Main

To investigate the molecular mechanism behind the resistance of CSCs to therapeutics, we studied the death of stem cells with different degrees of quiescence in the adult Drosophila digestive system, including intestinal stem cells (ISCs)2,3, renal and nephric stem cells (RNSCs)4 and hindgut intestinal stem cells (HISCs)5,6 (Fig. 1a and Extended Data Fig. 1a). We found that expression of the proapoptotic genes rpr and p53 effectively ablated differentiated cells but had little effect on stem cells (Extended Data Fig. 1b–n).

Figure 1: Activation of proliferation accelerates apoptotic cell death of hyperplastic stem cells but fails to completely eliminate neoplastic stem cells.
figure 1

a, Diagram of three types of stem cells near the hindgut–midgut junction, and the cells in which esgGal4 (esg) and wgGal4 (wg) were expressed. b–i, Representative images of the posterior midguts of flies with the indicated phenotypes. b, esgts > upd, 29 °C, 4 d (n = 33). c, esgts > upd + rpr, 29 °C, 4 d (n = 35). d, esgts > RasV12, 29 °C, 7 d (n = 34). e, esgts > RasV12 + rpr, 29°C, 7 d (n = 29). f, esgts > RasV12 + Arf79FRNAi, 29 °C, 7 d (n = 35). g, esgts > NDN, 29 °C, 7 d (n = 24). h, esgts > NDN + rpr, 29 °C, 7 d (n = 29). i, esgts > NDN + Arf79FRNAi, 29 °C, 7 d (n = 38). j, Quantification of GFP+ cells from midguts isolated from flies with the indicated genotypes. Data are represented as mean ± s.e.m. Statistical significance determined by Student’s t-test, ***P < 0.0001. The posterior midguts of flies with the indicated genotypes were dissected, stained with the GFP and Prospero (Pros) antibodies and analysed by confocal microscopy. White arrows in b and c point to the hindgut–midgut junction. g, h, Red arrows with white dotted lines point to clusters of ISCs enteroblasts and yellow arrows with yellow dotted lines point to clusters of enteroendocrine cells. i, Red and yellow arrows point to remaining ISCs/enteroblasts, and enteroendocrine cells, respectively. Scale bars in b–i, 10 μm.

PowerPoint slide

Source data

In mammals, treatment-resistant leukaemic stem cells (LSCs) can be eliminated by a two-step protocol involving initial activation by interferon-α (IFNα) or colony-stimulating factor (G-CSF), followed by targeted chemotherapy7. In Drosophila, activation of the hopscotch (also known as JAK)–Stat92E signalling pathway induces hyperplastic stem cells, which are overproliferating, but retain their apico-basal polarity and differentiation ability4,6,8. We conducted a slightly different two-step protocol in Drosophila stem cells by overexpressing the JAK–Stat92E pathway ligand unpaired (upd) and rpr together. The induction of upd + rpr using the temperature-sensitive (ts) mutant esg–Gal4 (esgts > upd + rpr; Fig. 1b, c, j and Extended Data Fig. 1o–q) effectively ablated all of the ISCs and RNSCs through apoptosis within four days. Consistent with this result, expressing a gain-of-function Raf mutant (Rafgof) also accelerated apoptotic cell death of hyperplastic ISCs9.

Expressing a constitutively active form of Ras oncogene at 85D (also known as RasV12) in RNSCs10 and the knockdown of Notch activity in ISCs11,12 can transform these cell types into CSC-like neoplastic stem cells, which were not only overproliferating, but also lost their apico-basal polarity and differentiation ability (Fig. 1d, g). We found that expressing rpr in RasV12-transformed RNSCs (esgts > RasV12 + rpr; Fig. 1e, j) or in ISCs expressing a dominant–negative form of Notch (NDN) (esgts > NDN + rpr; Fig. 1h, j) caused the ablation of only a proportion of the transformed RNSCs and few transformed ISCs and it did not affect differentiated cells (Extended Data Fig. 1r–u); substantial populations of the neoplastic stem cells remained even seven days after rpr induction.

These results suggest that the activation of proliferation can accelerate the apoptotic cell death of hyperplastic stem cells, but that a proportion of actively proliferating neoplastic RNSCs and ISCs are resistant to apoptotic cell death. Neoplastic tumours in Drosophila are more similar to high-grade malignant human tumours than are the hyperplastic Drosophila tumours13.

