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:

Adipocytes reprogram cancer cell metabolism by diverting glucose towards glycerol-3-phosphate thereby promoting metastasis

Abstract

In the tumor microenvironment, adipocytes function as an alternate fuel source for cancer cells. However, whether adipocytes influence macromolecular biosynthesis in cancer cells is unknown. Here we systematically characterized the bidirectional interaction between primary human adipocytes and ovarian cancer (OvCa) cells using multi-platform metabolomics, imaging mass spectrometry, isotope tracing and gene expression analysis. We report that, in OvCa cells co-cultured with adipocytes and in metastatic tumors, a part of the glucose from glycolysis is utilized for the biosynthesis of glycerol-3-phosphate (G3P). Normoxic HIF1α protein regulates the altered flow of glucose-derived carbons in cancer cells, resulting in increased glycerophospholipids and triacylglycerol synthesis. The knockdown of HIF1α or G3P acyltransferase 3 (a regulatory enzyme of glycerophospholipid synthesis) reduced metastasis in xenograft models of OvCa. In summary, we show that, in an adipose-rich tumor microenvironment, cancer cells generate G3P as a precursor for critical membrane and signaling components, thereby promoting metastasis. Targeting biosynthetic processes specific to adipose-rich tumor microenvironments might be an effective strategy against metastasis.

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: Adipocytes increase glucose metabolism and GPLs in OvCa cells.
Fig. 2: Adipocytes increase glycolysis but not oxidative phosphorylation in OvCa cells.
Fig. 3: GPAT3 regulates GPL synthesis.
Fig. 4: Adipocyte-mediated HIF1α expression regulates glucose utilization in cancer cells.
Fig. 5: Reduction of HIF1a expression inhibits omental metastasis.
Fig. 6: Cancer cells adjacent to adipocytes have increased PC.

Similar content being viewed by others

Data availability

Metabolomic data are deposited in the metabolomics workbench (https://www.metabolomicsworkbench.org/) and can be queried using the project https://doi.org/10.21228/M82S3K. Raw and processed microarray data are available in the Gene Expression Omnibus database (accession number GSE235641). Unique biological materials will be provided upon reasonable request. Source data are provided with this paper.

References

  1. Lengyel, E., Makowski, L., DiGiovanni, J. & Kolonin, M. G. Cancer as a matter of fat: the crosstalk between adipose tissue and tumors. Trends Cancer 4, 374–384 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Mukherjee, A., Bilecz, A. J. & Lengyel, E. The adipocyte microenvironment and cancer. Cancer Metastasis Rev. https://doi.org/10.1007/s10555-022-10059-x (2022).

    Article  PubMed  Google Scholar 

  3. Eckert, M. A. et al. Genomics of ovarian cancer progression reveals diverse metastatic trajectories including intraepithelial metastasis to the fallopian tube. Cancer Discov. 6, 1342–1351 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Nieman, K. M. et al. Adipocytes promote ovarian cancer metastasis and provide energy for rapid tumor growth. Nat. Med. 17, 1498–1503 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Mukherjee, A. et al. Adipocyte-induced FABP4 expression in ovarian cancer cells promotes metastasis and mediates carboplatin resistance. Cancer Res. 80, 1748–1761 (2020).

    Article  CAS  PubMed  Google Scholar 

  6. Ladanyi, A. et al. Adipocyte-induced CD36 expression drives ovarian cancer progression and metastasis. Oncogene 37, 2285–2301 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. 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 

  8. Fagone, P. & Jackowski, S. Membrane phospholipid synthesis and endoplasmic reticulum function. J. Lipid Res. 50, S311–S316 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

  9. Engelmann, B. & Wiedmann, M. K. Cellular phospholipid uptake: flexible paths to coregulate the functions of intracellular lipids. Biochim. Biophys. Acta 1801, 609–616 (2010).

    Article  CAS  PubMed  Google Scholar 

  10. Schug, Z. T. et al. Acetyl-CoA synthetase 2 promotes acetate utilization and maintains cancer cell growth under metabolic stress. Cancer Cell 27, 57–71 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Takeuchi, K. & Reue, K. Biochemistry, physiology, and genetics of GPAT, AGPAT, and lipin enzymes in triglyceride synthesis. Am. J. Physiol. Endocrinol. Metab. 296, E1195–E1209 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Buescher, J. M. et al. A roadmap for interpreting 13C metabolite labeling patterns from cells. Curr. Opin. Biotechnol. 34, 189–201 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Favaro, E. et al. Glucose utilization via glycogen phosphorylase sustains proliferation and prevents premature senescence in cancer cells. Cell Metab. 16, 751–764 (2012).

