Synthetic lethality is a potential strategy for cancer treatment by specifically promoting the death of cancer cells with particular defects such as the loss of the RB (RB1) tumor suppressor. We previously showed that inactivation of both RB and TSC2 induces synergistic apoptosis during the development of Drosophila melanogaster and in cancer cells. However, the in vivo mechanism of this synthetic-lethal interaction is not clear. Here, we show that synergistic cell death in tissues that have lost the RB and TSC orthologs rbf and dtsc1/gig, respectively, or overexpress Rheb and dE2F1, are correlated with synergistic defects in G1–S control, which causes cells to accumulate DNA damage. Coexpression of the G1–S inhibitor Dap, but not the G2–M inhibitor dWee1, decreases DNA damage and reduces cell death. In addition, we show that rbf and dtsc1 mutant cells are under energy stress, are sensitive to decreased energy levels and depend on the cellular energy stress-response pathway for survival. Decreasing mitochondrial ATP synthesis by inactivating cova or abrogating the energy-stress response by removing the metabolic regulator LKB1 both enhance the elimination of cells lacking either rbf or dtsc1. These observations, in conjunction with the finding that deregulation of TORC1 induces activation of JNK, indicate that multiple cellular stresses are induced and contribute to the synthetic-lethal interactions between RB and TSC1/TSC2 inactivation. The insights gained from this study suggest new approaches for targeting RB-deficient cancers.

Inactivation of the gene encoding retinoblastoma-associated protein (RB1, hereafter referred to as RB) by deletion, mutation, loss of expression or functional inactivation is considered a general feature of cancer (Sherr and McCormick, 2002). In addition to deregulating cell proliferation, loss of RB function increases apoptosis at least in part through de-repressing activator-type E2F proteins (Chen et al., 2009; Gordon and Du, 2011). Cells that lose tumor suppressors such as RB become dependent on supporting pathways for survival. This reliance on parallel signaling pathways can potentially be exploited by using a synthetic-lethal approach (Kaelin, 2005) – targeting the genes required for the survival of cells that lack RB function.

The Drosophila genetic model provides a unique opportunity to identify RB synthetic-lethal mutations in vivo, as this organism harbors well-conserved yet relatively simple RB–E2F gene families (van den Heuvel and Dyson, 2008). We conducted a forward screen to identify genes that are important for cell survival and differentiation in the absence of the RB ortholog Retinoblastoma family protein rbf (Steele et al., 2009; Sukhanova et al., 2011; Tanaka-Matakatsu et al., 2009) and identified the fly TSC2 ortholog gig (encoding the protein Gigas), which induces synergistic cell death upon rbf loss (Li et al., 2010). This synthetic-lethal interaction between RB and TSC2 is conserved between flies and human cancer cells, as we showed that inactivation of RB and TSC2 leads to the induction of excessive cellular stress, including ROS, which contributes to synergistic cell death in cancer cells in vitro and using a xenograft model (Danos et al., 2012; Li et al., 2010). However, the in vivo mechanisms that mediate the synthetic lethality of RB and TSC2 mutations are unclear.

The products of the tuberous sclerosis genes TSC2 and TSC1 function together to restrict cell growth by inhibiting activation of the rapamycin-sensitive complex TORC1 (Potter et al., 2001; Tapon et al., 2001). Mutation of either TSC2 or TSC1 causes tuberous sclerosis complex (TSC), an autosomal-dominant tumor syndrome (Orlova and Crino, 2010). Studies have shown that mutations in these genes induce endoplasmic reticulum (ER) stress, leading to activation of the unfolded protein response (UPR) and susceptibility to apoptosis (Ozcan et al., 2008). Additionally, deregulation of TORC1 causes glucose addiction (Inoki et al., 2003), as these cells are sensitive to reduced ATP levels and depend on energy stress signaling (Choo et al., 2010). Therefore deregulated TORC1 activity promotes cell growth but also sensitizes cells to nutrient deficiency and/or metabolic-stress-induced cell death.

Here, we show that aberrant S phase entry resulting from inactivation of both rbf and gig/dtsc1 causes increased DNA damage and cell death. Additionally, we show that loss of either rbf or gig/tsc1 induces energy stress and sensitizes cells to ATP depletion, leading to the dependence on signaling by the serine/threonine-protein kinase LKB1 for viability. These results provide new insights into the mechanisms that mediate synergistic cell death when RB and TSC2 are both inactivated and suggest new therapeutic approaches that potentially can be used to target RB-deficient tumors.

rbf and gig cooperate to regulate S phase during fly development

We previously showed that loss of gig causes synergistic apoptosis and ablation of rbf-mutant tissue in mosaic eyes and wings (Li et al., 2010). As mutation of either rbf or gig causes G1–S deregulation, we investigated the effect of inactivating both rbf and gig (hereafter indicated ‘rbf,gig’) on G1–S regulation and its impact on synergistic apoptosis. Cells in the morphogenetic furrow (MF) of developing eye discs arrest in G1. While mutation of neither rbf nor gig affects cell cycle arrest in the MF, ectopic S phase cells are observed in rbf,gig double-mutant clones (Fig. 1A–C′), indicating that rbf and gig cooperate to enforce G1 arrest. In addition, overexpressing dE2f1 (together with dDp) together with Rheb, encoding a GTPase that is regulated by gig and activates TORC1 directly (Saucedo et al., 2003; Zhang et al., 2003), leads to synergistic S phase in the MF and posterior (supplementary material Fig. S1). These results, in conjunction with the observations of ectopic S phase in rbf,tsc1 clones (Hsieh et al., 2010), indicate that deregulated E2F and Rheb or TORC1 signaling induce synergistic S phase.

Fig. 1.

gig restrains the proliferation of rbf mutants. Developing eye imaginal discs mosaic for mutations of the indicated genotypes, marked by the absence of GFP, were assayed for cells in S phase. Whereas neither rbf (A,A′) nor gig (B,B′) mutation is sufficient to deregulate S phase, rbf,gig double-mutants exhibit ectopic S phase in the morphogenetic furrow (MF). It should be noted that gig mutant clones show precocious S phase, probably a consequence of precocious differentiation (Bateman and McNeill, 2004). The ectopic S phase effect is suppressed by additional mutations to either de2f1 (D,D′) or s6k (E,E′) but not hid (F,F′). S phase cells are marked by incorporation of the nucleotide analogs EdU (A–C) or BrdU (D–F). White arrows point to clones in the MF. Scale bars: 0.1 mm.

Fig. 1.

gig restrains the proliferation of rbf mutants. Developing eye imaginal discs mosaic for mutations of the indicated genotypes, marked by the absence of GFP, were assayed for cells in S phase. Whereas neither rbf (A,A′) nor gig (B,B′) mutation is sufficient to deregulate S phase, rbf,gig double-mutants exhibit ectopic S phase in the morphogenetic furrow (MF). It should be noted that gig mutant clones show precocious S phase, probably a consequence of precocious differentiation (Bateman and McNeill, 2004). The ectopic S phase effect is suppressed by additional mutations to either de2f1 (D,D′) or s6k (E,E′) but not hid (F,F′). S phase cells are marked by incorporation of the nucleotide analogs EdU (A–C) or BrdU (D–F). White arrows point to clones in the MF. Scale bars: 0.1 mm.

We previously showed that removing the transcription activation function of de2f1 or inactivation of s6k suppressed the synergistic cell death effect of rbf,gig (Li et al., 2010). To investigate whether the effects of de2f1 or s6k mutation on the synergistic cell death of rbf,gig clones are correlated with their effects on increased S phase, we introduced either de2f1 or s6k mutation into the rbf,gig-mutant background and found that each suppresses ectopic S phase within the MF (Fig. 1D–E′). As caspase activation can induce compensatory proliferation in flies (Ryoo et al., 2004), we tested whether the proliferative phenotype is a consequence of apoptosis by removing the apoptotic gene hid (Tanaka-Matakatsu et al., 2009) from rbf,gig-mutant clones, which significantly reduces the level of apoptosis (Li et al., 2010). As shown in Fig. 1F, inhibition of cell death does not prevent ectopic S phase in the MF (Fig. 1F,F′). These results demonstrate that the transcription-activation function of dE2f1 (Royzman et al., 1999) and TORC1 signaling through S6k (Radimerski et al., 2002) are required for deregulated cell proliferation in the absence of both rbf and gig, which correlates with, but does not result from, apoptosis.

