Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

Cellular metabolic reprogramming controls sugar appetite in Drosophila

Abstract

Cellular metabolic reprogramming is an important mechanism by which cells rewire their metabolism to promote proliferation and cell growth. This process has been mostly studied in the context of tumorigenesis, but less is known about its relevance for nonpathological processes and how it affects whole-animal physiology. Here, we show that metabolic reprogramming in Drosophila female germline cells affects nutrient preferences of animals. Egg production depends on the upregulation of the activity of the pentose phosphate pathway in the germline, which also specifically increases the animal’s appetite for sugar, the key nutrient fuelling this metabolic pathway. We provide functional evidence that the germline alters sugar appetite by regulating the expression of the fat-body-secreted satiety factor Fit. Our findings demonstrate that the cellular metabolic program of a small set of cells is able to increase the animal’s preference for specific nutrients through inter-organ communication to promote specific metabolic and cellular outcomes.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: The germline undergoes a reprogramming of its carbohydrate metabolism, which is required for egg production.
Fig. 2: Dietary supply of sugars is required for egg production.
Fig. 3: Carbohydrate metabolism in a subset of germline cells modulates sucrose appetite.
Fig. 4: Germline-ablated females do not have increased available carbohydrates and are in a hyper-starved state.
Fig. 5: The activity of the PPP in the germline is required for egg production.
Fig. 6: PPP activity in the germline modulates sucrose appetite.
Fig. 7: The germline modulates sugar appetite by regulating the expression of the fat-body-secreted satiety peptide Fit.

Similar content being viewed by others

Data availability

The authors declare that the main data supporting the findings of this study are available within the article and its Supplementary Information files. The detailed genotypes of the animals used in this study are included in Supplementary Tables 1 and 2. Sequences of all oligonucleotides used in this study are included in Supplementary Table 4. The exonic DNA sequences of Hex-A and Pgd were retrieved from Ensembl genome browser 93 (https://www.ensembl.org/index.html) for synthesizing the probes for in situ hybridization. Source imaging data were deposited in figshare (https://doi.org/10.6084/m9.figshare.12600185.v2). Source data are provided with this paper.

References

  1. Pavlova, N. N. & Thompson, C. B. The emerging hallmarks of cancer metabolism. Cell Metab. 23, 27–47 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Vander Heiden, M. G., Cantley, L. C. & Thompson, C. B. Understanding the Warburg effect: the metabolic requirements of cell proliferation. Science 324, 1029–1033 (2009).

    Article  CAS  Google Scholar 

  3. Warburg, O. On the origin of cancer cells. Science 123, 309–314 (1956).

    Article  CAS  PubMed  Google Scholar 

  4. Warburg, O., Wind, F. & Negelein, E. Über den Stoffwechsel von Tumoren im Körper. Klin. Wochenschr. 5, 829–832 (1926).

    Article  CAS  Google Scholar 

  5. DeBerardinis, R. J. & Chandel, N. S. Fundamentals of cancer metabolism. Sci. Adv. 2, e1600200 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  6. Shyh-Chang, N., Daley, G. Q. & Cantley, L. C. Stem cell metabolism in tissue development and aging. Development 140, 2535–2547 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. DeBerardinis, R. J., Sayed, N., Ditsworth, D. & Thompson, C. B. Brick by brick: metabolism and tumor cell growth. Curr. Opin. Genet. Dev. 18, 54–61 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Heiden, M. G. V. & DeBerardinis, R. J. Understanding the intersections between metabolism and cancer biology. Cell 168, 657–669 (2017).

    Article  CAS  Google Scholar 

  9. Lunt, S. Y. & Vander Heiden, M. G. Aerobic glycolysis: meeting the metabolic requirements of cell proliferation. Annu. Rev. Cell Dev. Biol. 27, 441–464 (2011).

    Article  CAS  PubMed  Google Scholar 

  10. Stincone, A. et al. The return of metabolism: biochemistry and physiology of the pentose phosphate pathway. Biol. Rev. 90, 927–963 (2015).