    Article  CAS  PubMed  Google Scholar 

  14. Chen, H, C. et al. Hypoxia induces a metabolic shift and enhances the stemness and expansion of cochlear spiral ganglion stem/progenitor cells. Biomed. Res. Int. 2015, 359537 (2015).

    PubMed  Google Scholar 

  15. Sumi, C. et al. Suppression of mitochondrial oxygen metabolism mediated by the transcription factor HIF-1 alleviates propofol-induced cell toxicity. Sci. Rep. 8, 8987 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  16. Laurent, V. et al. Periprostatic adipose tissue favors prostate cancer cell invasion in an obesity-dependent manner: role of oxidative stress. Mol. Cancer Res. 17, 821–835 (2019).

    Article  CAS  PubMed  Google Scholar 

  17. Hart, P. C. et al. Mesothelial cell HIF1α expression is metabolically downregulated by metformin to prevent oncogenic tumor–stromal crosstalk. Cell Rep. 29, 4086–4098 e4086 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Safran, M. et al. Mouse model for noninvasive imaging of HIF prolyl hydroxylase activity: assessment of an oral agent that stimulates erythropoietin production. Proc. Natl Acad. Sci. USA 103, 105–110 (2006).

    Article  CAS  PubMed  Google Scholar 

  19. Coscia, F. et al. Multi-level proteomics identifies CT45 as mediator of chemosensitivity and immunotherapy target in ovarian cancer. Cell 175, 159–170 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Williamson, J. R. et al. Hyperglycemic pseudohypoxia and diabetic complications. Diabetes 42, 801–813 (1993).

    Article  CAS  PubMed  Google Scholar 

  21. Walter, M., Liang, S., Ghosh, S., Hornsby, P. J. & Li, R. Interleukin 6 secreted from adipose stromal cells promotes migration and invasion of breast cancer cells. Oncogene 28, 2745–2755 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Lengyel, E. Ovarian cancer development and metastasis. Am. J. Pathol. 177, 1053–1064 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  23. Yang, W. S. & Stockwell, B. R. Ferroptosis: death by lipid peroxidation. Trends Cell Biol. 26, 165–176 (2016).

    Article  CAS  PubMed  Google Scholar 

  24. Yang, W. S. et al. Regulation of ferroptotic cancer cell death by GPX4. Cell 156, 317–331 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Dixon, S. J. et al. Ferroptosis: an iron-dependent form of nonapoptotic cell death. Cell 149, 1060–1072 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Lane, A. N., Yan, J. & Fan, T. W. 13C tracer studies of metabolism in mouse tumor xenografts. Bio Protoc. https://doi.org/10.21769/bioprotoc.1650 (2015).

  27. Scott, A. J. et al. Norharmane matrix enhances detection of endotoxin by MALDI–MS for simultaneous profiling of pathogen, host, and vector systems. Pathog. Dis. https://doi.org/10.1093/femspd/ftw097 (2016).

  28. Natsume, M. et al. Omental adipocytes promote peritoneal metastasis of gastric cancer through the CXCL2–VEGFA axis. Br. J. Cancer 123, 459–470 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Haverkamp, L., Brenkman, H. J., Ruurda, J. P., Ten Kate, F. J. & van Hillegersberg, R. The oncological value of omentectomy in gastrectomy for cancer. J. Gastrointest. Surg. 20, 885–890 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  30. Bristow, R. E., Tomacruz, R. S., Armstrong, D. K., Trimble, E. L. & Montz, F. J. Survival effect of maximal cytoreductive surgery for advanced ovarian carcinoma during the platinum era: a meta-analysis. J. Clin. Oncol. 20, 1248–1259 (2002).

    Article  PubMed  Google Scholar 

  31. Hosios, A. M. et al. mTORC1 regulates a lysosome-dependent adaptive shift in intracellular lipid species. Nat. Metab. https://doi.org/10.1038/s42255-022-00706-6 (2022).

    Article  PubMed  PubMed Central  Google Scholar 

  32. Szachowicz-Petelska, B. et al. Phospholipid composition and electric charge in healthy and cancerous parts of human kidneys. J. Membr. Biol. 246, 421–425 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Dobrzynska, I., Szachowicz-Petelska, B., Sulkowski, S. & Figaszewski, Z. Changes in electric charge and phospholipids composition in human colorectal cancer cells. Mol. Cell. Biochem. 276, 113–119 (2005).