rbf,gig mutants show increased sub-G1 and decreased G2–M cells

To characterize further the relationship between deregulated proliferation and synergistic cell death in rbf,gig double-mutants, we used fluorescence-activated cell sorting (FACS) to analyze cells dissociated from mosaic wing discs and compared the cell cycle profiles of the rbf and gig mutants alone or in combination. This technique separates mutant from wild-type cells based on GFP expression, thereby providing internal controls for each genotype (Danos et al., 2012). Wild-type cells in late third-instar wing discs arrest in G2. As RBF–E2F regulates both the G1–S as well as G2–M transitions, rbf loss causes a reduction of the G2 population compared with controls (Fig. 2A,A′,2E; P<0.01 between rbf and GFP control cells), consistent with other reported results (Datar et al., 2000). By contrast, mutation of gig, which only promotes the G1–S transition, causes an accumulation of G2–M cells (Fig. 2B,B′,E; P<0.05 between gig and GFP control cells) similar to that reported previously (Gao and Pan, 2001). We also found a modest accumulation of sub-G1 cells among gig mutants that is reminiscent of cell death observed in this tissue (Li et al., 2010). In rbf,gig double-mutants, FACS analysis shows a larger population of sub-G1 cells, reflecting elevated cell death. Interestingly, we also found a corresponding reduction in the G2–M peak (Fig. 2C,C′,E; P<0.01 between rbf,gig and gig), suggesting that cell cycle progression is important for the apoptotic effect. To further test this idea, we determined whether inhibiting cell death affects the rbf,gig cell cycle profile by introducing a mutation in the apical caspase dronc (Steele et al., 2009). We found that rbf,gig,dronc triple-mutants exhibit a decreased sub-G1 population compared with that of rbf,gig double-mutants (Fig. 2D,D′), consistent with the observation that dronc partially suppresses the death of rbf,gig mutants (Li et al., 2010). Importantly, we found that suppressing cell death also partially restores the G2–M peak of the cell cycle profile, whereas mutation of dronc alone did not affect cell cycle phasing (Fig. 2C–E; P<0.05 between rbf,gig and rbf,gig,dronc). Taken together, these results indicate that cell death in the absence of both rbf and gig occurs at the expense of cells that have passed the G1–S checkpoint.

Fig. 2.

G2 cells are under-represented in apoptotic rbf,gig clones. Cells dissociated from developing wings mosaic for the indicated genotypes were assayed for cell cycle profiles by FACS sorting. Gray peaks represent the DNA content of mutant (GFP−) cells, and green peaks represent control (GFP+) cells from the same tissue preparations. rbf,gig double-mutants show a reduction in the G2 population and an increase in sub-G1 compared with controls (C,C′) that depends on the presence of dronc (D,D′). rbf (A,A′) and gig (B,B′) profiles reflect previously reported phenotypes. (E) A diagram of the average G2–M cell fraction from the indicated mutant clones normalized by the corresponding wild-type controls. The results of FACS from three independent experiments are shown. Statistically significant differences between different pairs are indicated. Error bars indicate standard deviations together with their associated P values.

Fig. 2.

G2 cells are under-represented in apoptotic rbf,gig clones. Cells dissociated from developing wings mosaic for the indicated genotypes were assayed for cell cycle profiles by FACS sorting. Gray peaks represent the DNA content of mutant (GFP−) cells, and green peaks represent control (GFP+) cells from the same tissue preparations. rbf,gig double-mutants show a reduction in the G2 population and an increase in sub-G1 compared with controls (C,C′) that depends on the presence of dronc (D,D′). rbf (A,A′) and gig (B,B′) profiles reflect previously reported phenotypes. (E) A diagram of the average G2–M cell fraction from the indicated mutant clones normalized by the corresponding wild-type controls. The results of FACS from three independent experiments are shown. Statistically significant differences between different pairs are indicated. Error bars indicate standard deviations together with their associated P values.

Precocious G1–S transition contributes to synergistic cell death of rbf,gig mutants

As blocking death alters the G2–M peak of rbf,gig mutants, we further tested whether defective cell cycle regulation contributes to the synergistic cell death phenotype. We thus overexpressed G1 or G2 cell cycle inhibitors specifically in the posterior of mosaic wing discs, thus allowing for the comparison of rbf,gig-mutant clones either with or without transgene overexpression in the same tissue. Expressing the p27 KIP1 ortholog Dacapo (Dap) causes an extended G1 phase, whereas expression of dWee1 prolongs G2 during development (Reis and Edgar, 2004). We found that Dap expression decreases Caspase-3 activation in rbf,gig-mutant clones compared with control clones in the anterior (Fig. 3A,A′,H). This phenotype is evident despite Dap expression on its own causing reduced tissue growth and increased basal levels of apoptosis. By contrast, expressing dWee1 does not significantly alter cell death in rbf,gig-mutant clones (Fig. 3B,B′,H). These results indicate that rbf,gig-mutant wing disc cells that have a reduced ability to enter S phase are protected from apoptosis, whereas inhibiting cell cycle progression through the G2–M checkpoint does not have an observable effect.

Fig. 3.

S phase progression is important for rbf,gig cell death. (A–B) rbf,gig mosaic clones induced in wing imaginal discs expressing either Dap (A,A′) or dWee1 (B,B′) under control of the engrailed promoter allow for the assessment of cell death upon inhibition of S phase or mitosis, respectively. Yellow arrows point to clones in the posterior, where the constructs are expressed, whereas white arrows indicate control clones in the anterior. Quantification of C3 levels within mutant clones is shown in panel H, where the asterisk (*) denotes statistical significance (P<0.01). (C–G) Overexpression of dE2f1,dDp using the flipout technique (marked by the presence of GFP) causes a mild cell death effect (C,C′) that is enhanced by Rheb (D,D′), whereas coexpression of Dap (F,F′) or Puc (G,G′) reduces this synergistic effect. Quantification of cell death in flipout clones is shown in panel I, where asterisks denote statistically significant differences (P<0.01) between dE2f1,dDp,Rheb and either dE2f1,dDp or Rheb alone (*) and dE2f1,dDp,Rheb compared with coexpression of Dap or Puc (**). Scale bars: 0.1 mm. Error bars indicate standard deviations.

Fig. 3.

S phase progression is important for rbf,gig cell death. (A–B) rbf,gig mosaic clones induced in wing imaginal discs expressing either Dap (A,A′) or dWee1 (B,B′) under control of the engrailed promoter allow for the assessment of cell death upon inhibition of S phase or mitosis, respectively. Yellow arrows point to clones in the posterior, where the constructs are expressed, whereas white arrows indicate control clones in the anterior. Quantification of C3 levels within mutant clones is shown in panel H, where the asterisk (*) denotes statistical significance (P<0.01). (C–G) Overexpression of dE2f1,dDp using the flipout technique (marked by the presence of GFP) causes a mild cell death effect (C,C′) that is enhanced by Rheb (D,D′), whereas coexpression of Dap (F,F′) or Puc (G,G′) reduces this synergistic effect. Quantification of cell death in flipout clones is shown in panel I, where asterisks denote statistically significant differences (P<0.01) between dE2f1,dDp,Rheb and either dE2f1,dDp or Rheb alone (*) and dE2f1,dDp,Rheb compared with coexpression of Dap or Puc (**). Scale bars: 0.1 mm. Error bars indicate standard deviations.

We further tested whether inhibiting G1–S progression affects cell death induced by dE2F1,dDp and Rheb overexpression. Clones of cells overexpressing dE2f1,dDp in eye discs show a pattern of apoptosis near the MF (Fig. 3C,C′) similar to rbf mutation (Moon et al., 2006; Tanaka-Matakatsu et al., 2009). Coexpression of Rheb with dE2f1,dDp significantly increases the level of cell death in these eye discs, particularly in clones anterior to the MF (Fig. 3D–E′,3I). Importantly, overexpression of Dap, while inhibiting ectopic G1–S progression (supplementary material Fig. S1), significantly decreases cell death (Fig. 3F,F′,I). Taken together, these results show that precocious S phase in rbf,gig-mutant clones probably contributes to the synergistic cell death phenotype.

