mechanisms, such as oxidation in the mitochondrial respiratory chain and reduction of pyruvate to lactate in the last step of glycolysis.139 NAD+ is a classical coenzyme mediating redox reactions,139 but also plays an important role in regulation of NAD+ - consuming enzymes, including sirtuin family of NAD+-dependent deacetylases.140 Notably, in recent years, several mechanisms linking sirtuins to the cell cycle machinery have been described. SIRT2 controls mitotic exit and acts as a checkpoint protein in cells treated with microtubule poisons.141,142 Under genotoxic stress, SIRT1 deacetylates and hinders the activity of p53, thereby preventing p53-mediated transactivation of p21Cip1 and cell cycle arrest (Fig. 2).143,144 SIRT1 also regulates the cell cycle by deacetylation of the forkhead box O 3 (FOXO3) transcription factor.145 In response to oxidative stress, SIRT1 binds to and deacetylates FOXO3, resulting in increased levels of FOXO3 target p27Kip1 and induction of cell cycle arrest (Fig. 2). Moreover, SIRT1 has been described to regulate the components of the circadian clock machinery.146,147 Interestingly, among clock-controlled genes are those that have an essential role in cell cycle control, including cyclin D1 and inhibitor of CDK1-cyclin B1 complex, Wee1.148,149 Hence, by regulating the circadian clock, SIRT1 inhibits cells cycle progression in situations of stress (Fig. 2). Taken together, sirtuins translate the NAD+/NADH ratio to the components of the cell cycle machinery.
Concluding Remarks
The combined regulation of metabolic events and components of the cell cycle machinery described here delineates an intricate relationship between the 2. Their reciprocal regulation plays a pivotal role in the cellular decision to enter and progress through cell cycle and to adjust metabolic pathways in response to both intracellular and external cues. Cells strictly control the cell cycle machinery components, in order to produce the required macromolecules during specific stages of the cell cycle. However, sometimes the regulation is disturbed, which leads to severe pathologies, including cancer. Thus, studying the reciprocal regulatory networks linking the cell cycle and metabolism is likely to reveal potential targets for therapy.
Disclosure of Potential Conflicts of Interest
No potential conflicts of interest were disclosed.
Funding
This work was supported by a Vici grant from the Netherlands Organization for Scientific Research (NWO) and a Queen Wilhelmina Award grant from the Dutch Cancer Society (KWF Kankerbestrijding) to DSP.
References
- 1.Vander Heiden MG, Cantley LC, Thompson CB. Understanding the Warburg effect: the metabolic requirements of cell proliferation. Science 2009; 324:1029-33; PMID:19460998; http://dx.doi.org/ 10.1126/science.1160809 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Cantor JR, Sabatini DM. Cancer cell metabolism: one hallmark, many faces. Cancer Discov 2012; 2:881-98; PMID:23009760; http://dx.doi.org/ 10.1158/2159-8290.CD-12-0345 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Ward PS, Thompson CB. Metabolic reprogramming: a cancer hallmark even warburg did not anticipate. Cancer Cell 2012; 21:297-308; PMID:22439925; http://dx.doi.org/ 10.1016/j.ccr.2012.02.014 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Hedeskov CJ. Early effects of phytohaemagglutinin on glucose metabolism of normal human lymphocytes. Biochem J 1968; 110:373-80; PMID:5726214 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Brand K. Glutamine and glucose metabolism during thymocyte proliferation. Pathways of glutamine and glutamate metabolism. Biochem J 1985; 228:353-61; PMID:2861809 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Wang T, Marquardt C, Foker J. Aerobic glycolysis during lymphocyte proliferation. Nature 1976. June 24;261(5562):702-5; http://dx.doi.org/ 10.1038/261702a0 [DOI] [PubMed] [Google Scholar]
- 7.Steck TL, Kaufman S, Bader JP. Glycolysis in chick embryo cell cultures transformed by Rous sarcoma virus. Cancer Res 1968; 28:1611-9; PMID:4299826 [PubMed] [Google Scholar]
- 8.Kondoh H, Lleonart ME, Nakashima Y, Yokode M, Tanaka M, Bernard D, Gil J, Beach D. A high glycolytic flux supports the proliferative potential of murine embryonic stem cells. Antioxid Redox Signal 2007; 9:293-9; PMID:17184172; http://dx.doi.org/ 10.1089/ars.2006.1467 [DOI] [PubMed] [Google Scholar]
- 9.Warburg O, Wind F, Negelein E. The metabolism of tumors in the body. J Gen Physiol 1927; 8:519-30; PMID:19872213; http://dx.doi.org/ 10.1085/jgp.8.6.519 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Granot D, Snyder M. Glucose induces cAMP-independent growth-related changes in stationary-phase cells of Saccharomyces cerevisiae. Proc Natl Acad Sci USA 1991; 88:5724-8; PMID:1648229; http://dx.doi.org/ 10.1073/pnas.88.13.5724 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Chen Z, Odstrcil EA, Tu BP, McKnight SL. Restriction of DNA replication to the reductive phase of the metabolic cycle protects genome integrity. Science 2007; 316:1916-9; PMID:17600220; http://dx.doi.org/ 10.1126/science.1140958 [DOI] [PubMed] [Google Scholar]
- 12.Hartwell LH, Culotti J, Pringle JR, Reid BJ. Genetic control of the cell division cycle in yeast. Science 1974; 183:46-51; PMID:4587263; http://dx.doi.org/ 10.1126/science.183.4120.46 [DOI] [PubMed] [Google Scholar]