    Article  CAS  PubMed  Google Scholar 

  34. Piccolis, M. et al. Probing the global cellular responses to lipotoxicity caused by saturated fatty acids. Mol. Cell 74, 32–44 e38 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Listenberger, L. L. et al. Triglyceride accumulation protects against fatty acid-induced lipotoxicity. Proc. Natl Acad. Sci. USA 100, 3077–3082 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Petschnigg, J. et al. Good fat, essential cellular requirements for triacylglycerol synthesis to maintain membrane homeostasis in yeast. J. Biol. Chem. 284, 30981–30993 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Barrera, G. Oxidative stress and lipid peroxidation products in cancer progression and therapy. ISRN Oncol. 2012, 137289 (2012).

    PubMed  PubMed Central  Google Scholar 

  38. Yang, M. et al. Clockophagy is a novel selective autophagy process favoring ferroptosis. Sci. Adv. 5, eaaw2238 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Zou, Y. et al. Plasticity of ether lipids promotes ferroptosis susceptibility and evasion. Nature 585, 603–608 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Mukherjee, A. Isolation of primary normal and cancer-associated adipocytes from the omentum. Mol. Biol. 2424, 167–175 (2022).

    CAS  Google Scholar 

  41. Mitra, A. K. et al. MicroRNAs reprogram normal fibroblasts into cancer-associated fibroblasts in ovarian cancer. Cancer Discov. 2, 1100–1108 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Sellers, K. et al. Pyruvate carboxylase is critical for non-small-cell lung cancer proliferation. J. Clin. Invest. 125, 687–698 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  43. Faubert, B. et al. Lactate metabolism in human lung tumors. Cell 171, 358–371 e359 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Kind, T. et al. Fiehnlib: mass spectral and retention index libraries for metabolomics based on quadrupole and time-of-flight gas chromatography/mass spectrometry. Anal. Chem. 81, 10038–10048 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Meissen, J. K. et al. Induced pluripotent stem cells show metabolomic differences to embryonic stem cells in polyunsaturated phosphatidylcholines and primary metabolism. PLoS ONE 7, e46770 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Olmstead, K. I. et al. Insulin induces a shift in lipid and primary carbon metabolites in a model of fasting-induced insulin resistance. Metabolomics https://doi.org/10.1007/s11306-017-1186-y (2017).

  47. Agrawal, K. et al. Effects of atopic dermatitis and gender on sebum lipid mediator and fatty acid profiles. Prostaglandins Leukot. Essent. Fat. Acids 134, 7–16 (2018).

    Article  CAS  Google Scholar 

  48. Tyanova, S. et al. The Perseus computational platform for comprehensive analysis of (prote)omics data. Nat. Methods 13, 731–740 (2016).

    Article  CAS  PubMed  Google Scholar 

  49. Tusher, V. G., Tibshirani, R. & Chu, G. Significance analysis of microarrays applied to the ionizing radiation response. Proc. Natl Acad. Sci. USA 98, 5116–5121 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Belov, M. E. et al. Design and performance of a novel interface for combined matrix-assisted laser desorption ionization at elevated pressure and electrospray ionization with orbitrap mass spectrometry. Anal. Chem. 89, 7493–7501 (2017).

    Article  CAS  PubMed  Google Scholar 

  51. He, L., Diedrich, J., Chu, Y. Y. & Yates, J. R. 3rd Extracting accurate precursor information for tandem mass spectra by RawConverter. Anal. Chem. 87, 11361–11367 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Greco, F. et al. Mass spectrometry imaging as a tool to investigate region specific lipid alterations in symptomatic human carotid atherosclerotic plaques. Metabolites https://doi.org/10.3390/metabo11040250 (2021).

  53. Smith, C. A. et al. METLIN: a metabolite mass spectral database. Ther. Drug Monit. 27, 747–751 (2005).

    Article  CAS  PubMed  Google Scholar 

  54. Liebisch, G. et al. Shorthand notation for lipid structures derived from mass spectrometry. J. Lipid Res. 54, 1523–1530 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Curtis, M. et al. Fibroblasts mobilize tumor cell glycogen to promote proliferation and metastasis. Cell Metab. 29, 141–155 e149 (2019).