Precocious S phase induced by deregulated E2F and TORC1 signaling increases DNA damage and cell death

Recent studies show that RB protects cells against chromosome instability and DNA damage (Coschi et al., 2010; Longworth et al., 2008; Manning et al., 2010; van Harn et al., 2010). To test whether rbf mutation during larval development induces DNA damage, we labeled mosaic eye discs with antibodies specific for γ-H2Av (Mehrotra and McKim, 2006a), a phosphorylated form of the Drosophila histone H2A variant that serves as a marker for DNA double-strand breaks (DSBs). Significant levels of γ-H2Av foci are observed in rbf-mutant clones (Fig. 4A,A′,I). While loss of gig also causes a low level of γ-H2Av accumulation proportional to mutant clone area (Fig. 4B,B,I), rbf,gig double-mutant clones show significantly increased γ-H2Av foci per unit clone area (Fig. 4C,C′,I). Therefore synergistic cell death in rbf,gig double-mutant clones correlates with DSB accumulation. Importantly, the increased DSBs in rbf,gig double-mutant clones are dependent on dE2f1 and TORC1 activities, as both de2f1 and s6k mutants suppress the γ-H2Av phenotype of rbf,gig mutants (Fig. 4D–E,I).

Fig. 4.

Loss of gig increases DNA damage in rbf-mutant clones. (A–E) Eye discs mosaic for mutations of the indicated genotypes (marked by the absence of GFP) were labeled with antibodies specific for γ-H2Av. Quantification of the number of γ-H2Av-positive cells per unit clone area is shown in panel I, where asterisks indicate statistically significant differences (P<0.01) from rbf,gig. (F–H) MARCM clones (marked by the presence of GFP) that lack rbf,dtsc1 while coexpressing either Dap (G,G′) or dWee1 (H,H′) were also assayed for γ-H2Av. Quantification of gamma-H2Av level in MARCM clones is shown in panel J, where the asterisk (*) shows a significant change (P<0.01) from rbf,dtsc1. Scale bars: 0.1 mm. Error bars indicate standard deviations.

Fig. 4.

Loss of gig increases DNA damage in rbf-mutant clones. (A–E) Eye discs mosaic for mutations of the indicated genotypes (marked by the absence of GFP) were labeled with antibodies specific for γ-H2Av. Quantification of the number of γ-H2Av-positive cells per unit clone area is shown in panel I, where asterisks indicate statistically significant differences (P<0.01) from rbf,gig. (F–H) MARCM clones (marked by the presence of GFP) that lack rbf,dtsc1 while coexpressing either Dap (G,G′) or dWee1 (H,H′) were also assayed for γ-H2Av. Quantification of gamma-H2Av level in MARCM clones is shown in panel J, where the asterisk (*) shows a significant change (P<0.01) from rbf,dtsc1. Scale bars: 0.1 mm. Error bars indicate standard deviations.

To determine whether precocious S phase induced by deregulated RB–E2F and Rheb–TORC1 is important for the accumulation of DNA DSBs, we overexpressed Dap in rbf,dtsc1 double-mutant clones using the mosaic analysis with a repressible cell marker (MARCM) approach (Lee and Luo, 2001). Similar to rbf,gig, inactivation of both rbf and tsc1 leads to synergistic cell death (Hsieh et al., 2010), which we also found to be correlated with high levels of DSBs (Fig. 4F,F′,J). Interestingly, partial inhibition of the G1–S transition by Dap reduces cell death (supplementary material Fig. S2) and decreases the level of γ-H2Av foci (Fig. 4G,G′,J) in rbf,dtsc1 clones. By contrast, expression of the G2–M regulator dWee1 does not reduce the levels of either cell death (supplementary material Fig. S2) or γ-H2Av foci (Fig. 4H,H′,J) in rbf,dtsc1 clones. To characterize further the relationship between increased DNA damage and induction of cell death in rbf,gig double-mutant clones, we determined the effect of impairing DSB repair by mutation of spnA, the Drosophila rad51 homolog required for DNA repair by the homologous-recombination pathway (Sekelsky et al., 2000). As shown in supplementary material Fig. S3, spnA mutation significantly increased both the number of DNA breaks (supplementary material Fig. S3A–C; P<0.001) as well as the level of cell death in rbf,dtsc1 mutant clones (supplementary material Fig. S3D–F; P<0.05).

Taken together, these results indicate that G1–S deregulation in rbf,gig clones induces DNA DSBs, which contribute to synergistic cell death. However as cell death in rbf,gig double-mutants is only partially suppressed by inhibiting S phase, it is possible that other forms of stress beyond DNA damage also affect the survival of these cells.

Cells lacking rbf and dtsc1 show decreased ATP levels

In addition to regulating cell growth and proliferation, tumor suppressors such as RB and TSC2 also regulate the energy-intensive metabolic processes of DNA replication and protein synthesis. Therefore, inactivation of rbf and gig/dtsc1 could cause metabolic and energetic imbalance. To test this, we induced mutant clones in a Minute background to generate eye-antenna discs that comprised mostly mutant tissue and determined the ratio of intracellular ATP:ADP within these discs. As a control, we determined the effect of mutant cytochrome c oxidase Va (cova), which encodes a mitochondrial protein that was shown to abrogate ATP synthesis and induce cell cycle arrest (Mandal et al., 2005; Owusu-Ansah et al., 2008). A modest but statistically significant decrease in the ATP:ADP ratio is observed in cova-mutant eye-antenna discs compared with controls (Fig. 5A; P = 0.003). A similar decrease in the ATP:ADP ratio is also observed in rbf- or tsc1-mutant discs compared with controls (Fig. 5A; P≤0.02); however, an even more pronounced decrease in the ATP:ADP ratio is observed in rbf,tsc1 double-mutant discs (P<0.03 compared with rbf- or tsc1-mutant discs). These results indicate that inactivation of rbf or tsc1 induces energy stress, which is more severe in rbf,tsc1 double-mutants. To characterize further the effect of deregulating RB–E2F and TORC1 signaling on cellular energy levels, we determined the effect of overexpressing Rheb and dE2F1–dDp. Decreased ATP:ADP ratios were observed in dE2f1,dDp,Rheb-overexpressing eye discs, similar to rbf,tsc1 double-mutant discs (supplementary material Fig. S4), suggesting an important role of RB–E2F and TSC–TORC1 signaling pathways in regulating energy metabolism, in addition to their roles in the cell cycle and cell growth.

Fig. 5.

rbf, dtsc1 and rbf,dtsc1 mutants are energy-deficient and exhibit lethality with loss of LKB1. (A) The ratio of ATP:ADP levels was calculated for each of the indicated genotypes using mosaic eye discs in a Minute background. Asterisks denote statistically significant differences between single mutants and the wild-type control (*, P≤0.02) or between double-mutants and all the single-mutants (**, P<0.03). (B–H) Mosaic clones of the indicated genotypes were induced in normal eye discs. Mutation of lkb1 causes little apoptosis on its own (B) but increases cell death in the absence of dtsc1 (D), rbf (G) and rbf,dtsc1 (H). (I–O) The loss of lkb1 decreases mutant tissue (white patches) from adult eyes mosaic for dtsc1 (K), rbf (M) and rbf,dtsc1 (O). Scale bars: 0.1 mm. Error bars indicate standard deviations.

Fig. 5.

rbf, dtsc1 and rbf,dtsc1 mutants are energy-deficient and exhibit lethality with loss of LKB1. (A) The ratio of ATP:ADP levels was calculated for each of the indicated genotypes using mosaic eye discs in a Minute background. Asterisks denote statistically significant differences between single mutants and the wild-type control (*, P≤0.02) or between double-mutants and all the single-mutants (**, P<0.03). (B–H) Mosaic clones of the indicated genotypes were induced in normal eye discs. Mutation of lkb1 causes little apoptosis on its own (B) but increases cell death in the absence of dtsc1 (D), rbf (G) and rbf,dtsc1 (H). (I–O) The loss of lkb1 decreases mutant tissue (white patches) from adult eyes mosaic for dtsc1 (K), rbf (M) and rbf,dtsc1 (O). Scale bars: 0.1 mm. Error bars indicate standard deviations.

rbf and tsc1 mutants undergo increased cell death in the absence of LKB1

Signaling by LKB1 is known to be required for balancing cellular energetic needs and supply through AMPK-dependent and -independent pathways (Shackelford and Shaw, 2009). The altered metabolism and increased cellular stress we observed in rbf- and tsc1-mutant clones suggests the likelihood that the viability of rbf- or dtsc1-mutant cells depends on LKB1 function. Thus, we used an lkb1 mutation (Lee et al., 2006) to test this possibility directly. Although mutation of lkb1 alone does not show a significant apoptotic effect (Fig. 5B), lkb1 loss significantly enhances cell death in rbf-mutant clones in both wing (supplementary material Fig. S5) and eye imaginal discs (Fig. 5E,G) and significantly decreases the contribution of mutant clones to mosaic eye tissue in adults (Fig. 5I,L,M). As loss of lkb1 does not significantly affect ATP levels either on its own or in combination with rbf mutation (Fig. 5A), it is likely that the cell death effect of lkb1 mutation is mediated by the inability of mutant cells to respond properly to energy stress. Similarly, inactivation of lkb1 also increases cell death in dtsc1- and rbf,dtsc1-mutant clones during larval development (Fig. 5C–H) and leads to the development of adult eyes that are largely devoid of mutant tissue (Fig. 5I–O).