Vesicle-mediated COPI and COPII are essential components of the trafficking machinery for vesicle transportation between the endoplasmic reticulum and the Golgi14. In addition, the COPI complex regulates the transport of lipolysis enzymes to the surface of lipid droplets for lipid droplet usage15 (Extended Data Fig. 2a). In our previous screen, we found that knockdown of COPI components (including Arf79F, the Drosophila homologue of ADP-ribosylation factor 1 (Arf1)) rather than COPII components16 resulted in stem-cell death, suggesting that lipid-droplet usage (lipolysis) rather than the general trafficking machinery between the endoplasmic reticulum and Golgi is important for stem-cell survival.

To further investigate the roles of these genes in stem cells, we used a recombined double Gal4 line of esg–Gal4 and wg–Gal4 to express genes in ISCs, RNSCs, and HISCs (esgts wgts > X). Knockdown of these genes using RNA interference (RNAi) in stem cells (esgts wgts > XRNAi; Extended Data Fig. 2b–k) ablated most of the stem cells in 1 week. However, expressing Arf79FRNAi in enterocytes (NP1ts > Arf79FRNAi; Extended Data Fig. 2l–o) or in differentiated stellate cells in Malpighian tubules (tshts > Arf79FRNAi; Extended Data Fig. 2p, q) did not cause similar marked ablation. These results suggest that Arf79F knockdown selectively kills stem cells and not differentiated cells.

We also found that expressing Arf79FRNAi (esgts > RasV12 + Arf79FRNAi; Fig. 1f, j) or ζ-COPRNAi (esgts wgts >RasV12 + ζ-COPRNAi; Extended Data Fig. 2r) in RasV12-transformed RNSCs ablated almost all of the transformed stem cells. Similarly, expressing Arf79FRNAi (esgts > NDN + Arf79FRNAi; Fig. 1i, j) or δ-COPRNAi (esgts wgts > NDN + δ-COPRNAi; Extended Data Fig. 2s) in NDN-transformed ISCs ablated all of the cells within one week, but restored differentiated cells to close to their normal levels within one week (Extended Data Fig. 2t, u).

We further generated δ-COP- and γ-COP-mutant clones using the mosaic analysis with a repressible cell marker (MARCM) technique17 and found that the COPI complex cell-autonomously regulated stem cell survival (Extended Data Fig. 3a–h). In summary, knockdown of the COPI–Arf79F complex effectively ablated normal and transformed stem cells but not differentiated enterocytes or stellate cells.

In our RNAi screen we also identified acyl-CoA synthetase long-chain (ACSL), an enzyme in the Drosophila lipolysis–β-oxidation pathway16,18 (Extended Data Fig. 2a), and bubblegum (bgm), a very long-chain fatty acid-CoA ligase16,19. RNAi-mediated knockdown of Acsl (esgts wgts > AcslRNAi; Extended Data Fig. 2i, k) and bgm (esgts wgts > bgmRNAi; Extended Data Fig. 2j, k) effectively killed ISCs and RNSCs, but killed HISCs less effectively. Expressing AcslRNAi in RasV12-transformed RNSCs (esgts wgts > RasV12 + AcslRNAi; Extended Data Fig. 2v) also ablated almost all of the transformed RNSCs in one week.

Brummer (bmm) is a triglyceride lipase, the Drosophila homologue of mammalian ATGL, the first enzyme in the lipolysis pathway20 (Extended Data Fig. 3a). Scully (scu) is the Drosophila orthologue of hydroxy-acyl-CoA dehydrogenase, an enzyme in the β-oxidation pathway21. Hepatocyte nuclear factor 4 (Hnf4) regulates the expression of several genes involved in lipid mobilization and β-oxidation21. To determine whether the lipolysis–β-oxidation pathway is required for COPI–Arf79F-mediated stem cell survival, we expressed upstream activating sequence (UAS)-regulated constructs (UAS–bmm (Fig. 2c, h), UAS–Hnf4 (Fig. 2d, h), and UAS–scu (Fig. 2e, h)) in stem cells that were depleted of Arf79F (Fig. 2b–e), β-COP (Extended Data Fig. 2w), or ζ-COP (Extended Data Fig. 2x). Overexpressing either scu or Hnf4 significantly (P < 0.0001) attenuated the stem cell death caused by knockdown of the COPI–Arf79F complex. Expressing UAS–Hnf4 (Fig. 2i) and UAS–scu (Fig. 2j) in FRT82Bγ-COP10 MARCM clones also rescued the stem cell death phenotype induced by γ-COP knockdown (Extended Data Fig. 3f, h). However, bmm overexpression did not rescue the stem-cell death induced by Arf79F knockdown (Fig. 2c, h). Since there are several other triglyceride lipases in Drosophila in addition to bmm, another lipase may redundantly regulate the lipolysis pathway.