    Article  CAS  PubMed  Google Scholar 

  56. Kamburov, A., Cavill, R., Ebbels, T. M., Herwig, R. & Keun, H. C. Integrated pathway-level analysis of transcriptomics and metabolomics data with IMPaLA. Bioinformatics 27, 2917–2918 (2011).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank all patients at The University of Chicago for donating tissue to this study. We thank F. Coscia and M. Mann (Max Plank Institute of Biochemistry, Martinsried, Bavaria) for help with proteomic analysis, and J. Andrade at the Center for Research Informatics (University of Chicago) for assistance with microarray analysis. This work was supported by the Ann Sol Schreiber award (372898), Colleen’s dream foundation and the DOD pilot award (W81XWH2110376) to A.M., NIH grants (R01CA169604, R35CA264619) awarded to E.L. D.B. is supported by NIH grant F31CA239330. R.J.D. is supported by grants from NIH (R35CA22044901) and by the Howard Hughes Medical Institute. We thank G. Isenberg for editing the paper. Figure 2b and Supplementary Figs. 2e,f, 5f and 6a,f were created using BioRender.com.

Author information

Authors and Affiliations

Authors

Contributions

A.M. and E.L. supervised the study, and wrote and edited the paper. Data acquisition and analysis were carried out by all A.M., D.B., F.G., C.-Y.C., M.Z., T.S., H.S.V., B.F., M.T., J.F., D.G., M.R.L.F., J.W.N., O.F., R.J.D. and E.L. Adipocyte isolation, co-culture, Seahorse, gene and protein expression analysis (A.M. and C.-Y.C.), metabolomics (A.M., J.F., M.R.L.F., D.G. and T.S.), omics data integrations (D.G. and A.M.), IMS (F.G., A.M., L.A.M. and L.N.), [13C]-glucose isotope tracing (A.M., D.B., H.S.V., B.F. and R.J.D.), xenograft studies (A.M.), animal studies (A.M., M.Z. and M.T.) and proteomics (A.M.). All authors edited and approved the final paper.

Corresponding author

Correspondence to Ernst Lengyel.

Ethics declarations

Competing interests

There are no potential conflicts of interest. E.L. received funding from Abbvie and Arsenal Bioscience for preclinical research studies unrelated to the submitted paper.

Peer review

Peer review information

Nature Metabolism thanks Rugang Zhang and the other, anonymous, reviewers for their contribution to the peer review of this work. Alfredo Giménez-Cassina, in collaboration with the Nature Metabolism team.

Additional information

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

Extended data

Extended Data Fig. 1 Untargeted metabolomic analysis of adipocytes after co-culture with ovarian cancer cells.

(a) Network map showing altered levels (red = increased; green = decreased) of biochemically and structurally similar metabolites in adipocytes after co-culture with SKOV3ip1 OvCa cells for 18 h. Biochemically and structurally similar metabolites are clustered. Metabolites in the same biochemical pathway are connected by orange lines. Structurally similar metabolites are connected by blue lines. (b) Principle component analysis (PCA) showing lipidomic changes in SKOV3ip1 cells induced by conditioned media derived from patient matched adipocytes (Adi CM), fibroblasts (NOF CM) and preadipocytes (Pre-adi CM). (c) Venn diagram showing number of metabolites altered in adipocytes, cancer cells, and co-culture derived media (p < 0.05, two-way ANOVA, mixed model post hoc Tukey HSD). (d) Changes in metabolites in adipocytes, cancer cells, and co-culture derived media. C24-ceramide, C16-ceramide and 18:1 sphingosine were primarily secreted by adipocytes; endocannabinoids such as 2-Arachidonylglycerol and 2-Linoleoylglycerol were increased in co-culture conditions (p < 0.05, one-way ANOVA). (e) Changes in phosphatidylcholine levels in the secretome of cancer cells (blue), adipocytes (green), and cancer cells co-cultured with adipocytes (red). Data extracted from metabolomics described in Fig. 1c. Statistical significance was calculated using two-way ANOVA, * p < 0.05, ** p < 0.01, *** p < 0.001.

Source data

Extended Data Fig. 2 Joint gene-metabolite analysis of adipocytes co-cultured with cancer cells.