- 13.Jong AY, Kuo CL, Campbell JL. The CDC8 gene of yeast encodes thymidylate kinase. J Biol Chem 1984; 259:11052-9; PMID:6088527 [PubMed] [Google Scholar]
- 14.Maitra PK, Lobo Z. Pyruvate kinase mutants of Saccharomyces cerevisiae: biochemical and genetic characterisation. Mol Gen Genet 1977; 152:193-200; PMID:327275; http://dx.doi.org/ 10.1007/BF00268817 [DOI] [PubMed] [Google Scholar]
- 15.Game JC. Yeast cell-cycle mutant cdc21 is a temperature-sensitive thymidylate auxotroph. Mol Gen Genet 1976; 146:313-5; PMID:794696; http://dx.doi.org/ 10.1007/BF00701257 [DOI] [PubMed] [Google Scholar]
- 16.Dickinson JR, Williams AS. The cdc30 mutation in Saccharomyces cerevisiae results in a temperature-sensitive isoenzyme of phosphoglucose isomerase. J Gen Microbiol 1987; 133:135-40; PMID:3309140 [DOI] [PubMed] [Google Scholar]
- 17.Wrobel C, Schmidt EV, Polymenis M. CDC64 encodes cytoplasmic alanyl-tRNA synthetase, Ala1p, of Saccharomyces cerevisiae. J Bacteriol 1999; 181:7618-20; PMID:10601222 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Cho RJ, Campbell MJ, Winzeler EA, Steinmetz L, Conway A, Wodicka L, Wolfsberg TG, Gabrielian AE, Landsman D, Lockhart DJ, et al.. A genome-wide transcriptional analysis of the mitotic cell cycle. Mol Cell 1998; 2:65-73; PMID:9702192; http://dx.doi.org/ 10.1016/S1097-2765(00)80114-8 [DOI] [PubMed] [Google Scholar]
- 19.Spellman PT, Sherlock G, Zhang MQ, Iyer VR, Anders K, Eisen MB, Brown PO, Botstein D, Futcher B. Comprehensive identification of cell cycle-regulated genes of the yeast Saccharomyces cerevisiae by microarray hybridization. Mol Biol Cell 1998; 9:3273-97; PMID:9843569; http://dx.doi.org/ 10.1091/mbc.9.12.3273 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Lloyd AC. The regulation of cell size. Cell 2013; 154:1194-205; PMID:24034244; http://dx.doi.org/ 10.1016/j.cell.2013.08.053 [DOI] [PubMed] [Google Scholar]
- 21.Boer VM, Crutchfield CA, Bradley PH, Botstein D, Rabinowitz JD. Growth-limiting intracellular metabolites in yeast growing under diverse nutrient limitations. Mol Biol Cell 2010; 21:198-211; PMID:19889834; http://dx.doi.org/ 10.1091/mbc.E09-07-0597 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Conlon I, Raff M. Differences in the way a mammalian cell and yeast cells coordinate cell growth and cell-cycle progression. J Biol 2003; 2:7; PMID:12733998; http://dx.doi.org/ 10.1186/1475-4924-2-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Rathmell JC, Vander Heiden MG, Harris MH, Frauwirth KA, Thompson CB. In the absence of extrinsic signals, nutrient utilization by lymphocytes is insufficient to maintain either cell size or viability. Mol Cell 2000; 6:683-92; PMID:11030347; http://dx.doi.org/ 10.1016/S1097-2765(00)00066-6 [DOI] [PubMed] [Google Scholar]
- 24.Langan TJ, Chou RC. Synchronization of mammalian cell cultures by serum deprivation. Methods Mol Biol 2011; 761:75-83; PMID:21755442; http://dx.doi.org/ 10.1007/978-1-61779-182-6_5 [DOI] [PubMed] [Google Scholar]
- 25.Levine AJ, Puzio-Kuter AM. The Control of the Metabolic Switch in Cancers by Oncogenes and Tumor Suppressor Genes. Science 2010; 330:1340-4; PMID:21127244; http://dx.doi.org/ 10.1126/science.1193494 [DOI] [PubMed] [Google Scholar]
- 26.Jones RG, Thompson CB. Tumor suppressors and cell metabolism: a recipe for cancer growth. Genes Dev 2009; 23:537-48; PMID:19270154; http://dx.doi.org/ 10.1101/gad.1756509 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Tu BP, Mohler RE, Liu JC, Dombek KM, Young ET, Synovec RE, McKnight SL. Cyclic changes in metabolic state during the life of a yeast cell. Proc Natl Acad Sci USA 2007; 104:16886-91; PMID:17940006; http://dx.doi.org/ 10.1073/pnas.0708365104 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Benanti JA, Cheung SK, Brady MC, Toczyski DP. A proteomic screen reveals SCFGrr1 targets that regulate the glycolytic-gluconeogenic switch. Nat Cell Biol 2007; 9:1184-91; PMID:17828247; http://dx.doi.org/ 10.1038/ncb1639 [DOI] [PubMed] [Google Scholar]
- 29.Herrero-Mendez A, Almeida A, Fernández E, Maestre C, Moncada S, Bolaños JP. The bioenergetic and antioxidant status of neurons is controlled by continuous degradation of a key glycolytic enzyme by APC/C-Cdh1. Nat Cell Biol 2009; 11:747-52; PMID:19448625; http://dx.doi.org/ 10.1038/ncb1881 [DOI] [PubMed] [Google Scholar]
- 30.Hartwell LH, Weinert TA. Checkpoints: controls that ensure the order of cell cycle events. Science 1989; 246:629-34; PMID:2683079; http://dx.doi.org/ 10.1126/science.2683079 [DOI] [PubMed] [Google Scholar]
- 31.Malumbres M, Barbacid M. Cell cycle, CDKs and cancer: a changing paradigm. Nat Rev Cancer 2009; 9:153-66; PMID:19238148; http://dx.doi.org/ 10.1038/nrc2602 [DOI] [PubMed] [Google Scholar]
- 32.Sherr CJ. G1 phase progression: cycling on cue. Cell 1994; 79:551-5; PMID:7954821; http://dx.doi.org/ 10.1016/0092-8674(94)90540-1 [DOI] [PubMed] [Google Scholar]
- 33.Lim S, Kaldis P. Cdks, cyclins and CKIs: roles beyond cell cycle regulation. Development 2013; 140:3079-93; PMID:23861057; http://dx.doi.org/ 10.1242/dev.091744 [DOI] [PubMed] [Google Scholar]
- 34.Sherr CJ, Roberts JM. CDK inhibitors: positive and negative regulators of G1-phase progression. Genes Dev 1999; 13:1501-12; PMID:10385618; http://dx.doi.org/ 10.1101/gad.13.12.1501 [DOI] [PubMed] [Google Scholar]