    Article  PubMed  Google Scholar 

  11. Barton, L. J., LeBlanc, M. G. & Lehmann, R. Finding their way: themes in germ cell migration. Curr. Opin. Cell Biol. 42, 128–137 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Johnston, D. S. & Ahringer, J. Cell polarity in eggs and epithelia: parallels and diversity. Cell 141, 757–774 (2010).

    Article  CAS  Google Scholar 

  13. Lehmann, R. Germline stem cells: origin and destiny. Cell Stem Cell 10, 729–739 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Bastock, R. & Johnston, D. S. Drosophila oogenesis. Curr. Biol. 18, R1082–R1087 (2008).

    Article  CAS  PubMed  Google Scholar 

  15. de Cuevas, M., Lilly, M. & Spradling, A. Germline cyst formation in Drosophila. Annu. Rev. Genet. 31, 405–428 (1997).

    Article  PubMed  Google Scholar 

  16. McLaughlin, J. M. & Bratu, D. P. in Drosophila Oogenesis: Methods and Protocols (eds. Bratu, D. P. & McNeil, G. P.) 1–20 (Springer, 2015). https://doi.org/10.1007/978-1-4939-2851-4_1

  17. Slaidina, M. & Lehmann, R. Translational control in germline stem cell development. J. Cell Biol. 207, 13–21 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Sieber, M. H. & Spradling, A. C. The role of metabolic states in development and disease. Curr. Opin. Genet. Dev. 45, 58–68 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Cox, R. T. & Spradling, A. C. A Balbiani body and the fusome mediate mitochondrial inheritance during Drosophila oogenesis. Development 130, 1579–1590 (2003).

    Article  CAS  PubMed  Google Scholar 

  20. Dumollard, R., Duchen, M. & Carroll, J. Section I. The role of mithochondrial function in the oocyte and embryo. in Current Topics in Developmental Biology, Vol. 77 (ed., St. John, J. C.) 21–49 (Academic Press, 2007).

  21. Teixeira, F. K. et al. ATP synthase promotes germ cell differentiation independent of oxidative phosphorylation. Nat. Cell Biol. 17, 689–696 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Sieber, M. H., Thomsen, M. B. & Spradling, A. C. Electron transport chain remodeling by GSK3 during oogenesis connects nutrient state to reproduction. Cell 164, 420–432 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Carvalho-Santos, Z. & Ribeiro, C. Gonadal ecdysone titers are modulated by protein availability but do not impact protein appetite. J. Insect Physiol. 106, 30–35 (2018).

    Article  CAS  PubMed  Google Scholar 

  24. Drummond-Barbosa, D. & Spradling, A. C. Stem cells and their progeny respond to nutritional changes during Drosophila oogenesis. Dev. Biol. 231, 265–278 (2001).

    Article  CAS  PubMed  Google Scholar 

  25. Hsu, H. J. & Drummond-Barbosa, D. Insulin levels control female germline stem cell maintenance via the niche in Drosophila. Proc. Natl Acad. Sci. USA 106, 1117–1121 (2009).

    Article  CAS  PubMed  Google Scholar 

  26. Leitao-Goncalves, R. et al. Commensal bacteria and essential amino acids control food choice behavior and reproduction. PLoS Biol. 15, e2000862 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  27. Piper, M. D. et al. A holidic medium for Drosophila melanogaster. Nat. Methods 11, 100–105 (2014).

    Article  CAS  PubMed  Google Scholar 

  28. Søndergaard, L. et al. Nutritional response in a Drosophila yolk protein gene promoter. Mol. Gen. Genet. 248, 25–32 (1995).

    Article  PubMed  Google Scholar 

  29. Min, K. J., Hogan, M. F., Tatar, M. & O’Brien, D. M. Resource allocation to reproduction and soma in Drosophila: a stable isotope analysis of carbon from dietary sugar. J. Insect Physiol. 52, 763–770 (2006).