rbf and dtsc1 mutants are sensitive to ATP depletion

rbf,tsc1 double-mutants show significantly decreased ATP levels compared with those of either single mutant (Fig. 5A), raising the possibility that excessive metabolic stress and ATP depletion contribute to the synergistic cell death. To test this, we impaired ATP production using the aforementioned cova mutant. rbf,cova double-mutant discs have significantly lower ATP levels than either single-mutant (Fig. 5A; P<0.001). Importantly, this decrease in ATP levels correlates with increased apoptosis in rbf,cova double-mutants, particularly anterior to the MF (Fig. 6G–I; Fig. 6M). Furthermore adult eyes are largely devoid of rbf,cova double-mutant tissue, even when induced in a Minute background, whereas both single mutants are visible (Fig. 6A–C). Similarly loss of cova significantly increases cell death in dtsc1 and rbf,dtsc1 mutant clones (Fig. 6J–M) and significantly decreases the amount of dtsc1-mutant adult tissue while eliminating the rbf,dtsc1-mutant tissue from adult eyes (Fig. 6B,D–F). These results indicate that loss of rbf or gig/dtsc1 alters metabolism and sensitizes cells to additional metabolic stress or energy depletion and that combining these mutations results in excessive metabolic stress that contributes to synergistic cell death, resulting in the elimination of mutant tissue from adult structures.

Fig. 6.

Cells lacking rbf, dtsc1 or rbf,dtsc1 are sensitive to compromised ATP synthesis. (A–F) Adult eyes mosaic for rbf (A), cova (B), rbf,cova (C), dtsc1 (D), dtsc1,cova (E) and rbf,dtsc1,cova (F) mutant clones (white patches) were generated in a Minute (M) background to maximize their growth. Loss of cova severely limits the growth of clones lacking rbf (C), dtsc1 (E) and rbf,dtsc1 (F). (G–L) Eye discs with GFP-negative mutant clones for rbf (G), cova (H), rbf,cova (I), dtsc1 (J), dtsc1,cova (K) and rbf,dtsc1,cova (L) mutant clones were induced in a Minute background and labeled with activated C3. Loss of cova visibly enhances cell death in the absence of rbf (I) dtsc1 (K) and rbf,dtsc1 (L) despite the absence of a significant cell death phenotype in the single cova mutant (H). Quantification of C3 levels within mutant clones is shown in panel M, where the asterisk (*) denotes statistical significance (P<0.001). Scale bars: 0.1 mm. Error bars indicate standard deviations.

Fig. 6.

Cells lacking rbf, dtsc1 or rbf,dtsc1 are sensitive to compromised ATP synthesis. (A–F) Adult eyes mosaic for rbf (A), cova (B), rbf,cova (C), dtsc1 (D), dtsc1,cova (E) and rbf,dtsc1,cova (F) mutant clones (white patches) were generated in a Minute (M) background to maximize their growth. Loss of cova severely limits the growth of clones lacking rbf (C), dtsc1 (E) and rbf,dtsc1 (F). (G–L) Eye discs with GFP-negative mutant clones for rbf (G), cova (H), rbf,cova (I), dtsc1 (J), dtsc1,cova (K) and rbf,dtsc1,cova (L) mutant clones were induced in a Minute background and labeled with activated C3. Loss of cova visibly enhances cell death in the absence of rbf (I) dtsc1 (K) and rbf,dtsc1 (L) despite the absence of a significant cell death phenotype in the single cova mutant (H). Quantification of C3 levels within mutant clones is shown in panel M, where the asterisk (*) denotes statistical significance (P<0.001). Scale bars: 0.1 mm. Error bars indicate standard deviations.

TORC1 deregulation activates JNK signaling, which contributes to increased cell death

We previously showed that antagonizing JNK signaling during larval development reduces apoptosis in rbf,gig wing disc clones (Li et al., 2010), indicating that activated JNK signaling also contributes to rbf,gig cell death. JNK activation can be monitored by expression of puckered (puc) (Martín-Blanco et al., 1998; Riesgo-Escovar et al., 1996) or directly using a phospho-specific antibody against JNK (Ohsawa et al., 2011). We observed elevated JNK signaling in eye discs mosaic for gig loss (Fig. 7B,B′; supplementary material Fig. S6F,F′) or overexpressing a strong Rheb construct (Fig. 7E,E′), whereas neither loss of rbf (Fig. 7A,A′) nor overexpression of dE2f1,dDp (Fig. 7D,D′) causes observable JNK activation. Furthermore, loss of rbf does not obviously affect JNK activation due to loss of gig (Fig. 7C,C′), nor does dE2f1,Dp coexpression affect JNK activity induced by Rheb (Fig. 7F,F′). Notably, mutation of s6k blocked elevated levels of phospho-JNK induced by gig mutation (supplementary material Fig. S6G,G′), indicating that deregulated TORC1 signaling through S6k is required for the activation of JNK signaling. While inhibiting JNK signaling by overexpressing Puc reduces cell death in rbf,gig double-mutants (Li et al., 2010), we also found that Puc reduces the apoptotic effect of dE2f1,Dp,Rheb (Fig. 3G,G′) as well as that induced by overexpression of a strong Rheb construct on its own (supplementary material Fig. S6A–E). Therefore, deregulated TORC1 activity activates JNK-mediated stress signaling, which also contributes to the rbf,gig cell death phenotype.

Fig. 7.

Stress signaling is induced by deregulation of TORC1. The activity of JNK was assayed in eye discs with mutant clones (A–C) marked by absence of GFP or flipout overexpression clones (D–F) marked by the presence of GFP, using the puc-lacZ reporter. Neither rbf loss (A,A′) nor dE2f1,dDp expression (D,D′) is sufficient to activate puc-lacZ over background. Deregulation of TORC1 by either loss of gig (B,B′) or overexpression of a strong Rheb construct (E,E′) activates puc-lacZ regardless of rbf (C,C′) or dE2f1,dDP coexpression (F,F′). White arrows point to wild-type tissue, whereas yellow arrows point to mutant or overexpression clones. Scale bars: 0.1 mm.

Fig. 7.

Stress signaling is induced by deregulation of TORC1. The activity of JNK was assayed in eye discs with mutant clones (A–C) marked by absence of GFP or flipout overexpression clones (D–F) marked by the presence of GFP, using the puc-lacZ reporter. Neither rbf loss (A,A′) nor dE2f1,dDp expression (D,D′) is sufficient to activate puc-lacZ over background. Deregulation of TORC1 by either loss of gig (B,B′) or overexpression of a strong Rheb construct (E,E′) activates puc-lacZ regardless of rbf (C,C′) or dE2f1,dDP coexpression (F,F′). White arrows point to wild-type tissue, whereas yellow arrows point to mutant or overexpression clones. Scale bars: 0.1 mm.

We have shown that cell death induced by simultaneous inactivation of rbf and gig/dtsc1 correlates with synergistic deregulation of G1–S control and increased energy stress. While it is not surprising that deregulated RB–E2F and TSC–TORC1 pathways leads to uncontrolled S phase progression, it is not expected that the observed G1–S deregulation contributes to cell death. We found that inactivation of rbf and gig/dtsc1 also leads to the increased accumulation of DSBs. Manipulations that suppress deregulated G1–S progression, by de2f1 and s6k mutations or Dap overexpression, reduce the accumulation of DSBs and suppress synergistic cell death of double-mutants. By contrast, inhibition of G2–M progression by expression of dWee1 failed to affect double-mutant DSB accumulation or synergistic cell death. These observations suggest that inactivation of rbf and gig/dtsc1 causes deregulated G1–S progression, leading to DNA DSB accumulation and contributing to synergistic cell death (Fig. 8).

Fig. 8.

Model for the regulation of cell death by RB–E2F and TSC1/TSC2. In addition to the repression of apoptotic signaling through Hid and JNK, the RB and TSC tumor suppressors regulate cell death by cooperating to prevent precocious S phase and energy stress. While mutations to either rbf or gig/dtsc1 cause modest ATP depletion and cell cycle defects, combined inactivation of rbf and gig/dtsc1 induces deregulated S phase, leading to DNA DSBs and more severe energy deficiency, resulting in dependence on LKB1 signaling, both of which contribute to cell death during Drosophila development.