- 35.Kushner JA, Ciemerych MA, Sicinska E, Wartschow LM, Teta M, Long SY, Sicinski P, White MF. Cyclins D2 and D1 are essential for postnatal pancreatic β-cell growth. Mol Cell Biol 2005; 25:3752-62; PMID:15831479; http://dx.doi.org/ 10.1128/MCB.25.9.3752-3762.2005 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Sarruf DA, Iankova I, Abella A, Assou S, Miard S, Fajas L. Cyclin D3 promotes adipogenesis through activation of peroxisome proliferator-activated receptor gamma. Mol Cell Biol 2005; 25:9985-95; PMID:16260612; http://dx.doi.org/ 10.1128/MCB.25.22.9985-9995.2005 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Sakamaki T, Casimiro MC, Ju X, Quong AA, Katiyar S, Liu M, Jiao X, Li A, Zhang X, Lu Y, et al.. Cyclin D1 determines mitochondrial function in vivo. Mol Cell Biol 2006; 26:5449-69; PMID:16809779; http://dx.doi.org/ 10.1128/MCB.02074-05 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Hanse EA, Mashek DG, Becker JR, Solmonson AD, Mullany LK, Mashek MT, Towle HC, Chau AT, Albrecht JH. Cyclin D1 inhibits hepatic lipogenesis via repression of carbohydrate response element binding protein and hepatocyte nuclear factor 4α. cc 2012; 11:2681-90; http://dx.doi.org/ 10.4161/cc.21019 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Wang C, Li Z, Lu Y, Du R, Katiyar S, Yang J, Fu M, Leader JE, Quong A, Novikoff PM, et al.. Cyclin D1 repression of nuclear respiratory factor 1 integrates nuclear DNA synthesis and mitochondrial function. Proc Natl Acad Sci USA 2006; 103:11567-72; PMID:16864783; http://dx.doi.org/ 10.1073/pnas.0603363103 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Tchakarska G, Roussel M, Troussard X, Sola B. Cyclin D1 inhibits mitochondrial activity in B cells. Cancer Res 2011; 71:1690-9; PMID:21343394; http://dx.doi.org/ 10.1158/0008-5472.CAN-10-2564 [DOI] [PubMed] [Google Scholar]
- 41.Bienvenu F, Jirawatnotai S, Elias JE, Meyer CA, Mizeracka K, Marson A, Frampton GM, Cole MF, Odom DT, Odajima J, et al.. Transcriptional role of cyclin D1 in development revealed by a genetic-proteomic screen. Nature 2010; 463:374-8; PMID:20090754; http://dx.doi.org/ 10.1038/nature08684 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Buchakjian MR, Kornbluth S. The engine driving the ship: metabolic steering of cell proliferation and death. Nat Rev Mol Cell Biol 2010. October;11(10):715-27; http://dx.doi.org/ 10.1038/nrm2972 [DOI] [PubMed] [Google Scholar]
- 43.Yang K, Hitomi M, Stacey DW. Variations in cyclin D1 levels through the cell cycle determine the proliferative fate of a cell. Cell Division 2006; 1:32; PMID:17176475; http://dx.doi.org/ 10.1186/1747-1028-1-32 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Dimova DK, Dyson NJ. The E2F transcriptional network: old acquaintances with new faces. Oncogene 2005; 24:2810-26; PMID:15838517; http://dx.doi.org/ 10.1038/sj.onc.1208612 [DOI] [PubMed] [Google Scholar]
- 45.Emerging roles of E2Fs in cancer: an exit from cell cycle control. Nat Rev Cancer 2009; 9:785-97; PMID:19851314; http://dx.doi.org/ 10.1038/nrc2696 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Fajas L, Landsberg RL, Huss-Garcia Y, Sardet C, Lees JA, Auwerx J. E2Fs Regulate Adipocyte Differentiation. Dev Cell 2002; 3:39-49; PMID:12110166; http://dx.doi.org/ 10.1016/S1534-5807(02)00190-9 [DOI] [PubMed] [Google Scholar]
- 47.Abella A, Dubus P, Malumbres M, Rane SG, Kiyokawa H, Sicard A, Vignon F, Langin D, Barbacid M, Fajas L. Cdk4 promotes adipogenesis through PPARgamma activation. Cell Metab 2005; 2:239-49; PMID:16213226; http://dx.doi.org/ 10.1016/j.cmet.2005.09.003 [DOI] [PubMed] [Google Scholar]
- 48.Fajas L, Egler V, Reiter R, Hansen J, Kristiansen K, Debril M, Miard S, Auwerx J. The Retinoblastoma-Histone Deacetylase 3 Complex Inhibits PPARγ and Adipocyte Differentiation. Dev Cell 2002; 3:903-10; PMID:12479814; http://dx.doi.org/ 10.1016/S1534-5807(02)00360-X [DOI] [PubMed] [Google Scholar]
- 49.Chen PL, Riley DJ, Chen Y, Lee WH. Retinoblastoma protein positively regulates terminal adipocyte differentiation through direct interaction with C/EBPs. Genes Dev 1996; 10:2794-804; PMID:8946919; http://dx.doi.org/ 10.1101/gad.10.21.2794 [DOI] [PubMed] [Google Scholar]
- 50.Fajas L, Annicotte J-S, Miard S, Sarruf D, Watanabe M, Auwerx J. Impaired pancreatic growth, β cell mass, and β cell function in E2F1 (−/− )mice. J Clin Invest 2004; 113:1288-95; PMID:15124020; http://dx.doi.org/ 10.1172/JCI18555 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Annicotte J-S, Blanchet E, Chavey C, Iankova I, Costes S, Assou S, Teyssier J, Dalle S, Sardet C, Fajas L. The CDK4-pRB-E2F1 pathway controls insulin secretion. Nat Cell Biol 2009; 11:1017-23; PMID:19597485; http://dx.doi.org/ 10.1038/ncb1915 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Roche TE, Hiromasa Y. Pyruvate dehydrogenase kinase regulatory mechanisms and inhibition in treating diabetes, heart ischemia, and cancer. Cell Mol Life Sci 2007; 64:830-49; PMID:17310282; http://dx.doi.org/ 10.1007/s00018-007-6380-z [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Hsieh MCF, Das D, Sambandam N, Zhang MQ, Nahlé Z. Regulation of the PDK4 isozyme by the Rb-E2F1 complex. J Biol Chem 2008; 283:27410-7; PMID:18667418; http://dx.doi.org/ 10.1074/jbc.M802418200 [DOI] [PubMed] [Google Scholar]
- 54.Darville MI, Antoine IV, Mertens-Strijthagen JR, Dupriez VJ, Rousseau GG. An E2F-dependent late-serum-response promoter in a gene that controls glycolysis. Oncogene 1995; 11:1509-17; PMID:7478575 [PubMed] [Google Scholar]