    Article  CAS  PubMed  Google Scholar 

  30. Eisenreich, W., Ettenhuber, C., Laupitz, R., Theus, C. & Bacher, A. Isotopolog perturbation techniques for metabolic networks: metabolic recycling of nutritional glucose in Drosophila melanogaster. Proc. Natl Acad. Sci. USA 101, 6764–6769 (2004).

    Article  CAS  PubMed  Google Scholar 

  31. Itskov, P. M. & Ribeiro, C. The dilemmas of the gourmet fly: the molecular and neuronal mechanisms of feeding and nutrient decision making in Drosophila. Front. Neurosci. 7, 12 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  32. Münch, D., Ezra-Nevo, G., Francisco, A. P., Tastekin, I. & Ribeiro, C. Nutrient homeostasis—translating internal states to behavior. Curr. Opin. Neurobiol. 60, 67–75 (2020).

    Article  PubMed  CAS  Google Scholar 

  33. Simpson, S. J. & Raubenheimer, D. The Nature of Nutrition (Princeton University Press, 2012).

  34. Simpson, S. J., Couteur, D. G. L. & Raubenheimer, D. Putting the balance back in diet. Cell 161, 18–23 (2015).

    Article  CAS  PubMed  Google Scholar 

  35. Corrales-Carvajal, V. M., Faisal, A. A. & Ribeiro, C. Internal states drive nutrient homeostasis by modulating exploration–exploitation trade-off. eLife 5, e19920 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  36. Itskov, P. M. et al. Automated monitoring and quantitative analysis of feeding behaviour in Drosophila. Nat. Commun. 5, 4560 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Ribeiro, C. & Dickson, B. J. Sex peptide receptor and neuronal TOR/S6K signaling modulate nutrient balancing in Drosophila. Curr. Biol. 20, 1000–1005 (2010).

    Article  CAS  PubMed  Google Scholar 

  38. Simpson, S. J., Sword, G. A., Lorch, P. D. & Couzin, I. D. Cannibal crickets on a forced march for protein and salt. Proc. Natl Acad. Sci. USA 103, 4152–4156 (2006).

    Article  CAS  PubMed  Google Scholar 

  39. Trumper, S. & Simpson, S. J. Regulation of salt intake by nymphs of Locusta migratoria. J. Insect Physiol. 39, 857–864 (1993).

    Article  CAS  Google Scholar 

  40. Walker, S. J., Corrales-Carvajal, V. M. & Ribeiro, C. Postmating circuitry modulates salt taste processing to increase reproductive output in Drosophila. Curr. Biol. 25, 2621–2630 (2015).

    Article  CAS  PubMed  Google Scholar 

  41. Coll, A. P., Farooqi, I. S. & O’Rahilly, S. The hormonal control of food intake. Cell 129, 251–262 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Droujinine, I. A. & Perrimon, N. Interorgan communication pathways in physiology: focus on Drosophila. Annu. Rev. Genet. 50, 539–570 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Friedman, J. M. & Halaas, J. L. Leptin and the regulation of body weight in mammals. Nature 395, 763–770 (1998).

    Article  CAS  PubMed  Google Scholar 

  44. Leopold, P. & Perrimon, N. Drosophila and the genetics of the internal milieu. Nature 450, 186–188 (2007).

    Article  CAS  PubMed  Google Scholar 

  45. Pool, A. H. & Scott, K. Feeding regulation in Drosophila. Curr. Opin. Neurobiol. 29, 57–63 (2014).

    Article  CAS  PubMed  Google Scholar 

  46. Williams, K. W. & Elmquist, J. K. From neuroanatomy to behavior: central integration of peripheral signals regulating feeding behavior. Nat. Neurosci. 15, 1350–1355 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Sieber, M. H. & Spradling, A. C. Steroid signaling establishes a female metabolic state and regulates SREBP to control oocyte lipid accumulation. Curr. Biol. 25, 993–1004 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Stryer, L. Biochemistry (W. H. Freeman and Company, 1995).