Fig. 8.

Model for the regulation of cell death by RB–E2F and TSC1/TSC2. In addition to the repression of apoptotic signaling through Hid and JNK, the RB and TSC tumor suppressors regulate cell death by cooperating to prevent precocious S phase and energy stress. While mutations to either rbf or gig/dtsc1 cause modest ATP depletion and cell cycle defects, combined inactivation of rbf and gig/dtsc1 induces deregulated S phase, leading to DNA DSBs and more severe energy deficiency, resulting in dependence on LKB1 signaling, both of which contribute to cell death during Drosophila development.

In addition to inducing DNA DSBs, our results show that inactivation of rbf and gig/dtsc1 also induces energy stress. Evidence of the ability of oncogenes and tumor suppressors to modulate metabolism in addition to their roles in regulating cell proliferation and/or differentiation is emerging. TORC1 signaling, for example, is known to promote both G1–S progression as well as anabolic processes such as protein and lipid synthesis, and TORC1 deregulation sensitizes cells to various forms of stress, including energy stress. Indeed, studies have shown that mammalian cells lacking TSC1 or TSC2 are particularly sensitive to glucose deprivation (Inoki et al., 2003). Similarly recent studies show that RB–E2F regulates genes involved in oxidative metabolism (Bateman and McNeill, 2004) and that RB is required for the upregulation of mitochondrial biogenesis during erythropoiesis (Sankaran et al., 2008). Interestingly, cells with either rbf or gig/dtsc1 mutations crucially depend on cellular mechanisms that balance metabolic or energy supply and demand. While we found that inactivation of the metabolic regulator LKB1 significantly increases the death of rbf or dtsc1 mutants and largely eliminates mutant tissue from adult eyes, others showed LKB1 to be required for hematopoietic stem cell survival in mice (Gan et al., 2010; Gurumurthy et al., 2010; Nakada et al., 2010). Furthermore rbf and gig/dtsc1 mutants are sensitive to ATP depletion, as inactivation of cova, which impairs ATP production, also increases the death of either rbf or dtsc1 mutants and largely eliminates mutant tissue from adults. Therefore the enhanced energy stress we observed in rbf,gig double-mutants also probably contributes to the synergistic cell death phenotype (Fig. 8).

In an earlier study, we showed that another cellular stress, oxidative stress, is synergistically induced by inactivation of RB and TSC2 and contributes to the synergistic death of human cancer cells (Li et al., 2010). Notably, different types of stress in mammalian cells are often linked. For example, increased energy stress can induce accumulation of ROS as cells try to balance energy needs with supply by promoting mitochondrial oxidative phosphorylation, which is a principle intracellular source of oxidative stress. Although cancer cells often exhibit aerobic glycolysis, an observation known as the Warburg effect (Vander Heiden et al., 2009), they usually still depend on mitochondrial oxidative phosphorylation for survival (Fogal et al., 2010; Funes et al., 2007; Weinberg et al., 2010). Accumulation of ROS can also contribute to oncogene-induced DNA damage and cell death (Vafa et al., 2002), while increased DSBs increase ROS levels (Kang et al., 2011). Therefore increased energy stress and DSBs can induce accumulation of ROS, which contributes to the death of RB-mutant human cancer cells in which TSC2 is inactivated. By contrast, ROS do not appear to play a sufficient role in Drosophila apoptosis. For example, mutation of Pdsw, which encodes a component of mitochondrial complex I, causes accumulation of very high ROS levels and G1 cell cycle arrest but not cell death (Owusu-Ansah et al., 2008). Therefore, mechanisms in addition to ROS probably contribute to metabolic- or energy-stress-induced cell death in Drosophila. Interestingly, a recent study showed that caspase activation and cell death can be directly modulated by metabolic byproducts in flies (Kim et al., 2008).

A variety of cellular stresses can activate JNK signaling (Johnson and Nakamura, 2007). We found that JNK activity is involved in the sensitivity of rbf,gig cells to apoptosis. As Drosophila JNK signaling has been shown to promote expression of hid (Luo et al., 2007; Ryoo et al., 2004), an apoptotic gene similar to Diablo (Smac) that is involved in dE2f1-mediated cell death, it is possible JNK activation by deregulation of TORC1 can synergize with the loss of rbf to induce apoptosis owing to their effects on apoptotic gene expression (Fig. 8). The involvement of multiple mechanisms in the synergistic cell death of these double-mutants is consistent with our observations that hid and dronc are both important, but neither is absolutely required for the cell death phenotype of rbf,gig-mutant clones (Li et al., 2010). It should be noted that the ability of gig, tsc1, lkb1 or cova to induce synergistic cell death with rbf varies depending on the different regions of the eye disc. This is likely due to the fact that additional cell-intrinsic and extrinsic factors also influence the level of synergistic cell death. For example, it has been shown that Hid protein is preferentially accumulated near the MF in rbf-mutant eye discs (Tanaka-Matakatsu et al., 2009) and that epidermal growth factor receptor (EGFR) signaling, which is an important survival signal in the posterior of the eye disc (Yang and Baker, 2003), negatively regulates rbf-mediated cell death (Moon et al., 2006).

The mechanisms revealed by this and our previous research suggest there could be multiple benefits of transient activation of TORC1 signaling in therapies that target cancers with inactivated RB. In addition to inducing a significant death phenotype in RB-deficient cells, activation of TORC1 signaling in the absence of RB is expected to deregulate G1–S control synergistically and induce metabolic and energy stress. These effects could further sensitize RB-deficient cancer cells to conventional cancer treatments such as radiation and chemotherapy, as well as to new strategies that target cancer metabolism (Vander Heiden, 2011). As resistance to cancer therapy is believed to arise from the persistence of subsets of cells, such as cancer stem cells that survive the cytotoxic effects of chemotherapy or radiation by exiting the cell cycle and remaining dormant until growth conditions are more favorable (van Gent et al., 2001), transient activation of TORC1 activity might lead to better cure rates for RB-deficient cancers.

Drosophila stocks

The fly stocks used in this study were: rbf15aΔ (Tanaka-Matakatsu et al., 2009), de2f1i2 (Royzman et al., 1999), de2f1rm729 (Duronio et al., 1995), gig64 (Li et al., 2010), dtsc129 (Gao and Pan, 2001), s6kl1 (Radimerski et al., 2002), lkb1X5 (Lee et al., 2006), covatend (Mandal et al., 2005), hid138 (Tanaka-Matakatsu et al., 2009), dronc01 (Tanaka-Matakatsu et al., 2009), pucE69 (Riesgo-Escovar et al., 1996), UAS-E2f1,UAS-dDp (Bloomington stock no. 4774), UAS-Dap (Reis and Edgar, 2004), UAS-dWee1 (Price et al., 2002), UAS-Puc (Martín-Blanco et al., 1998), and UAS-Rheb(III) or strong UAS-Rheb(II) (Saucedo et al., 2003).

Drosophila genetics

Flies were maintained at 25°C on standard media containing yeast, cornmeal, sugar and agar. The genotypes used were as follows:

yw, hsFLP/Y; Ubi-GFP, FRT80B/gig, FRT80B

rbf15aΔ,w, hsFLP/Y;RBF-G3, Ubi-GFP, FRT80B/FRT80B

rbf15aΔ,w, hsFLP/Y;RBF-G3, Ubi-GFP, FRT80B/gig, FRT80B

rbf15aΔ,w, eyFLP/Y;RBF-G3, Ubi-GFP, FRT80B/FRT80B

rbf15aΔ,w, eyFLP/Y;RBF-G3, Ubi-GFP, FRT80B/gig, FRT80B

yw, eyFLP/Y; Ubi-GFP, FRT80B/gig, FRT80B, pucE69

rbf15aΔ,w, eyFLP/Y;RBF-G3, Ubi-GFP, FRT80B/gig, FRT80B, pucE69

rbf15aΔ,w, eyFLP/Y;RBF-G3, Ubi-GFP, FRT40A/FRT40A;pucE69/+

rbf15aΔ,w, eyFLP/Y;RBF-G3, Ubi-GFP, FRT80B/s6k, gig, FRT80B

rbf15aΔ,w, eyFLP/Y;RBF-G3, Ubi-GFP, FRT80B/hid, gig, FRT80B

rbf15aΔ,w, eyFLP/Y;RBF-G3, Ubi-GFP, FRT80B/dronc, gig, FRT80B

rbf15aΔ,w, eyFLP/Y;RBF-G3, Ubi-GFP, FRT80B, de2f1rm729/gig, FRT80B, de2f1i2

rbf15aΔ,w, hsFLP/Y;en-Gal4/UAS-Dap;RBF-G3, Ubi-GFP, FRT80B/gig, FRT80B

rbf15aΔ,w, hsFLP/Y;en-Gal4/UAS-dWee1;RBF-G3, Ubi-GFP, FRT80B/gig, FRT80B

rbf15aΔ,w, eyFLP/Y;Act >y >Gal4, UAS-GFP/+;FRT82B, RBF-G3, tub-Gal80/FRT82B dtsc1