Figure 2: The COPI–Arf79F complex regulates stem cell survival through a lipolysis pathway.
figure 2

ag, Representative images are shown. The genotypes of the flies in each panel were: a, esgts wgts > lacZRNAi, 29 °C, 7 d (n = 34). b, esgts wgts > Arf79FRNAi, 29 °C, 7 d (n = 38). c, esgts wgts > Arf79FRNAi + bmm, 29 °C, 7 d (n = 31). d, esgts wgts > Arf79FRNAi + Hnf4, 29 °C, 7 d (n = 29). e, esgts wgts > Arf79FRNAi + scu, 29 °C, 7 d (n = 32). f, esgts wgts > Arf79FRNAi + p35, 29 °C, 7 d (n = 37). g, esgts wgts > Arf79FRNAi + Atg12RNAi, 29 °C, 7 d (n = 32). h, Quantification of GFP+ cells from midguts isolated from flies with the indicated genotypes. Data are represented as mean ± s.d. Statistical significance determined by Student’s t-test, ***P < 0.0001. NS, not significant (P > 0.05). ik, MARCM clones of flies with the following genotypes: i, UASHnf4; FRT82B–γ-Cop10, 7 d (n = 30) after clonal induction (ACI). j, UAS-scu; FRT82B-γ-Cop10, 7 d ACI (n = 32). k, UAS–cat; FRT82Bγ-Cop10, 7 d ACI (n = 35). The posterior midguts of flies with the indicated genotypes were dissected, stained with the GFP, Prospero (Pros) and Armadillo (Arm) antibodies and analysed by confocal microscopy. White arrows in ag point to the hindgut–midgut junction. White dotted lines in ik outline GFP+ clones. Scale bars in ag and ik, 10 μm.

PowerPoint slide

Source data

To further investigate the function of lipolysis in stem cells, we investigated the expression of a lipolysis reporter (GAL4–dHFN4; UAS–nlacZ)21, which consisted of hsp70–GAL4–dHNF4 combined with a UAS–nlacZ reporter gene. The flies were either cultured continuously at 29 °C or heat-shocked for 30 min at 37 °C, 12 h before dissection. Without heat shock, the reporter was expressed only in ISCs and RNSCs of mature adult flies, but not in enteroendocrine cells, enterocytes, quiescent HISCs or quiescent ISCs of freshly emerged young adult flies (less than 3 days old) (Extended Data Fig. 3i–m). Expressing δ-COPRNAi (esgts > δ-COPRNAi + GAL4–dHFN4; UAS–nlacZ) almost completely eliminated the reporter expression (Extended Data Fig. 3n), suggesting that the reporter was specifically regulated by the COPI complex. After heat shock or when a constitutively active form of JAK (hopTum-l) was expressed, the reporter was strongly expressed in ISCs, RNSCs and HISCs, but not in enteroendocrine cells or enterocytes (Extended Data Fig. 3o, p). These data suggest that COPI-complex-regulated lipolysis was active in stem cells, but not in differentiated cells, and that the absence of the reporter expression in quiescent HISCs at 29 °C was probably owing to weak hsp70 promoter activity rather than to low lipolysis in these cells.

We further investigated lipid storage, and found that the size and number of lipid droplets were markedly increased in stem cells after knockdown of Arf79F (esgts > Arf79FRNAi) (Extended Data Fig. 3q–v).

We also used Arf1 inhibitors (brefeldin A, golgicide A, secin H3, LM11 and LG8) and fatty-acid-oxidation (FAO) inhibitors (triacsin C, mildronate, etomoxir and enoximone) and found that these inhibitors markedly reduced stem-cell tumours in Drosophila through the lipolysis pathway but had a negligible effect on normal stem cells (Extended Data Fig. 4).

These data together suggest that the COPI–Arf1 complex regulates stem-cell survival through the lipolysis–β-oxidation pathway, and that knockdown of these genes blocks lipolysis but promotes lipid storage. Further, the transformed stem cells are more sensitive to Arf1 inhibitors and may be selectively eliminated by controlling the concentration of Arf1 inhibitors.