- 55.Goto Y, Hayashi R, Kang D, Yoshida K. Acute loss of transcription factor E2F1 induces mitochondrial biogenesis in HeLa cells. J Cell Physiol 2006; 209:923-34; PMID:16972274; http://dx.doi.org/ 10.1002/jcp.20802 [DOI] [PubMed] [Google Scholar]
- 56.Blanchet E, Annicotte J-S, Lagarrigue S, Aguilar V, Clapé C, Chavey C, Fritz V, Casas F, Apparailly F, Auwerx J, et al.. E2F transcription factor-1 regulates oxidative metabolism. Nat Cell Biol 2011; 13:1146-52; PMID:21841792; http://dx.doi.org/ 10.1038/ncb2309 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Naaz A, Holsberger DR, Iwamoto GA, Nelson A, Kiyokawa H, Cooke PS. Loss of cyclin-dependent kinase inhibitors produces adipocyte hyperplasia and obesity. FASEB J 2004; 18:1925-7; PMID:15469964 [DOI] [PubMed] [Google Scholar]
- 58.Inoue N, Yahagi N, Yamamoto T, Ishikawa M, Watanabe K, Matsuzaka T, Nakagawa Y, Takeuchi Y, Kobayashi K, Takahashi A, et al.. Cyclin-dependent kinase inhibitor, p21WAF1/CIP1, is involved in adipocyte differentiation and hypertrophy, linking to obesity, and insulin resistance. J Biol Chem 2008; 283:21220-9; PMID:18445590; http://dx.doi.org/ 10.1074/jbc.M801824200 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Lin J, Della-Fera MA, Li C, Page K, Choi YH, Hartzell DL, Baile CA. P27 knockout mice: reduced myostatin in muscle and altered adipogenesis. Biochem Biophys Res Commun 2003; 300:938-42; PMID:12559964; http://dx.doi.org/ 10.1016/S0006-291X(02)02949-2 [DOI] [PubMed] [Google Scholar]
- 60.Uchida T, Nakamura T, Hashimoto N, Matsuda T, Kotani K, Sakaue H, Kido Y, Hayashi Y, Nakayama KI, White MF, et al.. Deletion of Cdkn1b ameliorates hyperglycemia by maintaining compensatory hyperinsulinemia in diabetic mice. Nat Med 2005; 11:175-82; PMID:15685168; http://dx.doi.org/ 10.1038/nm1187 [DOI] [PubMed] [Google Scholar]
- 61.Pagano M. Cell cycle regulation by the ubiquitin pathway. FASEB J 1997; 11:1067-75; PMID:9367342 [DOI] [PubMed] [Google Scholar]
- 62.Skaar JR, Pagano M. Cdh1: a master G0/G1 regulator. Nat Cell Biol 2008; 10:755-7; PMID:18591966; http://dx.doi.org/ 10.1038/ncb0708-755 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Matyskiela ME, Rodrigo-Brenni MC, Morgan DO. Mechanisms of ubiquitin transfer by the anaphase-promoting complex. J Biol 2009; 8:92; PMID:19874575; http://dx.doi.org/ 10.1186/jbiol184 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Skowyra D, Craig KL, Tyers M, Elledge SJ, Harper JW. F-box proteins are receptors that recruit phosphorylated substrates to the SCF ubiquitin-ligase complex. Cell 1997; 91:209-19; PMID:9346238; http://dx.doi.org/ 10.1016/S0092-8674(00)80403-1 [DOI] [PubMed] [Google Scholar]
- 65.Skaar JR, Pagan JK, Pagano M. Mechanisms and function of substrate recruitment by F-box proteins. Nat Rev Mol Cell Biol 2013; 14:369-81; PMID:23657496; http://dx.doi.org/ 10.1038/nrm3582 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Vodermaier HC. APC/C and SCF: controlling each other and the cell cycle. Curr Biol 2004; 14:R787-96; PMID:15380093; http://dx.doi.org/ 10.1016/j.cub.2004.09.020 [DOI] [PubMed] [Google Scholar]
- 67.Purnapatre K, Gray M, Piccirillo S, Honigberg SM. Glucose inhibits meiotic DNA replication through SCFGrr1p-dependent destruction of Ime2p kinase. Mol Cell Biol 2005; 25:440-50; PMID:15601864; http://dx.doi.org/ 10.1128/MCB.25.1.440-450.2005 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Carlson M. Glucose repression in yeast. Curr Opin Microbiol 1999; 2:202-7; PMID:10322167; http://dx.doi.org/ 10.1016/S1369-5274(99)80035-6 [DOI] [PubMed] [Google Scholar]
- 69.Ozcan S, Johnston M. Three different regulatory mechanisms enable yeast hexose transporter (HXT) genes to be induced by different levels of glucose. Mol Cell Biol 1995; 15:1564-72; PMID:7862149 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Spielewoy N, Flick K, Kalashnikova TI, Walker JR, Wittenberg C. Regulation and recognition of SCFGrr1 targets in the glucose and amino acid signaling pathways. Mol Cell Biol 2004; 24:8994-9005; PMID:15456873; http://dx.doi.org/ 10.1128/MCB.24.20.8994-9005.2004 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Kim J-H, Brachet V, Moriya H, Johnston M. Integration of transcriptional and posttranslational regulation in a glucose signal transduction pathway in Saccharomyces cerevisiae. Eukaryotic Cell 2006; 5:167-73; PMID:16400179; http://dx.doi.org/ 10.1128/EC.5.1.167-173.2006 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Tudzarova S, Colombo SL, Stoeber K, Carcamo S, Williams GH, Moncada S. Two ubiquitin ligases, APC/C-Cdh1 and SKP1-CUL1-F (SCF)-β-TrCP, sequentially regulate glycolysis during the cell cycle. PNAS 2011; 108:5278-83; PMID:21402913; http://dx.doi.org/ 10.1073/pnas.1102247108 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Almeida A, Bolaños JP, Moncada S. E3 ubiquitin ligase APC/C-Cdh1 accounts for the Warburg effect by linking glycolysis to cell proliferation. PNAS 2010; 107:738-41; PMID:20080744; http://dx.doi.org/ 10.1073/pnas.0913668107 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Colombo SL, Palacios-Callender M, Frakich N, De Leon J, Schmitt CA, Boorn L, Davis N, Moncada S. Anaphase-promoting complex/cyclosome-Cdh1 coordinates glycolysis and glutaminolysis with transition to S phase in human T lymphocytes. PNAS 2010; 107:18868-73; PMID:20921411; http://dx.doi.org/ 10.1073/pnas.1012362107 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Colombo