  49. Cavener, D. R. Genetics of male-specific glucose oxidase and the identification of other unusual hexose enzymes in Drosophila melanogaster. Biochem. Genet. 18, 929–937 (1980).

    Article  CAS  PubMed  Google Scholar 

  50. Chintapalli, V. R., Wang, J. & Dow, J. A. Using FlyAtlas to identify better Drosophila melanogaster models of human disease. Nat. Genet. 39, 715–720 (2007).

    Article  CAS  PubMed  Google Scholar 

  51. Giese, G. E., Nanda, S., Holdorf, A. D. & Walhout, A. J. M. Transcriptional regulation of metabolic flux: a Caenorhabditis elegans perspective. Curr. Opin. Syst. Biol. 15, 12–18 (2019).

    Article  Google Scholar 

  52. O’Brien, D. M., Min, K. J., Larsen, T. & Tatar, M. Use of stable isotopes to examine how dietary restriction extends Drosophila lifespan. Curr. Biol. 18, R155–R156 (2008).

    Article  PubMed  CAS  Google Scholar 

  53. Piper, M. D. et al. Matching dietary amino acid balance to the in silico-translated exome optimizes growth and reproduction without cost to lifespan. Cell Metab. 25, 610–621 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Steck, K. et al. Internal amino acid state modulates yeast taste neurons to support protein homeostasis in Drosophila. eLife 7, e31625 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  55. Ohlstein, B. & McKearin, D. Ectopic expression of the Drosophila Bam protein eliminates oogenic germline stem cells. Development 124, 3651–3662 (1997).

    CAS  PubMed  Google Scholar 

  56. Inagaki, H. K. et al. Visualizing neuromodulation in vivo: TANGO-mapping of dopamine signaling reveals appetite control of sugar sensing. Cell 148, 583–595 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Dethier, V. G. The Hungry Fly: A Physiological Study of the Behavior Associated With Feeding (Harvard University Press, 1976).

  58. Gvozdev, V. A., Gerasimova, T. I., Kogan, G. L. & Braslavskaya, O. Y. Role of the pentose phosphate pathway in metabolism of Drosophila melanogaster elucidated by mutations affecting glucose 6-phosphate and 6-phosphogluconate dehydrogenases. FEBS Lett. 64, 85–88 (1976).

    Article  CAS  PubMed  Google Scholar 

  59. Hughes, M. B. & Lucchesi, J. C. Genetic rescue of a lethal “null” activity allele of 6-phosphogluconate dehydrogenase in Drosophila melanogaster. Science 196, 1114–1115 (1977).

    Article  CAS  PubMed  Google Scholar 

  60. Malmanche, N. & Clark, D. V. Drosophila melanogaster Prat, a purine de novo synthesis gene, has a pleiotropic maternal-effect phenotype. Genetics 168, 2011–2023 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Staller, M. V. et al. Depleting gene activities in early Drosophila embryos with the “maternal-Gal4–shRNA” system. Genetics 193, 51–61 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. DeBerardinis, R. J. & Thompson, C. B. Cellular metabolism and disease: what do metabolic outliers teach us? Cell 148, 1132–1144 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Fujii, S. & Amrein, H. Genes expressed in the Drosophila head reveal a role for fat cells in sex-specific physiology. EMBO J. 21, 5353–5363 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Parisi, M. J. et al. Germline-dependent gene expression in distant non-gonadal somatic tissues of Drosophila. BMC Genom. 11, 346 (2010).

    Article  CAS  Google Scholar 

  65. Sun, J. et al. Drosophila FIT is a protein-specific satiety hormone essential for feeding control. Nat. Commun. 8, 14161 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. DeBerardinis, R. J., Lum, J. J., Hatzivassiliou, G. & Thompson, C. B. The biology of cancer: metabolic reprogramming fuels cell growth and proliferation. Cell Metab. 7, 11–20 (2008).

    Article  CAS  PubMed  Google Scholar 

  67. Warburg, O. & Christian, W. Optischer Nachweis der Hydrierung und Dehydrierung des Pyridins im Gärungs-Co-Ferment. Biochemische Z. 286, 81 (1936).