rbf15aΔ,w, eyFLP/Y;Act >y >Gal4, UAS-GFP/UAS-Dap;FRT82B, RBF-G3, tub-Gal80/FRT82B

dtsc1

rbf15aΔ,w, eyFLP/Y;Act >y >Gal4, UAS-GFP/UAS-dWee1;FRT82B, RBF-G3, tub-Gal80/FRT82B

dtsc1

rbf15aΔ,w, eyFLP/Y;FRT82B, RBF-G3, Ubi-GFP/FRT82B

yw, eyFLP/Y;FRT82B, Ubi-GFP/FRT82B, dtsc1

rbf15aΔ,w, eyFLP/Y;FRT82B, RBF-G3, Ubi-GFP/FRT82B, dtsc1

yw, eyFLP/Y;FRT82B, Ubi-GFP/FRT82B, lkb1

yw, eyFLP/Y;FRT82B, Ubi-GFP/FRT82B, lkb1, dtsc1

rbf15aΔ,w, eyFLP/Y;FRT82B, RBF-G3, Ubi-GFP/FRT82B, lkb1

rbf15aΔ,w, eyFLP/Y;FRT82B, RBF-G3, Ubi-GFP/FRT82B, lkb1, dtsc1

rbf15aΔ,w, eyFLP/Y;FRT82B, RBF-G3, Ubi-GFP, rps3/FRT82B

rbf15aΔ,w, eyFLP/+;FRT82B, RBF-G3, Ubi-GFP, rps3/FRT82B, dtsc1

rbf15aΔ,w, eyFLP/Y;FRT82B, RBF-G3, Ubi-GFP, rps3/FRT82B, dtsc1

rbf15aΔ,w, eyFLP/+;FRT82B, RBF-G3, Ubi-GFP, rps3/FRT82B, cova

rbf15aΔ,w, eyFLP/Y;FRT82B, RBF-G3, Ubi-GFP, rps3/FRT82B, cova

rbf15aΔ,w, eyFLP/+;FRT82B, RBF-G3, Ubi-GFP, rps3/FRT82B, cova, dtsc1

rbf15aΔ,w, eyFLP/Y;FRT82B, RBF-G3, Ubi-GFP, rps3/FRT82B, cova, dtsc1

rbf15aΔ,w, eyFLP/Y;FRT82B, RBF-G3, Ubi-GFP, rps3/FRT82B, cova, dtsc1

yw,hsFLP/+;UAS-E2f1, UAS-dDp/+; Act >y >Gal4, UAS-GFP/+

yw,hsFLP/+;UAS-E2f1, UAS-dDp/UAS-Rheb; Act >y >Gal4, UAS-GFP/+

yw,hsFLP/+;UAS-E2f1, UAS-dDp/Act >y >Gal4, UAS-GFP;UAS-Rheb/+

yw,hsFLP/+;UAS-E2f1, UAS-dDp/UAS-Dap;Act >y >Gal4, UAS-GFP/UAS-Rheb

yw,hsFLP/+;UAS-E2f1, UAS-dDp/Act >y >Gal4, UAS-GFP;UAS-Rheb/UAS-Puc

yw,hsFLP/+;UAS-E2f1,dDp/UAS-Rheb, UAS-Dap; Act >y >Gal4, UAS-GFP/+

yw,hsFLP;UAS-E2f1, UAS-dDp/UAS-Rheb; Act >y >Gal4, UAS-GFP/UAS-Puc

yw,hsFLP;UAS-Rheb/Act >y >Gal4, UAS-GFP;pucE69/+

yw,hsFLP;UAS-E2f1, UAS-dDp/+;Act >y >Gal4, UAS-GFP/pucE69

yw,hsFLP;Act >y >Gal4/UAS-Rheb;pucE69

yw,hsFLP;UAS-Rheb/Act >y >Gal4, UAS-GFP;UAS-Puc/pucE69

yw,hsFLP;UAS-E2f1, UAS-dDp/UAS-Rheb;Act >y >Gal4, UAS-GFP/pucE69

Immunohistochemistry

All steps were performed at room temperature. Larval imaginal discs were dissected in 1× PBS, fixed in 4% formaldehyde in 1×PBS for 30 minutes and incubated with primary antibody diluted in 1× PBS plus 0.3% Triton-X100 (PBSTx) with 10% normal goat serum overnight at 4°C. The primary antibodies used were: rabbit anti-activated caspase-3 (C3, 1:300 from Cell Signaling), anti-P-H2Av (Mehrotra and McKim, 2006b) at 1:500; mouse anti-β-galactosidase (1:500, DSHB), anti-BrdU (1:100, DSHB) and anti-P-JNK (1:50 from Cell Signaling). Following incubation with primary antibody, samples were washed three times (10 minutes each) in PBSTx and incubated with secondary antibodies from Jackson ImmunoResearch (1:200 to 1:400). Sample mounting was performed using 70% glycerol with 1,4-diazabicyclo[2.2.2]octane (DABCO) at 12.5 mg ml–1. The β-galactosidase and BrdU antibodies were obtained from the Developmental Studies Hybridoma Bank (DSHB), developed under the auspices of the NICHD and maintained by The University of Iowa (Department of Biology, Iowa City, IA, USA). Imaging was achieved with the Zeiss Axioscope–ApoTome microscope using the AxioCam CCD camera controlled by Zeiss software.

Determination of cell death and γ-H2Av levels in developing imaginal discs

The percentage of pixels that had an above-background level of Caspase 3 (C3) staining was determined in mutant clones using the histogram function in Adobe Photoshop. The background level of C3 staining was determined as the level that was ≥95% of the C3 signal in adjacent wild-type tissues that had no apoptosis. The levels of γ-H2Av were determined as the number of positive cells per unit clone area, as quantified by Adobe Photoshop.

Nucleotide-analog incorporation assays

Incorporation of BrdU (Sigma) was performed at 75 ug ml–1 for 60 minutes in 1× PBS, followed by standard fixation. After two washes in PBST, samples were treated with 0.02 U µl–1 DNase I (Promega) for 1 hour at room temperature and then labeled using antibodies against BrdU following standard procedures. The EdU (Invitrogen) incorporation assay was performed according to the manufacturer's instructions; the EdU labeling reaction was achieved at 10 µM for 60 minutes and detected using the AlexaFluor 594 azide. To minimize GFP quenching, the fluorophore reaction was performed for 5 minutes.

G.M.G., T.Z. and W.D. conceived the experiments, analyzed the data and wrote the manuscript. G.M.G. and T.Z. were involved in all aspects of the experiments and data generation in this manuscript. J.Z. contributes to the Facs analysis data generation and manuscript preparation.

FACS analysis of dissociated wing disc cells

The method used to analyze the dissociated wing disc cells using FACS was adapted from de la Cruz and Edgar (de la Cruz and Edgar, 2008). Briefly, at least 20 larvae were used to procure 40 wing discs for each genotype. Discs were cleanly dissected in 1× PBS and then transferred into 4 ml of 5× trypsin-EDTA solution for 2–4 hours, with gentle agitation. Cells were then labeled for 30 minutes before FACS analysis with 1 µl of Vybrant DyeCycle Violet stain per 4 ml of trypsin solution. Dissociated cells were then sorted using the LSR-II Flow Cytometer (BD Biosciences) at the University of Chicago Flow Cytometry Facility. Data were analyzed using FlowJo software version 6.0 (Tree Star).

Determination of the ATP:ADP ratio

Relative ATP levels were determined after treating dissected eye imaginal discs with 120 µl 1× passive lysis buffer (Promega) by pipetting 15 times in a 1.5 ml tube on ice, boiling for 5 minutes, then incubating on ice for 2 minutes. After centrifugation at 18,000 g for 2 minutes, 20 µl of each sample was used to assay the ADP:ATP ratio by using the Enzylight kit according to the manufacturer's protocol (BioAssay Systems).