Our data suggest that neither caspase-mediated apoptosis nor autophagy-regulated cell death regulates the stem-cell death induced by the knockdown of components of the COPI–Arf79F complex (Fig. 2f–h). We therefore investigated whether necrosis regulates the stem-cell death induced by knockdown of the COPI–Arf79F complex. Necrosis is characterized by early plasma membrane rupture, reactive oxygen species (ROS) accumulation and intracellular acidification22. Propidium iodide detects necrotic cells with compromised membrane integrity, the oxidant-sensitive dye dihydroethidium (DHE) indicates cellular ROS levels and LysoTracker staining detects intracellular acidification22. We detected the membrane rupture phenotype only in esgts.wgts > Arf79FRNAi ISCs but not in wild-type ISCs (Figs 3a–d, 4a and Extended Data Fig. 5a–i) and the propidium iodide signal was observed only in ISCs from flies that had RNAi-induced knockdown of expression of COPI–Arf79F components (esgts wgts > XRNAi, Fig. 3g–i, l and Extended Data Fig. 5k–p), and not in cells from wild-type (Fig. 3e, f, l and Extended Data Fig. 5j, p), scu-rescued Arf79FRNAi (esgts wgts > scu + Arf79FRNAi, Fig. 3j, l) or Hnf4-rescued Arf79FRNAi (esgts wgts > Hnf4 + Arf79FRNAi, Fig. 3k, l) flies. In the esgts wgts > AcslRNAi flies, all of the ISCs and RNSCs were ablated after four days at 29 °C, but a fraction of the HISCs remained, and these were also propidium iodide positive (Extended data Fig. 5n–p), indicating that the HISCs were dying slowly. This slowness may have been due to either a lower GAL4 (wg–Gal4) activity in these cells compared to ISCs and RNSCs (esg–Gal4) or quiescence of the HISCs. Furthermore, strong propidium iodide signals were detected in transformed ISCs from esgts > NDN + Arf79FRNAi but not esgts > NDN flies (Extended data Fig. 6a–d), indicating that the transformed stem cells were dying through necrosis.

Figure 3: Knockdown of components of the COPI–Arf79F–β-oxidation pathway kill stem cells through necrosis.
figure 3

The genotypes of the flies in each panel were: a, b, e, f, esgts wgts > lacZRNAi, 29 °C, 4 d (n = 32). c, d, g, h, esgts wgts > Arf79FRNAi, 29 °C, 4 d (n = 28). i, esgts wgts > δ-CopRNAi, 29 °C, 4 d (n = 30). j, esgts wgts > Arf79FRNAi + scu, 29 °C, 4 d (n = 28). k, esgts wgts > Arf79FRNAi + Hnf4, 29 °C, 4 d (n = 30). l, Quantification of PI+ cells from midguts isolated from flies with the indicated genotypes. Data are represented as mean ± s.d. Statistical significance determined by Student’s t-test, ***P < 0.0001; NS, not significant (P > 0.05). The posterior midguts of flies with the indicated genotypes were dissected, stained with GFP and Armadillo (Arm) antibodies or indicated reagents, and analysed by confocal microscopy. White arrows in ai point to esg–GFP+ cells and in jk point to the hindgut–midgut junction. White dotted lines in d outline ISCs/enteroblasts, yellow dotted lines outline enterocytes. Scale bars in ak, 10 μm.

PowerPoint slide

Source data

Figure 4: Dying ISCs are engulfed by neighbouring enterocytes through the draper–Rac–JNK (Bsk) pathway.
figure 4

a, Representative images from esgts > Arf79FRNAi cells, 29°C, 7 d (n = 32). A dying ISC is engulfed by a neighbouring enterocyte. The posterior midguts of flies were dissected, stained with the GFP and Armadillo (Arm) antibodies, and analysed by confocal microscopy. Arrow points to GFP+ stem cell/progenitors and the dotted line outlines a nucleus and cell membrane of an enterocyte. Scale bar, 10 μm. b, Quantification of Dl+ (Delta+) ISCs cells in flies with the NP1ts esgts driver and indicated genotypes (See images in Extended Data Fig. 9 for details). Data are represented as mean ± s.e.m. Statistical significance determined by Student’s t-test, *P < 0.05, ***P < 0.0001. NS, not significant (P > 0.05). c, Model of ISC death induced by knockdown of the COPI–Arf1 complex. Details are described in the text. The autophagosome is involved in the last step of phagocytosis (degradation of internalized cargo)27, and autophagy may both function downstream of and be regulated by the drpr–Mbc–Rac1–JNK pathway.

PowerPoint slide

Source data

Similarly, DHE (Extended Data Fig. 6e–h) or LysoTracker (Extended Data Fig. 6i–l) signals were detected only in ISCs from esgts > Arf79FRNAi flies (Extended Data Fig. 6g, h, k, l), but not from wild-type flies (Extended Data Fig. 6e, f, i, j), indicating that the dying ISCs had accumulated ROS and were intracellularly acidified. Overexpressing catalase (a ROS-chelating enzyme) rescued the stem-cell death specifically induced by the γ-COP mutant clone (Fig. 2k) or by Arf79F knockdown (Extended Data Fig. 7b), and the ROS inhibitor NAC blocked the Arf1 inhibitor-induced death of RasV12-induced RNSC tumours (Extended Data Fig. 4i, l). These data together suggest that knockdown of the COPI–Arf1 complex induced the death of stem cells or of transformed stem cells (RasV12–RNSCs, NDN–ISCs) through ROS-induced necrosis. Although ISCs, RNSCs, and HISCs exhibit different degrees of quiescence, they all rely on lipolysis for survival, suggesting that this is a general property of stem cells.