SL, Palacios-Callender M, Frakich N, Carcamo S, Kovacs I, Tudzarova S, Moncada S. Molecular basis for the differential use of glucose and glutamine in cell proliferation as revealed by synchronized HeLa cells. PNAS 2011; 108:21069-74; PMID:22106309; http://dx.doi.org/ 10.1073/pnas.1117500108 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Sun J, Karoulia Z, Wong EYM, Ahmed M, Itoh K, Xu P-X. The phosphatase-transcription activator EYA1 is targeted by anaphase-promoting complex/Cdh1 for degradation at M-to-G1 transition. Mol Cell Biol 2013; 33:927-36; PMID:23263983; http://dx.doi.org/ 10.1128/MCB.01516-12 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Xu PX, Adams J, Peters H, Brown MC, Heaney S, Maas R. Eya1-deficient mice lack ears and kidneys and show abnormal apoptosis of organ primordia. Nat Genet 1999; 23:113-7; PMID:10471511; http://dx.doi.org/ 10.1038/12722 [DOI] [PubMed] [Google Scholar]
- 78.Zou D, Silvius D, Fritzsch B, Xu P-X. Eya1 and Six1 are essential for early steps of sensory neurogenesis in mammalian cranial placodes. Development 2004; 131:5561-72; PMID:15496442; http://dx.doi.org/ 10.1242/dev.01437 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Zou D, Silvius D, Rodrigo-Blomqvist S, Enerbäck S, Xu P-X. Eya1 regulates the growth of otic epithelium and interacts with Pax2 during the development of all sensory areas in the inner ear. Dev Biol 2006; 298:430-41; PMID:16916509; http://dx.doi.org/ 10.1016/j.ydbio.2006.06.049 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Zhang L, Yang N, Huang J, Buckanovich RJ, Liang S, Barchetti A, Vezzani C, O'Brien-Jenkins A, Wang J, Ward MR, et al.. Transcriptional coactivator Drosophila eyes absent homologue 2 is up-regulated in epithelial ovarian cancer and promotes tumor growth. Cancer Res 2005; 65:925-32; PMID:15705892 [PubMed] [Google Scholar]
- 81.Pandey RN, Rani R, Yeo E-J, Spencer M, Hu S, Lang RA, Hegde RS. The Eyes Absent phosphatase-transactivator proteins promote proliferation, transformation, migration, and invasion of tumor cells. Oncogene 2010; 29:3715-22; PMID:20418914; http://dx.doi.org/ 10.1038/onc.2010.122 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Li C-M, Guo M, Borczuk A, Powell CA, Wei M, Thaker HM, Friedman R, Klein U, Tycko B. Gene expression in Wilms' tumor mimics the earliest committed stage in the metanephric mesenchymal-epithelial transition. Am J Pathol. 2002; 160:2181-90; PMID:12057921; http://dx.doi.org/ 10.1016/S0002-9440(10)61166-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Miller SJ, Lan ZD, Hardiman A, Wu J, Kordich JJ, Patmore DM, Hegde RS, Cripe TP, Cancelas JA, Collins MH, et al.. Inhibition of Eyes Absent Homolog 4 expression induces malignant peripheral nerve sheath tumor necrosis. Oncogene 2010; 29:368-79; PMID:19901965; http://dx.doi.org/ 10.1038/onc.2009.360 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Grifone R, Laclef C, Spitz F, Lopez S, Demignon J, Guidotti J-E, Kawakami K, Xu P-X, Kelly R, Petrof BJ, et al.. Six1 and Eya1 expression can reprogram adult muscle from the slow-twitch phenotype into the fast-twitch phenotype. Mol Cell Biol 2004; 24:6253-67; PMID:15226428; http://dx.doi.org/ 10.1128/MCB.24.14.6253-6267.2004 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Cassago A, Ferreira APS, Ferreira IM, Fornezari C, Gomes ERM, Greene KS, Pereira HM, Garratt RC, Dias SMG, Ambrosio ALB. Mitochondrial localization and structure-based phosphate activation mechanism of Glutaminase C with implications for cancer metabolism. PNAS 2012; 109:1092-7; PMID:22228304; http://dx.doi.org/ 10.1073/pnas.1112495109 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Vousden KH, Prives C. Blinded by the Light: The Growing Complexity of p53. Cell 2009; 137:413-31; PMID:19410540; http://dx.doi.org/ 10.1016/j.cell.2009.04.037 [DOI] [PubMed] [Google Scholar]
- 87.Schwartzenberg-Bar-Yoseph F, Armoni M, Karnieli E. The tumor suppressor p53 down-regulates glucose transporters GLUT1 and GLUT4 gene expression. Cancer Res 2004; 64:2627-33; PMID:15059920; http://dx.doi.org/ 10.1158/0008-5472.CAN-03-0846 [DOI] [PubMed] [Google Scholar]
- 88.Kondoh H, Lleonart ME, Gil J, Wang J, Degan P, Peters G, Martinez D, Carnero A, Beach D. Glycolytic enzymes can modulate cellular life span. Cancer Res 2005; 65:177-85; PMID:15665293 [PubMed] [Google Scholar]
- 89.Bensaad K, Tsuruta A, Selak MA, Vidal MNC, Nakano K, Bartrons R, Gottlieb E, Vousden KH. TIGAR, a p53-inducible regulator of glycolysis and apoptosis. Cell 2006; 126:107-20; PMID:16839880; http://dx.doi.org/ 10.1016/j.cell.2006.05.036 [DOI] [PubMed] [Google Scholar]
- 90.Boidot R, Vegran F, Meulle A, Le Breton A, Dessy C, Sonveaux P, Lizard-Nacol S, Feron O. Regulation of monocarboxylate transporter MCT1 expression by p53 mediates inward and outward lactate fluxes in tumors. Cancer Res 2012; 72:939-48; PMID:22184616; http://dx.doi.org/ 10.1158/0008-5472.CAN-11-2474 [DOI] [PubMed] [Google Scholar]