We thank Kim McKim, Duojia Pan, Shelagh Campbell, Utpal Banerjee, Jongkyeong Chung and Bruce Edgar for generously supplying antibodies, fly stocks and reagents. We thank the Bloomington Drosophila Stock Center and the Developmental Studies Hybridoma bank at the University of Iowa for providing fly stocks and antibodies. We also thank Binghui Li for assistance with FACS analysis and members of the Du laboratory, past and present, for helpful discussions.

Funding

This work was supported in part by the NIH [grant numbers NIH CA149275 to W.D., NIH GM074197 to W.D. and NIH/NCCAM AT004418 to W.D.]. Deposited in PMC for release after 12 months.

Bateman
J. M.
,
McNeill
H.
(
2004
).
Temporal control of differentiation by the insulin receptor/tor pathway in Drosophila.
Cell
119
,
87
96
.
Chen
H. Z.
,
Tsai
S. Y.
,
Leone
G.
(
2009
).
Emerging roles of E2Fs in cancer: an exit from cell cycle control.
Nat. Rev. Cancer
9
,
785
797
.
Choo
A. Y.
,
Kim
S. G.
,
Vander Heiden
M. G.
,
Mahoney
S. J.
,
Vu
H.
,
Yoon
S. O.
,
Cantley
L. C.
,
Blenis
J.
(
2010
).
Glucose addiction of TSC null cells is caused by failed mTORC1-dependent balancing of metabolic demand with supply.
Mol. Cell
38
,
487
499
.
Coschi
C. H.
,
Martens
A. L.
,
Ritchie
K.
,
Francis
S. M.
,
Chakrabarti
S.
,
Berube
N. G.
,
Dick
F. A.
(
2010
).
Mitotic chromosome condensation mediated by the retinoblastoma protein is tumor-suppressive.
Genes Dev.
24
,
1351
1363
.
Danos
A. M.
,
Liao
Y.
,
Li
X.
,
Du
W.
(
2012
).
Functional inactivation of Rb sensitizes cancer cells to TSC2 inactivation induced cell Death.
Cancer Lett.
328
,
36
43
.
Datar
S. A.
,
Jacobs
H. W.
,
de la Cruz
A. F.
,
Lehner
C. F.
,
Edgar
B. A.
(
2000
).
The Drosophila cyclin D-Cdk4 complex promotes cellular growth.
EMBO J.
19
,
4543
4554
.
de la Cruz
A. F.
,
Edgar
B. A.
(
2008
).
Flow cytometric analysis of Drosophila cells.
Methods Mol. Biol.
420
,
373
389
.
Duronio
R. J.
,
O’Farrell
P. H.
,
Xie
J. E.
,
Brook
A.
,
Dyson
N.
(
1995
).
The transcription factor E2F is required for S phase during Drosophila embryogenesis.
Genes Dev.
9
,
1445
1455
.
Fogal
V.
,
Richardson
A. D.
,
Karmali
P. P.
,
Scheffler
I. E.
,
Smith
J. W.
,
Ruoslahti
E.
(
2010
).
Mitochondrial p32 protein is a critical regulator of tumor metabolism via maintenance of oxidative phosphorylation.
Mol. Cell. Biol.
30
,
1303
1318
.
Funes
J. M.
,
Quintero
M.
,
Henderson
S.
,
Martinez
D.
,
Qureshi
U.
,
Westwood
C.
,
Clements
M. O.
,
Bourboulia
D.
,
Pedley
R. B.
,
Moncada
S.
 et al. (
2007
).
Transformation of human mesenchymal stem cells increases their dependency on oxidative phosphorylation for energy production.
Proc. Natl. Acad. Sci. USA
104
,
6223
6228
.
Gan
B.
,
Hu
J.
,
Jiang
S.
,
Liu
Y.
,
Sahin
E.
,
Zhuang
L.
,
Fletcher-Sananikone
E.
,
Colla
S.
,
Wang
Y. A.
,
Chin
L.
 et al. (
2010
).
Lkb1 regulates quiescence and metabolic homeostasis of haematopoietic stem cells.
Nature
468
,
701
704
.
Gao
X.
,
Pan
D.
(
2001
).
TSC1 and TSC2 tumor suppressors antagonize insulin signaling in cell growth.
Genes Dev.
15
,
1383
1392
.
Gordon
G. M.
,
Du
W.
(
2011
).
Conserved RB functions in development and tumor suppression.
Protein Cell
2
,
864
878
.
Gurumurthy
S.
,
Xie
S. Z.
,
Alagesan
B.
,
Kim
J.
,
Yusuf
R. Z.
,
Saez
B.
,
Tzatsos
A.
,
Ozsolak
F.
,
Milos
P.
,
Ferrari
F.
 et al. (
2010
).
The Lkb1 metabolic sensor maintains haematopoietic stem cell survival.
Nature
468
,
659
663
.
Hsieh
T. C.
,
Nicolay
B. N.
,
Frolov
M. V.
,
Moon
N. S.
(
2010
).
Tuberous sclerosis complex 1 regulates dE2F1 expression during development and cooperates with RBF1 to control proliferation and survival.
PLoS Genet.
6
,
e1001071
.
Inoki
K.
,
Zhu
T.
,
Guan
K. L.
(
2003
).
TSC2 mediates cellular energy response to control cell growth and survival.
Cell
115
,
577
590
.
Johnson
G. L.
,
Nakamura
K.
(
2007
).
The c-jun kinase/stress-activated pathway: regulation, function and role in human disease.
Biochim. Biophys. Acta
1773
,
1341
1348
.
Kaelin
W. G.
 Jr
(
2005
).
The concept of synthetic lethality in the context of anticancer therapy.
Nat. Rev. Cancer
5
,
689
698
.
Kang
Y. J.
,
Lu
M. K.
,
Guan
K. L.
(
2011
).
The TSC1 and TSC2 tumor suppressors are required for proper ER stress response and protect cells from ER stress-induced apoptosis.
Cell Death Differ.
18
,
133
144
.
Kim
I.
,
Xu
W.
,
Reed
J. C.
(
2008
).
Cell death and endoplasmic reticulum stress: disease relevance and therapeutic opportunities.
Nat. Rev. Drug Discov.
7
,
1013
1030
.
Lee
T.
,
Luo
L.
(
2001
).
Mosaic analysis with a repressible cell marker (MARCM) for Drosophila neural development.
Trends Neurosci.
24
,
251
254
.
Lee
J. H.
,
Koh
H.
,
Kim
M.
,
Park
J.
,
Lee
S. Y.
,
Lee
S.
,
Chung
J.
(
2006
).
JNK pathway mediates apoptotic cell death induced by tumor suppressor LKB1 in Drosophila.
Cell Death Differ.
13
,
1110
1122
.
Li
B.
,
Gordon
G. M.
,
Du
C. H.
,
Xu
J.
,
Du
W.
(
2010
).
Specific killing of Rb mutant cancer cells by inactivating TSC2.
Cancer Cell
17
,
469
480
.
Longworth
M. S.
,
Herr
A.
,
Ji
J. Y.
,
Dyson
N. J.
(
2008
).
RBF1 promotes chromatin condensation through a conserved interaction with the Condensin II protein dCAP-D3.
Genes Dev.
22
,
1011
1024
.
Luo
X.
,
Puig
O.
,
Hyun
J.
,
Bohmann
D.
,
Jasper
H.
(
2007
).
Foxo and Fos regulate the decision between cell death and survival in response to UV irradiation.
EMBO J.
26
,
380
390
.
Mandal
S.
,
Guptan
P.
,
Owusu-Ansah
E.
,
Banerjee
U.
(
2005
).
Mitochondrial regulation of cell cycle progression during development as revealed by the tenured mutation in Drosophila.
Dev. Cell
9
,
843
854
.
Manning
A. L.
,
Longworth
M. S.
,
Dyson
N. J.
(
2010
).
Loss of pRB causes centromere dysfunction and chromosomal instability.
Genes Dev.
24
,
1364
1376
.
Martín-Blanco
E.
,
Gampel
A.
,
Ring
J.
,
Virdee
K.
,
Kirov
N.
,
Tolkovsky
A. M.
,
Martinez-Arias
A.
(
1998
).
puckered encodes a phosphatase that mediates a feedback loop regulating JNK activity during dorsal closure in Drosophila.
Genes Dev.
12
,
557
570
.
Mehrotra
S.
,
McKim
K. S.
(
2006a
).
Temporal analysis of meiotic DNA double-strand break formation and repair in Drosophila females.