We noticed cases where the GFP-positive material of the dying ISCs was present within neighbouring enterocytes (Fig. 4a, Extended Data Fig. 5a–i), suggesting that these enterocytes had engulfed dying ISCs.

The JNK pathway, autophagy and engulfment genes are involved in the engulfment of dying cells23,24. We therefore investigated whether these genes are required for COPI–Arf79F-regulated ISC death. We found that: (1) ISC death activated JNK signalling and autophagy in neighbouring enterocytes (Extended Data Fig. 7i–n); (2) knockdown of these genes in enterocytes but not in ISCs rescued ISC death to different degrees (Fig. 4b and Extended Data Figs 8a–i, 9a–l); (3) the drpr–mbc–Rac1–JNK pathway in enterocytes is not only necessary but also sufficient for ISC death (Extended Data Figs 8j–n and 9m, n); and (4) inhibitors of JNK and Rac1 could block Arf1-inhibitor-induced cell death of the RasV12-induced RNSC tumours (Extended Data Fig. 4g, h, l). These data together suggest that the drpr–mbc–Rac1–JNK pathway in neighbouring differentiated cells controls the engulfment of dying or transformed stem cells (Fig. 4c).

Our finding that the COPI–Arf79F–lipolysis–β-oxidation pathway regulated transformed stem-cell survival in the fly led us to investigate whether the pathway has a similar role in CSCs. We tested two Arf1 inhibitors (brefeldin A and golgicide A) and two FAO inhibitors (triascin C and etomoxir) on human cancer cell lines, and found that the growth, tumoursphere formation and expression of tumour-initiating cell markers of the four cancer cell lines were significantly (P < 0.01) suppressed by these inhibitors (Extended Data Fig. 10), suggesting that these inhibitors suppress CSCs. In mouse xenografts of BSY-1 human breast cancer cells, a novel low-cytotoxicity Arf1-ArfGEF inhibitor called AMF-26 was reported to induce complete regression in vivo in five days25. Together, this report and our results suggest that inhibiting Arf1 activity or blocking the lipolysis pathway can kill CSCs and block tumour growth.

Stem cells or CSCs are usually localized to a hypoxic storage niche, surrounded by a dense extracellular matrix26, which may make them less accessible to sugar and amino acid nutrition from the body’s circulatory system. Most normal cells rely on sugar and amino acids for their energy supply, with lipolysis playing only a minor role in their survival. Our results suggest that stem cells and CSCs are metabolically unique; they rely mainly on lipid reserves for their energy supply, and blocking COPI–Arf1-mediated lipolysis can starve them to death. We further found that transformed stem cells were more sensitive than normal stem cells to Arf1 inhibitors (Extended Data Fig. 4). Thus, selectively blocking lipolysis may kill CSCs without severe side effects. Therefore, targeting the COPI–Arf1 complex or the lipolysis pathway may prove to be a well-tolerated, novel approach for eliminating CSCs.

Methods

Fly strains

The following fly strains were used: NP1Gal4 and FRT19Aδ-COPG0051 (from DGRC); esgGal4 and tshGal4 (from S. Hayashi); wgGal4 (from J.-P. Vincent); UASupd (generated in our laboratory); UASNDN(2X) (from M. Fortini); UASbmm (from R. P. Kuhnlein); UASHnf4 and GAL4dHFN4 ; UASnlacZ (from C. Thummel); miraGFP (from F. Schweisguth); pmCherryAtg8a (from E. Baehrecke); UASGAP43mCherry (from T. Lecuit); UASdrprRNAi and UASdrpr (from M. Freeman); UASDJunAsp (from M. Mlodzik); UAShepCA, UASRasV12, UASscu, UASrpr, UASp53, UAS–p35, UAS–Cat, UAS–Sod, UAS–Sod2, UAS–RacDN(N17), UAS–RacV12, UAS–bskDN, hopTum-l, puc–lacZ (pucE69), UAS–2XEYFP, tub–Gal80ts, and fly lines used for MARCM clones (FRT19A, tub–Gal80; FRT82B, tub–Gal80; SM6, hs–flp; MKRS, hs–flp; act>y+>Gal4, UAS–GFP; FRT82Bγ-COP10; and UAS–γ-COP FRT82Bγ-COP10) were obtained from the Bloomington Drosophila Stock Center at Indiana University.