Gene expression analysis. Heat map showing clustering of (a) cancer cells +/− adipocyte co-culture, (b) adipocytes +/− co-culture with SKOV3ip1 cells for 12 h. Gene Set Enrichment Analysis (cell cycle) based on the gene expression (microarray) of (c) cancer cells +/− adipocyte co-culture and (d) adipocytes +/− SKOV3ip1 co-culture. (One-way ANOVA, FDR, q value < 0.05). Integration of gene expression and metabolites using IMPala, depicting changes in the (e) cancer cells +/− adipocyte co-culture and (f) adipocytes +/− co-culture with SKOV3ip1 cells. Genes and metabolites increased with co-culture are in red, those reduced are in green. Fold changes of significantly altered genes from microarray analysis are shown in a table on left (One-way ANOVA, FDR, q value < 0.05). (E, F Created with BioRender.com (2023)).

Extended Data Fig. 3 Seahorse and metabolomics of ovarian cancer cells co-cultured with adipocytes.

(a) Glycolytic (ECAR) profiles of SKOV3ip1 cells treated with adipocyte conditioned media (Adi CM) for 18 h measured using a Seahorse XFe96 analyzer (n = 3 independent experiments, mean +/− SEM are plotted as bar graphs, two-way ANOVA, ** p < 0.05). (b) ECAR profile of OVCAR5 cells treated with Adi CM for 18 h (n = 3 independent experiments). (cd), Ex-vivo culture of omental tumor explants. Stable isotope tracing using [13C]-glucose (data from Fig. 2c) for 24 h. Isotopologue prolife of (C) TCA cycle intermediates and (D) Lysophosphocholine (LPC) 18:1 for three patient tumors is plotted (n = 3 independent biological samples). Mean +/− SEM are plotted as bar graphs. (e) Stable isotope tracing. SKOV3ip1 cells were treated with either control or Adi CM for 6 h and glucose utilization traced using uniformly labeled [13C]-glucose. Bar graph depict contribution of 13C vs 12C carbon to the total pool of glycerol-3-phosphate under specific treatment conditions (n = 3 independent experiments). Mean +/− SEM are plotted as bar graphs. (p = 0.11, two-way ANOVA; and p = 0.001 unpaired T-test).

Source data

Extended Data Fig. 4 Knockdown of GPAT3 reduces metastatic tumor burden in mice.

Scrambled (Control shRNA) or GPAT3 targeting shRNA (GPAT3 shRNA) transduced, luciferase expressing, HeyA8 cancer cells were injected intraperitoneally into athymic nude mice and tumor burden visualized after 2 weeks using the IVIS spectrum in vivo imaging system. Images show end-point difference in tumor burden.

Extended Data Fig. 5 Adipocyte-induced HIF1α regulates glycolysis.

(a) Immunoblot of HIF1α in ovarian cancer cell lines co-cultured with Adi for 16 h (n = 3 independent experiments). (b) Immunoblot for HIF1α, showing stable knockdown of HIF1α in SKOV3ip1 cells (n = 3 independent experiments). (c) Principal component analysis using adipocyte co-cultured samples (16 h) stably expressing either control shRNA or HIF1α shRNA cells. (d) qPCR analysis to determine glycerol-3-phosphate dehydrogenase (GPD1) mRNA expression in stable HIF1α knockdown SKOV3ip1 cells after 12 h adipocyte co-culture. Mean +/− SEM are plotted as bar graphs. (n = 3 independent experiments, two-tailed t-test, ** p < 0.005). (e-f) SKOV3ip1 HIF1α knockdown cells were treated with Adi CM for 18 h, followed by Seahorse analysis to determine changes in ECAR (n = 3 independent experiments). (Mean +/− SEM, two-way ANOVA, *** p = 0.0005, **** p < 0.0001). g) Adi CM was fractioned into metabolite and non-metabolite fractions based on size (3 kd). SKOV3ip1 cells were treated with both fractions for 6 h and immunoblot carried out (n = 3 independent experiments). (h) Immunoblot for HIF1α after treatment of SKOV3i1p cells with 10 ng/ml of recombinant human cytokines (IL-6, IL-8, and MCP1) for 6 hr (n = 3 independent experiments. (i) Immunoblot of HIF1α expression in SKOV3ip1 cells treated with Adi CM (6 h) +/− neutralizing antibodies against human IL-6, IL-8, or MCP-1(n = 3 independent experiments. (j) SKOV3ip1 cells were pretreated with the STAT-3 inhibitor STATTIC (10 µM), the JAK2 inhibitor AZD-1480 (10 µM), or a MEK inhibitor, Trametinib (1 µM) for 30 min, followed by incubation with Adi CM for 6 h. Immunoblot of HIF1α expression (n = 3 independent experiments).