    CAS  Google Scholar 

  68. Warburg, O., Christian, W. & Griese, A. Wasserstoffübertragendes Co-Ferment, seine Zusammensetzung und Wirkungsweise. Biochemische Z. 282, 157–205 (1935).

    CAS  Google Scholar 

  69. Walker, S. J., Goldschmidt, D. & Ribeiro, C. Craving for the future: the brain as a nutritional prediction system. Curr. Opin. Insect Sci. 23, 96–103 (2017).

    Article  PubMed  Google Scholar 

  70. Song, Y. et al. Dynamic control of dNTP synthesis in early embryos. Dev. Cell 42, 301–308 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Djabrayan, N. J.-V. et al. Metabolic regulation of developmental cell cycles and zygotic transcription. Curr. Biol. 29, 1193–1198 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Liu, B., Winkler, F., Herde, M., Witte, C.-P. & Großhans, J. A link between deoxyribonucleotide metabolites and embryonic cell-cycle control. Curr. Biol. 29, 1187–1192 (2019).

    Article  CAS  PubMed  Google Scholar 

  73. Andermann, M. L. & Lowell, B. B. Toward a wiring diagram understanding of appetite control. Neuron 95, 757–778 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Hudry, B. et al. Sex differences in intestinal carbohydrate metabolism promote food intake and sperm maturation. Cell 178, 901–918 (2019).

    Article  CAS  Google Scholar 

  75. Goncalves, M. D., Hopkins, B. D. & Cantley, L. C. Dietary fat and sugar in promoting cancer development and progression. Annu. Rev. Cancer Biol. 3, 255–273 (2019).

    Article  Google Scholar 

  76. Goncalves, M. D. et al. High-fructose corn syrup enhances intestinal tumor growth in mice. Science 363, 1345–1349 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Hirabayashi, S., Baranski, T. J. & Cagan, R. L. Transformed Drosophila cells evade diet-mediated insulin resistance through Wingless signaling. Cell 154, 664–675 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Perkins, L. A. et al. The Transgenic RNAi Project at Harvard Medical School: resources and validation. Genetics 201, 843–852 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Morris, C. A., Benson, E. & White-Cooper, H. Determination of gene expression patterns using in situ hybridization to Drosophila testes. Nat. Protoc. 4, 1807–1819 (2009).

    Article  CAS  PubMed  Google Scholar 

  80. Tennessen, J. M., Barry, W. E., Cox, J. & Thummel, C. S. Methods for studying metabolism in Drosophila. Methods 68, 105–115 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Miyamoto, T., Slone, J., Song, X. & Amrein, H. A fructose receptor functions as a nutrient sensor in the Drosophila brain. Cell 151, 1113–1125 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank B. Dickson (Howard Hughes Medical Institute, Janelia Research Campus, USA), R. Neumüller (Institute of Molecular Biotechnology, Austria), D. McKearin (Howard Hughes Medical Institute, University of Minnesota, USA), M. Buszczak (University of Texas Southwestern Medical Center, USA), Y. Li (Institute of Biophysics, Chinese Academy of Sciences, China), R. Martinho (Instituto de Biomedicina, Universidade de Aveiro, Portugal) and P. Prudencio (Instituto de Medicina Molecular, Portugal) for providing fly stocks and reagents. Lines obtained from the Bloomington Drosophila Stock Center (NIH P40OD018537) were used in this study. We thank R. Martinho and P. Prudencio for help with experimental protocol optimization. We thank R. Martinho, C. Pereira, A. Gontijo, S. Walker, G. Ezra, D. Goldschmidt, P. Francisco, D. Münch and all members of the Behavior and Metabolism Laboratory for helpful discussions and comments on the manuscript and G. Costa for illustrations. We thank M. Anjos and N. Archer for technical assistance. This project was supported by Bial grant 279/16 and Portuguese Foundation for Science and Technology (FCT) postdoctoral fellowship SFRH/BPD/79325/2011 to Z.C.-S. Work by Z.C.-S. was also financed by national funds through the FCT in the framework of the financing of the Norma Transitória DL 57/2016. Work by R.C.-F. was financed by the FCT doctoral fellowship SFRH/BD/143862/2019. Work by I.T. was financed by Marie Skłodowska-Curie Actions postdoctoral fellowship MSCA-IF-EF-ST 867459. Research at the Centre for the Unknown is supported by the Champalimaud Foundation and the research infrastructure Congento, co-financed by Lisboa Regional Operational Programme (Lisboa2020), under the PORTUGAL 2020 Partnership Agreement, through the European Regional Development Fund (ERDF) and Fundação para a Ciência e Tecnologia (Portugal) under the project LISBOA-01-0145-FEDER-022170.