RNAi stocks

An upstream activating sequence (UAS)-regulated double-stranded inverse-repeat construct was designed to target Arf79F: (UASArf79FRNAi)-VDRC Transformant ID: 23082 (v23082). The RNA level was reduced to 39.0% in the Act–Gal4UAS–Arf79FRNAi flies (ref. 16), and the phenotypes were confirmed by two independent RNAi lines (v103572 and v23080). The other RNAi lines used were: δ-COP–v41551; RNA level was reduced to 14.3% in Act–Gal4UAS–δ-COPRNAi flies (ref. 16), and phenotypes were confirmed by an independent RNAi line (Bloomington stock number 31764 (BL31764 (TRiP ID HM04076)). β-COP–BL31709 (TRiP ID HM04016); RNA level was reduced to 13.3% in ActGal4UAS–β-COPRNAi flies (ref. 16), and phenotypes were confirmed by two independent RNAi lines (v109641 and v15418). β’-COP–BL31710 (TRiP ID HM04017); RNA level was reduced to 3.2% in ActGal4UAS–β’-COPRNAi flies (ref. 16). ζ-COP-BL28960 (TRiP ID HM05171); RNA level was reduced to 47.0% in ActGal4UASζ-COPRNAi flies (ref. 16), and phenotypes were confirmed by two independent RNAi lines (v34768 and v104405). garz-BL31232 (TRiP ID JF01013); RNA level was reduced to 52.4% in the ActGal4UASgarzRNAi flies (ref. 16). Acsl–BL27729 (TRiP ID JF02811); RNA level was reduced to 25.5% in ActGal4UASAcslRNAi flies (ref. 16). bgm-v34854; RNA level was reduced to 56.2% in ActGal4UASbgmRNAi flies (ref. 16), and phenotypes were confirmed by two independent RNAi lines (v105635 and BL28639 (TRiP ID JF03054)). γ-COP–BL28889 (TRiP IDHM05099); Atg5–BL34899 (TRiP ID HMS01244); Atg12RNAi (from E. Baehrecke, ref. 28); mbc–BL32355 (TRiP ID HMS00346); PSR–BL33700 (TRiP ID HMS00576); mys–BL33642 (TRiP ID HMS00043); CycT–BL32976 (TRiP ID HMS00776). The sequences used for each VDRC knock-down strain are available at https://stockcenter.vdrc.at) and for each Bloomington knock-down strain at http://flystocks.bio.indiana.edu. The data presented in all figures were generated by using the first RNAi line for all genes.

MARCM clone assay

To induce MARCM clones, three- or four-day-old adult female flies were heat-shocked twice with an interval of 8–12 h at 37 °C for 60 min. The flies were transferred to fresh food daily after the final heat shock and their posterior midguts were processed for staining at the indicated times.

RNAi-mediated gene depletion

To target the expression of UAS-linked genes in the cell types of interest, we used specific drivers. The posterior midgut of adult Drosophila is maintained by multipotent ISCs2,3, which differentiate into secretory enteroendocrine cells and absorptive enterocytes through immature enteroblasts. Enterocytes are polyploid and express the transcription factor Pdm1. Enteroendocrine cells are diploid and express the transcription factor Prospero (Pros). UAS-linked genes can be targeted to enterocytes by the MyolAGal4 (NP1Gal4) driver8 or to ISCs and enteroblasts by the escargot (esg)–Gal4 driver2. To target expression of UAS-linked genes in RNSCs, we also used esgGal4 (ref. 4); for the quiescent HISCs, the wingless (wg)–Gal4 driver was used5,6. To investigate the response of the different cells to cell-death effectors, we first overexpressed reaper (rpr, an inhibitor of Death-associated inhibitor of apoptosis 1; Diap-1) in them, using the cell-type-specific Gal4 drivers combined with the temperature-sensitive Gal4 repressor tub–Gal80ts (ref. 29).

We used the inducible NP1Gal4; tub–Gal80tsUAS–rpr; UAS–GFP to express rpr in enterocytes (NP1ts > rpr), esg–Gal4; tubGal80tsUAS–rpr; UAS–GFP (esgts > rpr) in ISCs, enteroblasts and RNSCs, and wg–Gal4; tubGal80tsUAS–rpr; UAS–GFP (wgts > rpr) in HISCs. The NP1ts > rpr flies were raised to adulthood at 18 °C and shifted to 29 °C for 24 h to induce rpr expression.

Four male UASRNAi transgene flies were crossed with 8 female virgins of NP1ts, esgts, and wgts at 18 °C. Three- to five-day-old adult flies with the appropriate genotype were transferred to new vials at 29 °C for the indicated times before dissection. For p53, we did not find a significant change in esg+ progenitors and enteroendocrine cell numbers after the flies (esgts > p53) were cultured for 7 days at 29 °C, although a previous study found that a 15-day induction ablated nearly all esg+ cells and reduced enteroendocrine cell numbers8.