PLoS Genet.
2
,
e200
.
Mehrotra
S.
,
McKim
K. S.
(
2006b
).
Temporal analysis of meiotic DNA double-strand break formation and repair in Drosophila females.
PLoS Genet.
2
,
e200
.
Moon
N. S.
,
Di Stefano
L.
,
Dyson
N.
(
2006
).
A gradient of epidermal growth factor receptor signaling determines the sensitivity of rbf1 mutant cells to E2F-dependent apoptosis.
Mol. Cell. Biol.
26
,
7601
7615
.
Nakada
D.
,
Saunders
T. L.
,
Morrison
S. J.
(
2010
).
Lkb1 regulates cell cycle and energy metabolism in haematopoietic stem cells.
Nature
468
,
653
658
.
Ohsawa
S.
,
Sugimura
K.
,
Takino
K.
,
Xu
T.
,
Miyawaki
A.
,
Igaki
T.
(
2011
).
Elimination of oncogenic neighbors by JNK-mediated engulfment in Drosophila.
Dev. Cell
20
,
315
328
.
Orlova
K. A.
,
Crino
P. B.
(
2010
).
The tuberous sclerosis complex.
Ann. New York Acad. Sci.
1184
,
87
105
.
Owusu-Ansah
E.
,
Yavari
A.
,
Mandal
S.
,
Banerjee
U.
(
2008
).
Distinct mitochondrial retrograde signals control the G1-S cell cycle checkpoint.
Nat. Genet.
40
,
356
361
.
Ozcan
U.
,
Ozcan
L.
,
Yilmaz
E.
,
Düvel
K.
,
Sahin
M.
,
Manning
B. D.
,
Hotamisligil
G. S.
(
2008
).
Loss of the tuberous sclerosis complex tumor suppressors triggers the unfolded protein response to regulate insulin signaling and apoptosis.
Mol. Cell
29
,
541
551
.
Potter
C. J.
,
Huang
H.
,
Xu
T.
(
2001
).
Drosophila Tsc1 functions with Tsc2 to antagonize insulin signaling in regulating cell growth, cell proliferation, and organ size.
Cell
105
,
357
368
.
Price
D. M.
,
Jin
Z.
,
Rabinovitch
S.
,
Campbell
S. D.
(
2002
).
Ectopic expression of the Drosophila Cdk1 inhibitory kinases, Wee1 and Myt1, interferes with the second mitotic wave and disrupts pattern formation during eye development.
Genetics
161
,
721
731
.
Radimerski
T.
,
Montagne
J.
,
Hemmings-Mieszczak
M.
,
Thomas
G.
(
2002
).
Lethality of Drosophila lacking TSC tumor suppressor function rescued by reducing dS6K signaling.
Genes Dev.
16
,
2627
2632
.
Reis
T.
,
Edgar
B. A.
(
2004
).
Negative regulation of dE2F1 by cyclin-dependent kinases controls cell cycle timing.
Cell
117
,
253
264
.
Riesgo-Escovar
J. R.
,
Jenni
M.
,
Fritz
A.
,
Hafen
E.
(
1996
).
The Drosophila Jun-N-terminal kinase is required for cell morphogenesis but not for DJun-dependent cell fate specification in the eye.
Genes Dev.
10
,
2759
2768
.
Royzman
I.
,
Austin
R. J.
,
Bosco
G.
,
Bell
S. P.
,
Orr-Weaver
T. L.
(
1999
).
ORC localization in Drosophila follicle cells and the effects of mutations in dE2F and dDP.
Genes Dev.
13
,
827
840
.
Ryoo
H. D.
,
Gorenc
T.
,
Steller
H.
(
2004
).
Apoptotic cells can induce compensatory cell proliferation through the JNK and the Wingless signaling pathways.
Dev. Cell
7
,
491
501
.
Sankaran
V. G.
,
Orkin
S. H.
,
Walkley
C. R.
(
2008
).
Rb intrinsically promotes erythropoiesis by coupling cell cycle exit with mitochondrial biogenesis.
Genes Dev.
22
,
463
475
.
Saucedo
L. J.
,
Gao
X.
,
Chiarelli
D. A.
,
Li
L.
,
Pan
D.
,
Edgar
B. A.
(
2003
).
Rheb promotes cell growth as a component of the insulin/TOR signalling network.
Nat. Cell Biol.
5
,
566
571
.
Sekelsky
J. J.
,
Brodsky
M. H.
,
Burtis
K. C.
(
2000
).
DNA repair in Drosophila: insights from the Drosophila genome sequence.
J. Cell Biol.
150
,
F31
F36
.
Shackelford
D. B.
,
Shaw
R. J.
(
2009
).
The LKB1-AMPK pathway: metabolism and growth control in tumour suppression.
Nat. Rev. Cancer
9
,
563
575
.
Sherr
C. J.
,
McCormick
F.
(
2002
).
The RB and p53 pathways in cancer.
Cancer Cell
2
,
103
112
.
Steele
L.
,
Sukhanova
M. J.
,
Xu
J.
,
Gordon
G. M.
,
Huang
Y.
,
Yu
L.
,
Du
W.
(
2009
).
Retinoblastoma family protein promotes normal R8-photoreceptor differentiation in the absence of rhinoceros by inhibiting dE2F1 activity.
Dev. Biol.
335
,
228
236
.
Sukhanova
M. J.
,
Steele
L. J.
,
Zhang
T.
,
Gordon
G. M.
,
Du
W.
(
2011
).
RBF and Rno promote photoreceptor differentiation onset through modulating EGFR signaling in the Drosophila developing eye.
Dev. Biol.
359
,
190
198
.
Tanaka-Matakatsu
M.
,
Xu
J.
,
Cheng
L.
,
Du
W.
(
2009
).
Regulation of apoptosis of rbf mutant cells during Drosophila development.
Dev. Biol.
326
,
347
356
.
Tapon
N.
,
Ito
N.
,
Dickson
B. J.
,
Treisman
J. E.
,
Hariharan
I. K.
(
2001
).
The Drosophila tuberous sclerosis complex gene homologs restrict cell growth and cell proliferation.
Cell
105
,
345
355
.
Vafa
O.
,
Wade
M.
,
Kern
S.
,
Beeche
M.
,
Pandita
T. K.
,
Hampton
G. M.
,
Wahl
G. M.
(
2002
).
c-Myc can induce DNA damage, increase reactive oxygen species, and mitigate p53 function: a mechanism for oncogene-induced genetic instability.
Mol. Cell
9
,
1031
1044
.
van den Heuvel
S.
,
Dyson
N. J.
(
2008
).
Conserved functions of the pRB and E2F families.
Nat. Rev. Mol. Cell Biol.
9
,
713
724
.
van Gent
D. C.
,
Hoeijmakers
J. H.
,
Kanaar
R.
(
2001
).
Chromosomal stability and the DNA double-stranded break connection.
Nat. Rev. Genet.
2
,
196
206
.
van Harn
T.
,
Foijer
F.
,
van Vugt
M.
,
Banerjee
R.
,
Yang
F. T.
,
Oostra
A.
,
Joenje
H.
,
te Riele
H.
(
2010
).
Loss of Rb proteins causes genomic instability in the absence of mitogenic signaling.
Genes Dev.
24
,
1377
1388
.
Vander Heiden
M. G.
(
2011
).
Targeting cancer metabolism: a therapeutic window opens.
Nat. Rev. Drug Discov.
10
,
671
684
.
Vander Heiden
M. G.
,
Cantley
L. C.
,
Thompson
C. B.
(
2009
).
Understanding the Warburg effect: the metabolic requirements of cell proliferation.
Science
324
,
1029
1033
.
Weinberg
F.
,
Hamanaka
R.
,
Wheaton
W. W.
,
Weinberg
S.
,
Joseph
J.
,
Lopez
M.
,
Kalyanaraman
B.
,
Mutlu
G. M.
,
Budinger
G. R.
,
Chandel
N. S.
(
2010
).
Mitochondrial metabolism and ROS generation are essential for Kras-mediated tumorigenicity.
Proc. Natl. Acad. Sci. USA
107
,
8788
8793
.
Yang
L.
,
Baker
N. E.
(
2003
).
Cell cycle withdrawal, progression, and cell survival regulation by EGFR and its effectors in the differentiating Drosophila eye.
Dev. Cell
4
,
359
369
.
Zhang
Y.
,
Gao
X.
,
Saucedo
L. J.
,
Ru
B.
,
Edgar
B. A.
,
Pan
D.
(
2003
).
Rheb is a direct target of the tuberous sclerosis tumour suppressor proteins.
Nat. Cell Biol.
5
,
578
581
.

Supplementary information