- 91.Jiang P, Du W, Wang X, Mancuso A, Gao X, Wu M, Yang X. p53 regulates biosynthesis through direct inactivation of glucose-6-phosphate dehydrogenase. Nat Cell Biol 2011; 13:310-6; PMID:21336310; http://dx.doi.org/ 10.1038/ncb2172 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Aird KM, Zhang G, Li H, Tu Z, Bitler BG, Garipov A, Wu H, Wei Z, Wagner SN, Herlyn M, et al.. Suppression of nucleotide metabolism underlies the establishment and maintenance of oncogene-induced senescence. CellReports 2013; 3:1252-65 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Kulawiec M, Ayyasamy V, Singh KK. p53 regulates mtDNA copy number and mitocheckpoint pathway. J Carcinog 2009; 8:8; PMID:19439913; http://dx.doi.org/ 10.4103/1477-3163.50893 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Matoba S, Kang J-G, Patino WD, Wragg A, Boehm M, Gavrilova O, Hurley PJ, Bunz F, Hwang PM. p53 regulates mitochondrial respiration. Science 2006; 312:1650-3; PMID:16728594; http://dx.doi.org/ 10.1126/science.1126863 [DOI] [PubMed] [Google Scholar]
- 95.Okamura S, Ng CC, Koyama K, Takei Y, Arakawa H, Monden M, Nakamura Y. Identification of seven genes regulated by wild-type p53 in a colon cancer cell line carrying a well-controlled wild-type p53 expression system. Oncol Res 1999; 11:281-5; PMID:10691030 [PubMed] [Google Scholar]
- 96.Contractor T, Harris CR. p53 negatively regulates transcription of the pyruvate dehydrogenase kinase Pdk2. Cancer Res 2012; 72:560-7; PMID:22123926; http://dx.doi.org/ 10.1158/0008-5472.CAN-11-1215 [DOI] [PubMed] [Google Scholar]
- 97.Jiang P, Du W, Mancuso A, Wellen KE, Yang X. Reciprocal regulation of p53 and malic enzymes modulates metabolism and senescence. Nature 2013; 493:689-93; PMID:23334421; http://dx.doi.org/ 10.1038/nature11776 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Hu W, Zhang C, Wu R, Sun Y, Levine A, Feng Z. Glutaminase 2, a novel p53 target gene regulating energy metabolism and antioxidant function. PNAS 2010; 107:7455-60; PMID:20378837; http://dx.doi.org/ 10.1073/pnas.1001006107 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Maddocks ODK, Berkers CR, Mason SM, Zheng L, Blyth K, Gottlieb E, Vousden KH. Serine starvation induces stress and p53-dependent metabolic remodelling in cancer cells. Nature 2013; 493:542-6; PMID:23242140; http://dx.doi.org/ 10.1038/nature11743 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Yahagi N, Shimano H, Matsuzaka T, Najima Y, Sekiya M, Nakagawa Y, Ide T, Tomita S, Okazaki H, Tamura Y, et al.. p53 Activation in adipocytes of obese mice. J Biol Chem 2003; 278:25395-400; PMID:12734185; http://dx.doi.org/ 10.1074/jbc.M302364200 [DOI] [PubMed] [Google Scholar]
- 101.Faubert B, Berger SL, Jones RG, Thompson CB, Mak TW. Carnitine palmitoyltransferase 1C promotes cell survival and tumor growth under conditions of metabolic stress. Genes Dev 2011; 25:1041-51; PMID:21576264; http://dx.doi.org/ 10.1101/gad.1987211 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Ide T, Brown-Endres L, Chu K, Ongusaha PP, Ohtsuka T, El-Deiry WS, Aaronson SA, Lee SW. GAMT, a p53-inducible modulator of apoptosis, is critical for the adaptive response to nutrient stress. Mol Cell 2009; 36:379-92; PMID:19917247; http://dx.doi.org/ 10.1016/j.molcel.2009.09.031 [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- 103.Velletri T, Romeo F, Tucci P, Peschiaroli A, Annicchiarico-Petruzzelli M, Niklison-Chirou MV, Amelio I, Knight RA, Mak TW, Melino G, et al.. GLS2 is transcriptionally regulated by p73 and contributes to neuronal differentiation. cc 2013; 12:3564-73; http://dx.doi.org/ 10.4161/cc.26771 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Giacobbe A, Bongiorno-Borbone L, Bernassola F, Terrinoni A, Markert EK, Levine AJ, Feng Z, Agostini M, Zolla L, Agr∫ AF, et al.. p63 regulates glutaminase 2 expression. cc 2013; 12:1395-405; http://dx.doi.org/ 10.4161/cc.24478 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Rufini A, Niklison-Chirou MV, Inoue S, Tomasini R, Harris IS, Marino A, Federici M, Dinsdale D, Knight RA, Melino G, et al.. TAp73 depletion accelerates aging through metabolic dysregulation. Genes Dev 2012; 26:2009-14; PMID:22987635; http://dx.doi.org/ 10.1101/gad.197640.112 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Du W, Jiang P, Mancuso A, Stonestrom A, Brewer MD, Minn AJ, Mak TW, Wu M, Yang X. TAp73 enhances the pentose phosphate pathway and supports cell proliferation. Nat Cell Biol 2013; 15:991-1000; PMID:23811687; http://dx.doi.org/ 10.1038/ncb2789 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Amelio I, Markert EK, Rufini A, Antonov AV, Sayan BS, Tucci P, Agostini M, Mineo TC, Levine AJ, Melino G. p73 regulates serine biosynthesis in cancer. Oncogene 2014; 33(42):5039-46; PMID:24186203 [DOI] [PubMed] [Google Scholar]
- 108.Su X, Gi YJ, Chakravarti D, Chan IL, Zhang A, Xia X, Tsai KY, Flores ER. TAp63 is a master transcriptional regulator of lipid and glucose metabolism. Cell Metab 2012; 16:511-25; PMID:23040072; http://dx.doi.org/ 10.1016/j.cmet.2012.09.006 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Sabbisetti V, Di Napoli A, Seeley A, Amato AM, O'Regan E, Ghebremichael M, Loda M, Signoretti S. p63 promotes cell survival through fatty acid synthase. PLoS ONE 2009; 4:e5877; PMID:19517019; http://dx.doi.org/ 10.1371/journal.pone.0005877 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Pardee AB. A restriction point for control of normal animal cell proliferation. Proc Natl Acad Sci USA 1974; 71:1286-90; PMID:4524638; http://dx.doi.org/ 10.1073/pnas.71.4.1286 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Yalcin A, Clem BF, Simmons A, Lane A, Nelson K, Clem AL, Brock E, Siow D, Wattenberg B, Telang S, et al.. Nuclear targeting of 6-phosphofructo-2-kinase (PFKFB3) increases proliferation via cyclin-dependent kinases. J Biol Chem 2009; 284:24223-32; PMID:19473963; http://dx.doi.org/ 10.1074/jbc.M109.016816 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Christofk HR, Vander Heiden MG, Harris MH, Ramanathan A, Gerszten RE, Wei R, Fleming MD, Schreiber SL, Cantley LC. The M2 splice isoform of pyruvate kinase is important for cancer metabolism and tumour growth. Nature 2008; 452:230-3; PMID:18337823; http://dx.doi.org/ 10.1038/nature06734 [DOI] [PubMed] [Google Scholar]