Histology and image capture

Fly intestines were dissected in PBS and fixed in PBS containing 4% formaldehyde for 30 min. After three 5-min rinses with 1× PBT (PBS + 0.1% Triton X-100), the samples were blocked in 1× PBT containing 5% normal goat serum overnight at 4 °C and incubated first with primary antibody overnight at 4 °C or at room temperature for 2 h, and then with a fluorescence-conjugated secondary antibody for 2 h at room temperature. Samples were mounted in Vectashield mounting medium with DAPI (Vector Laboratories). The following antibodies were used: rabbit polyclonal anti-β-Gal (1:1,000; Cappel); mouse anti-Dl (Delta, 1:20; DSHB); mouse monoclonal anti-Prospero (Pros, 1:50; DSHB); rabbit polyclonal anti-Pdm1 (1:1,000, a gift from X. Yang); mouse monoclonal anti-Arm N27A1 (1:20; DSHB); Rabbit monoclonal anti-Phospho-SAPK/JNK (1:200; Cell Signaling); rabbit-polyclonal anti-GFP (1:500, Invitrogen); mouse monoclonal anti-GFP (1:100; Invitrogen), and chicken polyclonal anti-GFP (1:3,000; Abcam). Secondary antibodies were goat anti-mouse, anti-chicken, and goat anti-rabbit IgG conjugated to Alexa488 or Alexa568 (1:400; Invitrogen). DAPI (Sigma) was used to stain DNA. CellMask Deep Red plasma membrane dye (Life Technologies, C10046) was used to visualize the plasma membrane. Midguts were labelled with CellMask Deep Red Plasma Membrane Stains (1:2,000) for 7 min30.

ROS detection by DHE

DHE staining was performed as described previously31. In brief, guts were dissected in 1× PBS, incubated in 30 μM DHE (Invitrogen) in PBS for 5 min at room temperature in the dark, washed twice, mounted and immediately imaged by confocal microscopy.

Lysotracker staining

Guts were dissected in 1× PBS and then stained without fixation in 0.5 μM Lysotracker Red DND-99 (Invitrogen) for 3 min at room temperature. They were then washed three times in 1× PBS, fixed for 20 min in 4% formaldehyde, washed three times in 1× PBT, rinsed twice with 1× PBS, mounted in Vectashield with DAPI and analysed on a confocal microscope.

Apoptosis detection

Apoptosis was detected by TUNEL with the ApopTag Red In situ Apoptosis Detection Kit (Chemicon International) according to the manufacturer’s instructions.

Necrosis evaluation by propidium iodide

Guts were dissected in 1× PBS and then stained in 1.5 μM PI (Invitrogen) for 15 min at room temperature. The guts were then fixed for 20 min in 4% formaldehyde, washed three times in 1× PBT, rinsed twice with 1× PBS, mounted in Vectashield with DAPI and analysed on a confocal microscope.

Oil Red O staining

Oil Red O staining was performed as described previously32. In brief, Drosophila midguts were dissected in 1× PBS and fixed in 4% formaldehyde for 30 min. Midguts were washed three times in 1× PBS, double-distilled water and a 60% isopropanol solution. From the stock solution of Oil Red O (Sigma-Aldrich; 0.1% solution in isopropanol), a working solution was prepared by mixing 6 ml of 0.1% Oil Red O in isopropanol and 4 ml of double-distilled water. Midguts were incubated for 20 min in this solution and then washed in 60% isopropanol and water. The midguts were mounted in Vectashield mounting medium with DAPI (Vector Laboratories) and were imaged by confocal microscopy. Images were captured with the Zeiss LSM 510 confocal system and processed with LSM Image Browser and Adobe Photoshop.

Quantification and statistical analysis

To determine the percentage of GFP+ cells, the GFP+ cells and total cells were counted in a 5,000-μm2 area of a single confocal plane. In esgts samples, cells were counted in the posterior midgut and Malpighian tubules; in wgts samples, they were counted in the hindgut–midgut junction; and in esgtswgts samples, they were counted in the hindgut–midgut junction, the posterior midgut and the Malpighian tubules. The number of Pros+ nuclei was counted in a 0.08-mm2 surface area of a microscopic image from a similar region of each posterior midgut33. Cells per tumour were determined by counting the total number of nuclei within GFP+ tumours. All of the images were taken with the LSM5 Image Browser using the same confocal settings (Zeiss). Statistical analyses were performed using GraphPad Prism. Sample sizes (n) reported reflect the number of individual midguts. All experiments were performed in triplicate. P values were obtained between two groups using the Student’s t-test. For all statistical analysis, differences were considered to be statistically significant at values of P < 0.05.