Source data

Extended Data Fig. 6 Adipocyte-induced HIF1α alters the lipidome of ovarian cancer cells.

Lipidomics. HIF1α shRNA or control shRNA transduced SKOV3ip1 cells were co-cultured with primary omental adipocytes for 18 h, and mass spectrometry performed. The heat map depicts fold changes of significantly altered lipids (two-tailed t-test, p value of <s0.05) with adipocyte co-culture and HIF1α knockdown. Lipids: Triacylglycerol, TG; Phosphatidylethanolamine, PE; Lysophosphatidylcholine, LPC; phosphatidylcholine, PC; carnitine, CAR; ceramide, Cer; fatty acid, FA; diacylglycerol, DG; phosphatidylglycerol, PG; phosphatidylinositol, PI; phosphatidylserine, PS; sphingomyelin, SM.

Source data

Extended Data Fig. 7 Effect of HIF1α knockdown on ovarian cancer cells.

(a, b) Explant assay. SKOV3ip1 cells transduced with either control shRNA or HIF1α shRNA were cultured with non-cancerous human omentum for (A) 18 h and (B) 72 h to measure cellular adherence capacity and proliferation, respectively (n = 3 biologically independent samples). Mean +/− SEM are plotted as bar graphs. (c) C11 Bodipy stained cells (control and HIF1α) were treated with adipocyte-derived conditioned media and mean fluorescent intensity measured after 24 h using Incucyte (data extracted from Fig. 5f). (n = 3 independent experiments, Mean +/− SEM are plotted as bar graphs, one-way ANOVA * p < 0.05, *** p = 0.0002, p < 0.0001.) (D-E) Lipid ROS measurements. HeyA8 cells transduced with either scrambled shRNA (Control shRNA) or GPAT3 shRNA was labelled with C11-Bodipy dye was treated adipocyte-derived conditioned media (Adi CM). (d) Images were taken every 2 h using Incucyte and total green fluorescence intensity plotted. (n = 3 independent experiments, Mean +/− SEM are plotted). (e) Bar graph depicts green fluorescence intensity measured at the 24 h time point. (n = 3 independent experiments, two-way ANOVA, ** p = 0.003, mean +/− SEM are plotted). (f) Immunohistochemistry for 4-HNE adducts in serial sections of xenograft omental tumors (from Fig. 3d). Staining intensities of the images (left) were quantified using ImageJ (right). (n = 3 independent experiments, two-tailed t-test, mean +/− SEM are plotted as bar graphs). (g) MTT assay to determine the viability of HeyA8 cells after treatment with the indicated compounds. (n = 3 independent experiments, two-way ANOVA, p < 0.05, mean +/− SEM are plotted as bar graphs).

Source data

Supplementary information

Reporting Summary

Supplementary Table 1

Significantly altered (two-tailed t-test, P < 0.05) metabolites in SKOV3ip1 cells treated with conditioned medium derived from patient-matched primary adipocytes, fibroblasts and pre-adipocytes.

Supplementary Table 2

Differentially expressed genes in both adipocyte and SKOV3i1p OvCa cells after co-culture.

Supplementary Table 3

Joint gene–metabolite analysis using IMPaLa.

Supplementary Table 4

Proteomics to determine the effect of HIF1α knockdown on adipocyte-induced protein changes.

Supplementary Table 5

Lipidome of cancer cells co-cultured with primary human adipocytes after HIF1α knockdown.

Supplementary Table 6

IMS.

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 Extended Data Fig./Table 1

Statistical source data.

Source Data Extended Data Fig./Table 3

Statistical source data.

Source Data Extended Data Fig./Table 5

Statistical source data.

Source Data Extended Data Fig./Table 6

Statistical source data.

Source Data Extended Data Fig./Table 7

Statistical source data.

Source Data Fig. 4

Unprocessed western blots.

Source Data Extended Data Fig./Table 5

Unprocessed western blots.

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Mukherjee, A., Bezwada, D., Greco, F. et al. Adipocytes reprogram cancer cell metabolism by diverting glucose towards glycerol-3-phosphate thereby promoting metastasis. Nat Metab 5, 1563–1577 (2023). https://doi.org/10.1038/s42255-023-00879-8

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s42255-023-00879-8

Search

Quick links

Nature Briefing: Cancer

Sign up for the Nature Briefing: Cancer newsletter — what matters in cancer research, free to your inbox weekly.

Get what matters in cancer research, free to your inbox weekly. Sign up for Nature Briefing: Cancer