- 113.Yang W, Xia Y, Ji H, Zheng Y, Liang J, Huang W, Gao X, Aldape K, Lu Z. Nuclear PKM2 regulates β-catenin transactivation upon EGFR activation. Nature 2011; 480:118-22; PMID:22056988; http://dx.doi.org/ 10.1038/nature10598 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Yang W, Zheng Y, Xia Y, Ji H, Chen X, Guo F, Lyssiotis CA, Aldape K, Cantley LC, Lu Z. ERK1/2-dependent phosphorylation and nuclear translocation of PKM2 promotes the Warburg effect. Nat Cell Biol 2012; 14:1295-304; PMID:23178880; http://dx.doi.org/ 10.1038/ncb2629 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Yang W, Xia Y, Hawke D, Li X, Liang J, Xing D, Aldape K, Hunter T, Alfred Yung WK, Lu Z. PKM2 phosphorylates histone H3 and promotes gene transcription and tumorigenesis. Cell 2012; 150:685-96; PMID:22901803; http://dx.doi.org/ 10.1016/j.cell.2012.07.018 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Jiang Y, Li X, Yang W, Hawke DH, Zheng Y, Xia Y, Aldape K, Wei C, Guo F, Chen Y, et al.. PKM2 regulates chromosome segregation and mitosis progression of tumor cells. Mol Cell 2014; 53:75-87; PMID:24316223; http://dx.doi.org/ 10.1016/j.molcel.2013.11.001 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Wellen KE, Hatzivassiliou G, Sachdeva UM, Bui TV, Cross JR, Thompson CB. ATP-citrate lyase links cellular metabolism to histone acetylation. Science 2009; 324:1076-80; PMID:19461003; http://dx.doi.org/ 10.1126/science.1164097 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Galdieri L, Vancura A. Acetyl-CoA carboxylase regulates global histone acetylation. J Biol Chem 2012; 287:23865-76; PMID:22580297; http://dx.doi.org/ 10.1074/jbc.M112.380519 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Li B, Carey M, Workman JL. The role of chromatin during transcription. Cell 2007; 128:707-19; PMID:17320508; http://dx.doi.org/ 10.1016/j.cell.2007.01.015 [DOI] [PubMed] [Google Scholar]
- 120.Berger SL. The complex language of chromatin regulation during transcription. Nature 2007; 447:407-12; PMID:17522673; http://dx.doi.org/ 10.1038/nature05915 [DOI] [PubMed] [Google Scholar]
- 121.Love DC, Hanover JA. The hexosamine signaling pathway: deciphering the “O-GlcNAc code.” Sci STKE 2005; 2005:re13; PMID:16317114 [DOI] [PubMed] [Google Scholar]
- 122.Yang WH, Kim JE, Nam HW, Ju JW, Kim HS, Kim YS, Cho JW. Modification of p53 with O-linked N-acetylglucosamine regulates p53 activity and stability. Nat Cell Biol 2006; 8:1074-83; PMID:16964247; http://dx.doi.org/ 10.1038/ncb1470 [DOI] [PubMed] [Google Scholar]
- 123.Drougat L, Olivier-Van Stichelen S, Mortuaire M, Foulquier F, Lacoste A-S, Michalski J-C, Lefebvre T, Vercoutter-Edouart A-S. Characterization of O-GlcNAc cycling and proteomic identification of differentially O-GlcNAcylated proteins during G1/S transition. Biochim Biophys Acta 2012; 1820:1839-48; PMID:22967762; http://dx.doi.org/ 10.1016/j.bbagen.2012.08.024 [DOI] [PubMed] [Google Scholar]
- 124.Olivier-Van Stichelen S, Drougat L, Dehennaut V, Yazidi-Belkoura El I, Guinez C, Mir A-M, Michalski J-C, Vercoutter-Edouart A-S, Lefebvre T. Serum-stimulated cell cycle entry promotes ncOGT synthesis required for cyclin D expression. Oncogenesis 2012; 1:e36; PMID:23552487; http://dx.doi.org/ 10.1038/oncsis.2012.36 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Zhang S, Roche K, Nasheuer H-P, Lowndes NF. Modification of histones by sugar β-N-acetylglucosamine (GlcNAc) occurs on multiple residues, including histone H3 serine 10, and is cell cycle-regulated. J Biol Chem 2011; 286:37483-95; PMID:21896475; http://dx.doi.org/ 10.1074/jbc.M111.284885 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Hanover JA, Krause MW, Love DC. Bittersweet memories: linking metabolism to epigenetics through O-GlcNAcylation. Nat Rev Mol Cell Biol 2012; 13:312-21; PMID:22522719; http://dx.doi.org/ 10.1038/nrm3334 [DOI] [PubMed] [Google Scholar]
- 127.Yang YR, Song M, Lee H, Jeon Y, Choi E-J, Jang H-J, Moon HY, Byun H-Y, Kim E-K, Kim DH, et al.. O-GlcNAcase is essential for embryonic development and maintenance of genomic stability. Aging Cell 2012; 11:439-48; PMID:22314054; http://dx.doi.org/ 10.1111/j.1474-9726.2012.00801.x [DOI] [PubMed] [Google Scholar]
- 128.Dehennaut V, Slomianny M-C, Page A, Vercoutter-Edouart A-S, Jessus C, Michalski J-C, Vilain J-P, Bodart J-F, Lefebvre T. Identification of structural and functional O-linked N-acetylglucosamine-bearing proteins in Xenopus laevis oocyte. Mol Cell Proteomics 2008; 7:2229-45; PMID:18617508; http://dx.doi.org/ 10.1074/mcp.M700494-MCP200 [DOI] [PubMed] [Google Scholar]
- 129.Dehennaut V, Hanoulle X, Bodart J-F, Vilain J-P, Michalski J-C, Landrieu I, Lippens G, Lefebvre T. Microinjection of recombinant O-GlcNAc transferase potentiates Xenopus oocytes M-phase entry. Biochem Biophys Res Commun 2008; 369:539-46; PMID:18298951; http://dx.doi.org/ 10.1016/j.bbrc.2008.02.063 [DOI] [PubMed] [Google Scholar]