Flow cytometry

Cell surface markers were analysed by flow cytometry. Cultured or treated cells were dissociated by 0.05% trypsin-EDTA and centrifuged. Single cells were resuspended with PBS containing 2% FBS and fluorescent-conjugated antibodies FITC-CD44 (clone G44-26, BD Biosciences) and PE-CD24 (clone ML5, Biolegend), and incubated on ice for 30 min. After washing three times with PBS containing 2% FBS, cells were resuspended with PBS containing 2% FBS and analysed by BD FACS Caliber (BD Biosciences).

Inhibitors

Arf1 inhibitors are BFA (brefeldin A)34 from Sigma, GCA (golgicide A)35 from Santa Cruz, secin H336 from Cayman chemical, LM1137 from A. Chavanieu, LG838 from L. Frigerio. 2-deoxy-d-Glucose (2-DG)39; JNK inhibitor SP 60012540; FAO inhibitors:triascin C41, etomoxir42, and mildronate43 from Cayman chemical, Enoximone44 from Tocris. Rac1 inhibitor from Santa Cruz. N-Acetyl-L-cysteine (NAC) 45 from Sigma. For control experiments, a DMSO control (100 μl in 10 ml food) was used. All inhibitors were mixed in the fly food with following concentrations; Arf1 inhibitors: BFA (50 ng ml−1 and 200 ng ml−1), GCA (5 μM), LM11 (50 μM), LG8 (100 μM), and secin H3 (50 μM); JNK inhibitor: Sp600125 (50 μM); Rac1 inhibitor (100 μM); ROS inhibitor: NAC (10 mM); FAO inhibitors: triacsin C (5 μM), mildronate (100 μM), etomoxir (100 μM), enoximone (100 μM); and glycolysis inhibitor 2-DG (50 mM). We mixed each inhibitor in fly food and tested different concentrations of these inhibitors. We used the concentration in which the inhibitors could kill tumour cells. At the beginning of experiments, to find out whether the flies would eat the inhibitors, we added green food dye to fly food. Flies were fasted for 1 h and then 8–10 flies were transferred to a vial containing coloured fly food mixed with inhibitors. We found that within 1 h of feeding the green dye could be seen through the abdomen of each fly, which suggest that the fly food mixed with inhibitors was edible to the flies. However, we excluded the food dye from the food used in the main experiments. For the main experiments, we fed flies with food containing inhibitors for 4 days. We repeated each inhibitor treatment three times.

Cell lines and culture

The human prostate cancer cell line DU145, colon cancer cell line HT29, and breast cancer cell lines MCF7, MDA-MB-231 (provided by the DCTD Tumour Repository) were cultured in RPMI1640 supplemented with 10% fetal bovine serum and 100 units per ml penicillin/streptomycin at 37 °C in a humidified atmosphere containing 5% CO2.

Cell survival assay

Cells were seeded at 2 × 105 cells per well in 6-well plates. Treatments with the indicated chemicals were started the next day, and cells were incubated for 2 more days. The surviving cells were stained with Crystal Violet (EMD Millipore). For quantification, the stained cells were solubilized in 1% SDS, and the absorbance at 595 nm was determined with a microplate reader.

Sphere formation assay

Single cells were cultured in a Corning Costar Ultra-Low attachment 24-well plate (Sigma-Aldrich) in sphere culture medium, consisting of DMEM/F12 (1:1), B27 (Invitrogen), and 20 ng ml−1 EGF (Invitrogen), with the indicated chemicals. The number of spheres was counted after 10 days of culture.

RNA isolation and real-time PCR

An RNeasy Mini Kit (Qiagen) was used to extract the total RNA from human cancer cells. The cDNA was synthesized from 1 μg RNA from each sample using a reverse transcription kit (Promega). Real-time PCR was performed in a 15-μl reaction system using SYBR Advantage qPCR Premix (Clontech). All of the reactions were performed in triplicate in a RealPlex 2 system (Eppendorf). The relative gene expression was quantified as described previously46. The sequence of each primer was as follows: ACTB, 5′-GATCATTGCTCCTCCTGAGC-3′ and 5′-ACTCCTGCTTGCTGATCCAC-3;CDH1, 5′-ACCAGAATAAAGACCAAGTGACCA-3′ and 5′-AGCAAGAGCAGCAGAATCAGAAT-3′; CD44, 5′-GAGCATCGGATTTGAGA-3′ and 5′-CATACTGGGAGGTGTTGG-3′.