Author information

Authors and Affiliations

Authors

Contributions

C.R. and Z.C.-S. conceived and designed the experiments. Z.C.-S., R.C.-F., A.P.E. and C.B. performed the experiments. C.R., Z.C.-S., R.C.-F., A.P.E., I.T. and C.B. analysed the data. C.R. and Z.C.-S. wrote the paper. C.R. administered the project. C.R. acquired funding. All authors read and approved the final manuscript.

Corresponding authors

Correspondence to Zita Carvalho-Santos or Carlos Ribeiro.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Peer review information Primary Handling Editor: Christoph Schmitt.

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Extended data

Extended Data Fig. 1 Related to Figs. 1, 3, 5, and 6. Expression of shRNAs for genes encoding carbohydrate metabolism enzymes in the female germline leads to efficient mRNA knockdown.

Normalized Hex-A (a), Pgi (b), Pfk (c), PyK (d), and Rpi (e) mRNA levels in females fed on holidic medium lacking sucrose. The MTD-GAL4 was used to drive shRNAs in the germline and matching GFP knockdown lines were used as controls (light orange, controls; dark orange, females with PPP knocked down germlines). Grey circles represent independent measurements (n), and the line represents the mean and the error bars the s.e.m. Black filled circles represent the presence and open black circles the absence of a given dietary nutrient or transgene. Full genotypes can be found in Supplementary Tables 1 and 2. Statistical significance was tested using a One-sided unpaired t-test.

Source data

Extended Data Fig. 2 Related to Fig. 3. Germline-ablated virgin and mated females show a specific decrease in sugar appetite.

a, Virgin females were assayed for changes in nutrient choice using the flyPAD. Bars represent the difference in sucrose feeding of flies (light orange, controls; dark orange, germline-ablated) fed on holidic medium lacking sucrose (carb deprived) vs full holidic medium (full). Columns represent the mean and the error bars show 95% confidence interval. Black filled circles represent the presence and open black circles the absence of a transgene. The method used to calculate the plotted values is described in the Methods section. b, Sugar preference was assayed using the red and blue food choice assay of flies (light colours, controls; dark colours, germline-ablated) kept on full holidic medium (green) or medium lacking sucrose (orange). Colored circles in the plot represent sugar preference in single assays, and the line represents the median and error bars the interquartile range. The number of independent biological replicates is represented as n. a,b, Full genotypes can be found in Supplementary Tables 1 and 2. Statistical significance was tested using the Wilcoxon rank-sum test (a) or the Kruskal-Wallis test followed by Dunn’s multiple comparison test (b).

Source data

Extended Data Fig. 3 Related to Fig. 4. Germline-ablated females do not have increased levels of trehalose or fructose.

Trehalose (a) or fructose (b) measurements from the heads of females (light colours, controls; dark colours, germline-ablated) reared on full holidic medium (green) or holidic medium lacking sucrose (orange) normalized to protein concentrations. Grey circles represent independent measurements (n), and the line represents the mean and the error bars the s.e.m. Black filled circles represent the presence and open black circles the absence of a given dietary nutrient or transgene. Full genotypes can be found in Supplementary Tables 1 and 2. Statistical significance was tested using an ordinary one-way ANOVA followed by Sidak’s multiple comparisons test.