- 130.Dehennaut V, Lefebvre T, Leroy Y, Vilain J-P, Michalski J-C, Bodart J-F. Survey of O-GlcNAc level variations in Xenopus laevis from oogenesis to early development. Glycoconj J 2009; 26:301-11; PMID:18633701; http://dx.doi.org/ 10.1007/s10719-008-9166-0 [DOI] [PubMed] [Google Scholar]
- 131.Hardie DG. AMP-activated/SNF1 protein kinases: conserved guardians of cellular energy. Nat Rev Mol Cell Biol 2007; 8:774-85; PMID:17712357; http://dx.doi.org/ 10.1038/nrm2249 [DOI] [PubMed] [Google Scholar]
- 132.Hardie DG, Ross FA, Hawley SA. AMPK: a nutrient and energy sensor that maintains energy homeostasis. Nat Rev Mol Cell Biol 2012; 13:251-62; PMID:22436748; http://dx.doi.org/ 10.1038/nrm3311 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Imamura K, Ogura T, Kishimoto A, Kaminishi M, Esumi H. 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 2001; 287:562-7; PMID:11554766; http://dx.doi.org/ 10.1006/bbrc.2001.5627 [DOI] [PubMed] [Google Scholar]
- 134.Jones RG, Plas DR, Kubek S, Buzzai M, Mu J, Xu Y, Birnbaum MJ, Thompson CB. AMP-activated protein kinase induces a p53-dependent metabolic checkpoint. Mol Cell 2005; 18:283-93; PMID:15866171; http://dx.doi.org/ 10.1016/j.molcel.2005.03.027 [DOI] [PubMed] [Google Scholar]
- 135.Liang J, Shao SH, Xu Z-X, Hennessy B, Ding Z, Larrea M, Kondo S, Dumont DJ, Gutterman JU, Walker CL, et al.. The energy sensing LKB1-AMPK pathway regulates p27(kip1) phosphorylation mediating the decision to enter autophagy or apoptosis. Nat Cell Biol 2007; 9:218-24; PMID:17237771; http://dx.doi.org/ 10.1038/ncb1537 [DOI] [PubMed] [Google Scholar]
- 136.Poteet E, Choudhury GR, Winters A, Li W, Ryou M-G, Liu R, Tang L, Ghorpade A, Wen Y, Yuan F, et al.. Reversing the Warburg effect as a treatment for glioblastoma. J Biol Chem 2013; 288:9153-64; PMID:23408428; http://dx.doi.org/ 10.1074/jbc.M112.440354 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Mandal S, Freije WA, Guptan P, Banerjee U. Metabolic control of G1-S transition: cyclin E degradation by p53-induced activation of the ubiquitin-proteasome system. J Cell Biol 2010; 188:473-9; PMID:20176921; http://dx.doi.org/ 10.1083/jcb.200912024 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Owusu-Ansah E, Yavari A, Mandal S, Banerjee U. Distinct mitochondrial retrograde signals control the G1-S cell cycle checkpoint. Nat Genet 2008; 40:356-61; PMID:18246068; http://dx.doi.org/ 10.1038/ng.2007.50 [DOI] [PubMed] [Google Scholar]
- 139.Berg JM, Tymoczko JL, Stryer L. Biochemistry. 7th ed New York: WH Freeman & Company; 2012. [Google Scholar]
- 140.Imai S, Armstrong CM, Kaeberlein M, Guarente L. Transcriptional silencing and longevity protein Sir2 is an NAD-dependent histone deacetylase. Nature 2000; 403:795-800; PMID:10693811; http://dx.doi.org/ 10.1038/35001622 [DOI] [PubMed] [Google Scholar]
- 141.Dryden SC, Nahhas FA, Nowak JE, Goustin A-S, Tainsky MA. Role for human SIRT2 NAD-dependent deacetylase activity in control of mitotic exit in the cell cycle. Mol Cell Biol 2003; 23:3173-85; PMID:12697818; http://dx.doi.org/ 10.1128/MCB.23.9.3173-3185.2003 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142.Inoue T, Hiratsuka M, Osaki M, Yamada H, Kishimoto I, Yamaguchi S, Nakano S, Katoh M, Ito H, Oshimura M. SIRT2, a tubulin deacetylase, acts to block the entry to chromosome condensation in response to mitotic stress. Oncogene 2007; 26:945-57; PMID:16909107; http://dx.doi.org/ 10.1038/sj.onc.1209857 [DOI] [PubMed] [Google Scholar]
- 143.Vaziri H, Dessain SK, Ng Eaton E, Imai SI, Frye RA, Pandita TK, Guarente L, Weinberg RA. hSIR2(SIRT1) functions as an NAD-dependent p53 deacetylase. Cell 2001; 107:149-59; PMID:11672523; http://dx.doi.org/ 10.1016/S0092-8674(01)00527-X [DOI] [PubMed] [Google Scholar]
- 144.Luo J, Nikolaev AY, Imai S, Chen D, Su F, Shiloh A, Guarente L, Gu W. Negative control of p53 by Sir2alpha promotes cell survival under stress. Cell 2001; 107:137-48; PMID:11672522; http://dx.doi.org/ 10.1016/S0092-8674(01)00524-4 [DOI] [PubMed] [Google Scholar]
- 145.Brunet A, Sweeney LB, Sturgill JF, Chua KF, Greer PL, Lin Y, Tran H, Ross SE, Mostoslavsky R, Cohen HY, et al.. Stress-dependent regulation of FOXO transcription factors by the SIRT1 deacetylase. Science 2004; 303:2011-5; PMID:14976264; http://dx.doi.org/ 10.1126/science.1094637 [DOI] [PubMed] [Google Scholar]
- 146.Nakahata Y, Kaluzova M, Grimaldi B, Sahar S, Hirayama J, Chen D, Guarente LP, Sassone-Corsi P. The NAD+-dependent deacetylase SIRT1 modulates CLOCK-mediated chromatin remodeling and circadian control. Cell 2008; 134:329-40; PMID:18662547; http://dx.doi.org/ 10.1016/j.cell.2008.07.002 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147.Asher G, Gatfield D, Stratmann M, Reinke H, Dibner C, Kreppel F, Mostoslavsky R, Alt FW, Schibler U. SIRT1 regulates circadian clock gene expression through PER2 deacetylation. Cell 2008; 134:317-28; PMID:18662546; http://dx.doi.org/ 10.1016/j.cell.2008.06.050 [DOI] [PubMed] [Google Scholar]
- 148.Matsuo T, Yamaguchi S, Mitsui S, Emi A, Shimoda F, Okamura H. Control mechanism of the circadian clock for timing of cell division in vivo. Science 2003; 302:255-9; PMID:12934012; http://dx.doi.org/ 10.1126/science.1086271 [DOI] [PubMed] [Google Scholar]
- 149.Fu L, Pelicano H, Liu J, Huang P, Lee C. The circadian gene Period2 plays an important role in tumor suppression and DNA damage response in vivo. Cell 2002; 111:41-50; PMID:12372299; http://dx.doi.org/ 10.1016/S0092-8674(02)00961-3 [DOI] [PubMed] [Google Scholar]