Source data

Extended Data Fig. 4 Related to Fig. 5. The activity of the pentose phosphate pathway in the germline is required for oogenesis.

Ovariole morphology revealed by immunostaining of ovaries from females in which Zw (a) or Pgd (b) was knocked down in the germline. The dashed line in the germaria (G) delimits the region where St1 egg chambers would be formed. 1B1: spectrosomes and fusomes, Phalloidin: actin, DAPI: DNA. Scale bars, 25 μm (Germarium) and 50 μm (Ovariole). Full genotypes can be found in Supplementary Tables 1 and 2. Data shown are representative of 2 independent experiments of at least 6 ovaries/condition.

Extended Data Fig. 5 Related to Fig. 6. Knockdown of Hex-A, Pgd, or Rpi in ovaries using a mid-oogenesis driver leads to changes in sugar appetite.

a-c, Females in which the germline was knockdown of PPP enzymes (dark orange) using a mid-oogenesis driver (matα4-GAL4) were assayed for an effect in nutrient choice using the flyPAD. Bars represent the difference in sucrose feeding of flies maintained on holidic medium lacking sucrose (carb deprived) vs full holidic medium (full). Genotype matched GFP knockdown lines were used as negative controls (light orange). Columns represent the mean and the error bars show 95% confidence interval. Black filled circles represent the presence and open black circles the absence of a given transgene. The raw data used to derive these plots as well as the number of individuals tested per condition is indicated in Supplementary Fig. 2. The method used to calculate the plotted values is described in the Methods section. Statistical significance was tested using the Wilcoxon rank-sum test. d, Ovariole morphology revealed by immunostaining of ovaries from females in which Pgd was knocked down in the germline using matα4-GAL4 and the corresponding negative control. The region where the germarium is localized within the ovariole is marked with a G. Phalloidin: actin, DAPI: DNA. Scale bars, 25 μm (Germarium) and 50 μm (Ovariole). Data shown are representative of 1 experiment of at least 6 ovaries/condition. a-d, Full genotypes can be found in Supplementary Tables 1 and 2.

Source data

Extended Data Fig. 6 Related to Fig. 6. PPP activity in the germline is required and sufficient for controlling sugar appetite.

a, Schematic depicting the model of how the knockdown of different PPP enzymes may lead to opposite metabolic and behavioral outcomes. Enzymes or metabolites depicted in red are knocked down or decreased while those represented in green accumulate. b, Circles represent the nº of eggs laid/female in single assays (n) and the line represents the mean. The MTD-GAL4 was used to drive shRNAs in the germline and GFP knockdown used as a negative control (light green). Black filled circles represent the presence and open black circles the absence of a transgene. Statistical significance was tested using a One-sided unpaired t-test. Ovariole morphology revealed by immunostaining of ovaries from females in which Rpi was knocked down in the germline (c) or were mutants for Pgd and Zw (d). The region where the germarium is localized within the ovariole is marked with a G. 1B1: spectrosomes and fusomes, Phalloidin: actin, DAPI: DNA. Scale bars, 25 μm (Germarium) and 50 μm (Ovariole). Full genotypes can be found in Supplementary Tables 1 and 2. Data shown are representative of 2 experiments of at least 6 ovaries/condition.

Source data

Extended Data Fig. 7 Females with severely affected germline induced by AA deprivation still strongly increase sucrose appetite upon sugar deprivation.

a, Wild type females were assayed for an effect in nutrient choice using the flyPAD after being fed on a complete holidic medium, a medium lacking sucrose or a medium lacking both AAs and sucrose. Bars represent the difference in sucrose feeding of flies maintained on holidic medium lacking sucrose or sucrose and AAs (deprived) vs full holidic medium (full) (orange vs grey). Columns represent the mean and the error bars show 95% confidence interval. Raw data used to generate these plots and the number of individuals tested/condition is indicated in (b). The method used to calculate the plotted values is described in the Methods section. The raw data in (b) is represented by boxes showing the median with upper/lower quartiles. The number of independent biological replicates is represented as n. a, b, Black filled circles represent the presence and open black circles the absence of a particular nutrient (green, full HM, orange, HM without sucrose; grey, HM without both AAs and sucrose). Full genotypes can be found in Supplementary Tables 1 and 2. Statistical significance was tested using the Wilcoxon rank-sum test (a) the One-sided Mann-Whitney test (b).

Source data

Extended Data Fig. 8 Related to Fig. 7. Carbohydrate metabolism in the germline controls the expression levels of fit.

a, Normalized fit mRNA levels in females fed on holidic medium lacking sucrose. The MTD-GAL4 was used to drive shRNA in the germline. A genotype matched GFP knockdown line was used as a control (light orange). b, Normalized fit mRNA levels in control females fed on a complete holidic medium (green) or a medium lacking sucrose (orange). a-b, Grey circles represent independent measurements (n), and the line represents the mean and the error bars the s.e.m. c, Females (light colours, controls; dark colours, females mutant for fit) were assayed for an effect in nutrient choice using the flyPAD after fed on a complete holidic medium (green), or one lacking sucrose (orange). Boxes represent median with upper/lower quartiles. The number of independent biological replicates is represented as n. a-c, Black filled circles represent the presence and open black circles the absence of a nutrient, transgene, the germline, or the fit gene. Full genotypes can be found in Supplementary Tables 1 and 2. Statistical significance was tested using the One-sided unpaired t-test (a-b) or the Kruskal-Wallis test followed by Dunn’s multiple comparison test (c).

Source data

Extended Data Fig. 9 The male germline also regulates carbohydrate appetite but not via the PPP.

Males in which the germline was ablated (nos-GAL4>UAS-bam) (a) or metabolically manipulated (MTD-GAL4>Zw shRNA, MTD-GAL4>Pgd shRNA) (b) were assayed for an effect in nutrient choice using the flyPAD. Bars represent the difference in sucrose feeding of flies (light orange, controls; dark orange, germline-ablated) maintained on holidic medium lacking sucrose (carb deprived) vs full holidic medium (full). Genotype matched GFP knockdown lines were used as negative controls in (b). Columns (light orange, controls; dark orange, males with PPP knocked down germlines) represent the mean and the error bars show 95% confidence interval. Black filled circles represent the presence and open black circles the absence of a transgene. The raw data used to derive these plots as well as the number of individuals tested per condition is indicated in Supplementary Fig. 3. The method used to calculate the plotted values is described in the Methods section. Full genotypes can be found in Supplementary Tables 1 and 2. Statistical significance was tested using the Wilcoxon rank-sum test.

Source data

Supplementary information

Supplementary Information

Supplementary Figs. 1–3 and Supplementary Tables 1–5.

Reporting Summary

Source data

Source Data Fig. 1

Statistical source data.

Source Data Fig. 2

Statistical source data.

Source Data Fig. 3

Statistical source data.

Source Data Fig. 4

Statistical source data.

Source Data Fig. 5

Statistical source data.

Source Data Fig. 6

Statistical source data.

Source Data Fig. 7

Statistical source data.

Source Data Extended Data Fig. 1

Statistical source data.

Source Data Extended Data Fig. 2

Statistical source data.

Source Data Extended Data Fig. 3

Statistical source data.

Source Data Extended Data Fig. 5

Statistical source data.

Source Data Extended Data Fig. 6

Statistical source data.

Source Data Extended Data Fig. 7

Statistical source data.

Source Data Extended Data Fig. 8

Statistical source data.

Source Data Extended Data Fig. 9

Statistical source data.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Carvalho-Santos, Z., Cardoso-Figueiredo, R., Elias, A.P. et al. Cellular metabolic reprogramming controls sugar appetite in Drosophila. Nat Metab 2, 958–973 (2020). https://doi.org/10.1038/s42255-020-0266-x

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s42255-020-0266-x

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing