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:

A palmitate-rich metastatic niche enables metastasis growth via p65 acetylation resulting in pro-metastatic NF-κB signaling

Abstract

Metabolic rewiring is often considered an adaptive pressure limiting metastasis formation; however, some nutrients available at distant organs may inherently promote metastatic growth. We find that the lung and liver are lipid-rich environments. Moreover, we observe that pre-metastatic niche formation increases palmitate availability only in the lung, whereas a high-fat diet increases it in both organs. In line with this, targeting palmitate processing inhibits breast cancer-derived lung metastasis formation. Mechanistically, breast cancer cells use palmitate to synthesize acetyl-CoA in a carnitine palmitoyltransferase 1a-dependent manner. Concomitantly, lysine acetyltransferase 2a expression is promoted by palmitate, linking the available acetyl-CoA to the acetylation of the nuclear factor-kappaB subunit p65. Deletion of lysine acetyltransferase 2a or carnitine palmitoyltransferase 1a reduces metastasis formation in lean and high-fat diet mice, and lung and liver metastases from patients with breast cancer show coexpression of both proteins. In conclusion, palmitate-rich environments foster metastases growth by increasing p65 acetylation, resulting in a pro-metastatic nuclear factor-kappaB signaling.

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: High-fat diet enhances overall fatty acid availability in the lung and liver, whereas palmitate availability is specifically increased in the lung during pre-metastatic niche formation.
Fig. 2: Lung-resident alveolar type II cells increase surfactant-related gene expression and palmitate release during pre-metastatic niche formation.
Fig. 3: Intracellular palmitate levels and CPT1a expression are increased in breast cancer spheroids and lung metastases.
Fig. 4: Silencing CPT1a counteracts palmitate-induced spheroid growth and inhibits metastasis formation in lean and HFD mice.
Fig. 5: CPT1a activity sustains acetyl-CoA levels in spheroids and lung metastases.
Fig. 6: CPT1a is required for p65 acetylation and NF-κB signaling.
Fig. 7: CPT1a and KAT2a are coexpressed in palmitate-enriched environments and their silencing impairs metastasis formation.

Similar content being viewed by others

Data availability

Mouse scRNA-seq, RNA-seq and ChIP-seq data generated in this study have been deposited in the GEO under accession code GSE196993. The human expression data from patients with primary breast cancer from the publicly available TCGA Research Network and METABRIC are available in TCGA (http://tumorsurvival.org/download.html) and cBioportal (https://www.cbioportal.org/datasets). The dataset for gene expression from breast cancer metastases at different organ sites was downloaded from GEO under accession code GSE14018. Source data and gel source images are provided with this paper. All other data supporting the findings of this study are available within the Article and the Supplementary Information and from the corresponding author on reasonable request. Source data are provided with this paper.

References

  1. Bergers, G. & Fendt, S. M. The metabolism of cancer cells during metastasis. Nat. Rev. Cancer 21, 162–180 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Martin-Perez, M., Urdiroz-Urricelqui, U., Bigas, C. & Benitah, S. A. The role of lipids in cancer progression and metastasis. Cell Metab. 34, 1675–1699 (2022).

    Article  CAS  PubMed  Google Scholar 

  3. Broadfield, L. A., Pane, A. A., Talebi, A., Swinnen, J. V. & Fendt, S. M. Lipid metabolism in cancer: new perspectives and emerging mechanisms. Dev. Cell 56, 1363–1393 (2021).

    Article  CAS  PubMed  Google Scholar 

  4. Altea-Manzano, P., Cuadros, A. M., Broadfield, L. A. & Fendt, S. M. Nutrient metabolism and cancer in the in vivo context: a metabolic game of give and take. EMBO Rep. 21, e50635 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Peinado, H. et al. Pre-metastatic niches: organ-specific homes for metastases. Nat. Rev. Cancer 17, 302–317 (2017).

    Article  CAS  PubMed  Google Scholar 

  6. Doglioni, G., Parik, S. & Fendt, S. M. Interactions in the (pre)metastatic niche support metastasis formation. Front. Oncol. 9, 219 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  7. Fong, M. Y. et al. Breast-cancer-secreted miR-122 reprograms glucose metabolism in premetastatic niche to promote metastasis. Nat. Cell Biol. 17, 183–194 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Christen, S. et al. Breast cancer-derived lung metastasis show increased pyruvate carboxylase-dependent anaplerosis. Cell Rep. 17, 837–848 (2016).

    Article  CAS  PubMed  Google Scholar 

  9. Rinaldi, G. et al. In vivo evidence for serine biosynthesis-defined sensitivity of lung metastasis, but not of primary breast tumors, to mTORC1 inhibition. Mol. Cell 81, 386–397 (2021).

    Article  CAS  PubMed  Google Scholar 

  10. Heppner, G., Miller, F. & Malathy Shekhar, P. V. Nontransgenic models of breast cancer. Breast Cancer Res. 2, 331–334 (2000).

    Article  CAS  PubMed  Google Scholar 

  11. Lee, E., Pandey, N. B. & Popel, A. S. Pre-treatment of mice with tumor-conditioned media accelerates metastasis to lymph nodes and lungs: a new spontaneous breast cancer metastasis model. Clin. Exp. Metastasis 31, 67–79 (2014).

    Article  PubMed  Google Scholar 

  12. Erler, J. T. et al. Hypoxia-Induced lysyl oxidase is a critical mediator of bone marrow cell recruitment to form the premetastatic niche. Cancer Cell 15, 35–44 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Sceneay, J. et al. Primary tumor hypoxia recruits CD11b+/Ly6Cmed/Ly6G+ immune suppressor cells and compromises NK cell cytotoxicity in the premetastatic niche. Cancer Res. 72, 3906 (2012).

    Article  CAS  PubMed  Google Scholar 

  14. Peinado, H. et al. Melanoma exosomes educate bone marrow progenitor cells toward a pro-metastatic phenotype through MET. Nat. Med. 18, 883–891 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Agudelo, C. W., Samaha, G. & Garcia-Arcos, I. Alveolar lipids in pulmonary disease: a review. Lipids Health Dis. 19, 122 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Ombrato, L. et al. Metastatic-niche labelling reveals parenchymal cells with stem features. Nature 572, 603–608 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Hegab, A. E. et al. High-fat diet activates adult mouse lung stem cells and accelerates several aging-induced effects. Stem Cell Res. 33, 25–35 (2018).

    Article  CAS  PubMed  Google Scholar 

  18. Elia, I. et al. Proline metabolism supports metastasis formation and could be inhibited to selectively target metastasizing cancer cells. Nat. Commun. 8, 15267 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  19. Elia, I. et al. Breast cancer cells rely on environmental pyruvate to shape the metastatic niche. Nature 568, 117–121 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Lien, E. C. et al. Low glycaemic diets alter lipid metabolism to influence tumour growth. Nature 599, 302–307 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Vivas-García, Y. et al. Lineage-restricted regulation of SCD and fatty acid saturation by MITF controls melanoma phenotypic plasticity. Mol. Cell 77, 120–137 (2020).

    Article  PubMed  Google Scholar 

  22. Broadfield, L. A. et al. Fat induces glucose metabolism in nontransformed liver cells and promotes liver tumorigenesis. Cancer Res. 81, 1988–2001 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Ciriello, G. et al. Comprehensive molecular portraits of invasive lobular breast cancer. Cell 163, 506–519 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Curtis, C. et al. The genomic and transcriptomic architecture of 2,000 breast tumours reveals novel subgroups. Nature 486, 346–352 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Kalucka, J. et al. Quiescent endothelial cells upregulate fatty acid β-oxidation for vasculoprotection via redox homeostasis. Cell Metab. 28, 881–894 (2018).

    Article  CAS  PubMed  Google Scholar 

  26. Campbell, S. L. & Wellen, K. E. Metabolic signaling to the nucleus in cancer. Mol. Cell 71, 398–408 (2018).

    Article  CAS  PubMed  Google Scholar 

  27. Choudhary, C., Weinert, B. T., Nishida, Y., Verdin, E. & Mann, M. The growing landscape of lysine acetylation links metabolism and cell signalling. Nat. Rev. Mol. Cell Biol. 15, 536–550 (2014).

    Article  CAS  PubMed  Google Scholar 

  28. Krämer, A., Green, J., Pollard, J. Jr. & Tugendreich, S. Causal analysis approaches in ingenuity pathway analysis. Bioinformatics 30, 523–530 (2014).

    Article  PubMed  Google Scholar 

  29. Huber, M. A. et al. NF-κB is essential for epithelial-mesenchymal transition and metastasis in a model of breast cancer progression. J. Clin. Invest. 114, 569–581 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Pino, A. M. & Rodríguez, J. P. Is fatty acid composition of human bone marrow significant to bone health. Bone 118, 53–61 (2019).

    Article  CAS  PubMed  Google Scholar 

  31. Ferraro, G. B. et al. Fatty acid synthesis is required for breast cancer brain metastasis. Nat. Cancer 2, 414–428 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Parik, S. et al. GBM tumors are heterogeneous in their fatty acid metabolism and modulating fatty acid metabolism sensitizes cancer cells derived from recurring GBM tumors to temozolomide. Front. Oncol. https://doi.org/10.3389/fonc.2022.988872 (2022).

  33. Calao, M., Burny, A., Quivy, V., Dekoninck, A. & Van Lint, C. A pervasive role of histone acetyltransferases and deacetylases in an NF-kB-signaling code. Trends Biochem. Sci. 33, 339–349 (2008).

    Article  CAS  PubMed  Google Scholar 

  34. Qin, J. D. et al. Effect of ammonium pyrrolidine dithiocarbamate (PDTC) on NF-κB activation and CYP2E1 content of rats with immunological liver injury. Pharm. Biol. 52, 1460–1466 (2014).

    Article  CAS  PubMed  Google Scholar 

  35. Haque, M. E. et al. The GCN5: its biological functions and therapeutic potentials. Clin. Sci. 135, 231–257 (2021).

    Article  Google Scholar 

  36. Bondy-Chorney, E., Denoncourt, A., Sai, Y. & Downey, M. Nonhistone targets of KAT2A and KAT2B implicated in cancer biology (1). Biochem. Cell Biol. 97, 30–45 (2019).

    Article  CAS  PubMed  Google Scholar 

  37. Stilling, R. M. et al. K-lysine acetyltransferase 2a regulates a hippocampal gene expression network linked to memory formation. EMBO J. 33, 1912–1927 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Chimenti, F. et al. A novel histone acetyltransferase inhibitor modulating Gcn5 network: cyclopentylidene-[4-(4′-chlorophenyl)thiazol-2-yl)hydrazone. J. Med. Chem. 52, 530–536 (2009).

  39. Ubellacker, J.M. et al. Lymph protects metastasizing melanoma cells from ferroptosis. Nature 585, 113–118 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Pascual, G. et al. Dietary palmitic acid promotes a prometastatic memory via Schwann cells. Nature 599, 485–490 (2021).

    Article  CAS  PubMed  Google Scholar 

  41. Cacicedo, J. M., Yagihashi, N., Keaney, J. F. Jr., Ruderman, N. B. & Ido, Y. AMPK inhibits fatty acid-induced increases in NF-κB transactivation in cultured human umbilical vein endothelial cells. Biochem. Biophys. Res. Commun. 324, 1204–1209 (2004).

    Article  CAS  PubMed  Google Scholar 

  42. Ajuwon, K. M. & Spurlock, M. E. Palmitate activates the NF-κB transcription factor and induces IL-6 and TNFα expression in 3T3-L1 adipocytes. J. Nutr. 135, 1841–1846 (2005).

    Article  CAS  PubMed  Google Scholar 

  43. Laine, P. S. et al. Palmitic acid induces IP-10 expression in human macrophages via NF-κB activation. Biochem. Biophys. Res. Commun. 358, 150–155 (2007).

    Article  CAS  PubMed  Google Scholar 

  44. Maloney, E. et al. Activation of NF-κB by palmitate in endothelial cells: a key role for NADPH oxidase-derived superoxide in response to TLR4 activation. Arterioscler. Thromb. Vasc. Biol. 29, 1370–1375 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Khandekar, M. J., Cohen, P. & Spiegelman, B. M. Molecular mechanisms of cancer development in obesity. Nat. Rev. Cancer 11, 886–895 (2011).

    Article  CAS  PubMed  Google Scholar 

  46. Wang, X. et al. Epigenetic silencing of miR-33b promotes peritoneal metastases of ovarian cancer by modulating the TAK1/FASN/CPT1A/NF-κB axis. Cancers 13, 4795 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Rios Garcia, M. et al. Acetyl-CoA carboxylase 1-dependent protein acetylation controls breast cancer metastasis and recurrence. Cell Metab. 26, 842–855 (2017).

    Article  CAS  PubMed  Google Scholar 

  48. Loo, S. Y. et al. Fatty acid oxidation is a druggable gateway regulating cellular plasticity for driving metastasis in breast cancer. Sci. Adv. 7, eabh2443 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. McDonnell, E. et al. Lipids reprogram metabolism to become a major carbon source for histone acetylation. Cell Rep. 17, 1463–1472 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Lu, M. et al. ACOT12-dependent alteration of acetyl-CoA drives hepatocellular carcinoma metastasis by epigenetic induction of epithelial-mesenchymal transition. Cell Metab. 29, 886–900 (2019).

    Article  CAS  PubMed  Google Scholar 

  51. Chen, Q. et al. Acetyl-CoA derived from hepatic mitochondrial fatty acid β-oxidation aggravates inflammation by enhancing p65 acetylation. iScience 24, 103244 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Mao, X. et al. GCN5 is a required cofactor for a ubiquitin ligase that targets NF-κB/RelA. Genes Dev. 23, 849–861 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Xiong, Y. et al. CPT1A regulates breast cancer-associated lymphangiogenesis via VEGF signaling. Biomed. Pharmacother. 106, 1–7 (2018).

    Article  CAS  PubMed  Google Scholar 

  54. Jariwala, N. et al. CPT1A and fatty acid β-oxidation are essential for tumor cell growth and survival in hormone receptor-positive breast cancer. NAR Cancer 3, zcab035 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  55. Pucci, S. et al. Carnitine palmitoyl transferase-1A (CPT1A): a new tumor specific target in human breast cancer. Oncotarget 7, 19982–19996 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  56. Park, JunH. et al. Fatty acid oxidation-driven Src links mitochondrial energy reprogramming and oncogenic properties in triple-negative breast cancer. Cell Rep. 14, 2154–2165 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Wang, Y. N. et al. CPT1A-mediated fatty acid oxidation promotes colorectal cancer cell metastasis by inhibiting anoikis. Oncogene 37, 6025–6040 (2018).

    Article  CAS  PubMed  Google Scholar 

  58. Sawyer, B. T. et al. Targeting fatty acid oxidation to promote anoikis and inhibit ovarian cancer progression. Mol. Cancer Res. 18, 1088 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Lee, C.-k et al. Tumor metastasis to lymph nodes requires YAP-dependent metabolic adaptation. Science 363, 644 (2019).

    Article  CAS  PubMed  Google Scholar 

  60. Barone, I., Giordano, C., Bonofiglio, D., Andò, S. & Catalano, S. The weight of obesity in breast cancer progression and metastasis: clinical and molecular perspectives. Semin. Cancer Biol. 60, 274–284 (2020).

    Article  CAS  PubMed  Google Scholar 

  61. von Lintig, F. C. et al. Ras activation in human breast cancer. Breast Cancer Res. Treat. 62, 51–62 (2000).

    Article  Google Scholar 

  62. Hoenerhoff, M. J. et al. BMI1 cooperates with H-RAS to induce an aggressive breast cancer phenotype with brain metastases. Oncogene 28, 3022–3032 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Lock, R., Kenific, C. M., Leidal, A. M., Salas, E. & Debnath, J. Autophagy-dependent production of secreted factors facilitates oncogenic RAS-driven invasion. Cancer Discov. 4, 466–479 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Leamy, A. K. et al. Enhanced synthesis of saturated phospholipids is associated with ER stress and lipotoxicity in palmitate-treated hepatic cells. J. Lipid Res. 55, 1478–1488 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Czerkies, M. et al. Cell fate in antiviral response arises in the crosstalk of IRF, NF-κB and JAK/STAT pathways. Nat. Commun. 9, 493 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  66. Jacque, E., Tchenio, T., Piton, G., Romeo, P.-H. & Baud, V. RelA repression of RelB activity induces selective gene activation downstream of TNF receptors. Proc. Natl Acad. Sci. 102, 14635–14640 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Eluard, B. et al. The alternative RelB NF-κB subunit is a novel critical player in diffuse large B-cell lymphoma. Blood 139, 384–398 (2022).

    Article  CAS  PubMed  Google Scholar 

  68. van Gorsel, M., Elia, I. & Fendt, S.-M. 13C tracer analysis and metabolomics in 3D cultured cancer cells. Methods Mol. Biol. https://doi.org/10.1007/978-1-4939-8769-6_4 (2019).

  69. Lorendeau, D. et al. Dual loss of succinate dehydrogenase (SDH) and complex I activity is necessary to recapitulate the metabolic phenotype of SDH mutant tumors. Metab. Eng. 43, 187–197 (2017).

    Article  CAS  PubMed  Google Scholar 

  70. Vriens, K. et al. Evidence for an alternative fatty acid desaturation pathway increasing cancer plasticity. Nature 566, 403–406 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  71. Major, J. et al. Type I and III interferons disrupt lung epithelial repair during recovery from viral infection. Science 369, 712–717 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Fernandez, C. A., Des Rosiers, C., Previs, S. F., David, F. & Brunengraber, H. Correction of 13C mass isotopomer distributions for natural stable isotope abundance. J. Mass Spectrom. 31, 255–262 (1996).

    Article  CAS  PubMed  Google Scholar 

  73. 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 

  74. Karch, K. R., Sidoli, S. & Garcia, B. A. Identification and quantification of histone PTMs using high-resolution mass spectrometry. Methods Enzymol. 574, 3–29 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Rappsilber, J., Ishihama, Y. & Mann, M. Stop and go extraction tips for matrix-assisted laser desorption/ionization, nanoelectrospray, and LC/MS sample pretreatment in proteomics. Anal. Chem. 75, 663–670 (2003).

    Article  CAS  PubMed  Google Scholar 

  76. Broux, M. et al. Suz12 inactivation cooperates with JAK3 mutant signaling in the development of T-cell acute lymphoblastic leukemia. Blood 134, 1323–1336 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Sinha, M. & Lowell, C. A. Isolation of highly pure primary mouse alveolar epithelial type II cells by flow cytometric cell sorting. Bio. Protoc. 6, e2013 (2016).

    Article  PubMed  Google Scholar 

  78. Barkauskas, C. E. et al. Type 2 alveolar cells are stem cells in adult lung. J. Clin. Invest. 123, 3025–3036 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Butler, A., Hoffman, P., Smibert, P., Papalexi, E. & Satija, R. Integrating single-cell transcriptomic data across different conditions, technologies, and species. Nat. Biotechnol. 36, 411–420 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. Cao, J. et al. The single-cell transcriptional landscape of mammalian organogenesis. Nature 566, 496–502 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Blondel, V. D., Guillaume, J.-L., Lambiotte, R. & Lefebvre, E. Fast unfolding of communities in large networks. J. Stat. Mech. 2008, P10008 (2008).

    Article  Google Scholar 

  82. Hänzelmann, S., Castelo, R. & Guinney, J. GSVA: gene set variation analysis for microarray and RNA-seq data. BMC Bioinform. 14, 7 (2013).

    Article  Google Scholar 

  83. Kim, D. et al. TopHat2: accurate alignment of transcriptomes in the presence of insertions, deletions and gene fusions. Genome Biol. 14, R36 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  84. Langmead, B. & Salzberg, S. L. Fast gapped-read alignment with Bowtie 2. Nat. Methods 9, 357–359 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  85. Hruz, T. et al. Genevestigator v3: a reference expression database for the meta-analysis of transcriptomes. Adv. Bioinform. https://doi.org/10.1155/2008/420747 (2008).

  86. Xu, J. et al. 14-3-3ζ turns TGF-β’s function from tumor suppressor to metastasis promoter in breast cancer by contextual changes of Smad partners from p53 to Gli2. Cancer Cell 27, 177–192 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  87. Liberzon, A. et al. The Molecular Signatures Database hallmark gene set collection. Cell Syst. 1, 417–425 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  88. Gui, J. et al. Activation of p38α stress-activated protein kinase drives the formation of the pre-metastatic niche in the lungs. Nat. Cancer 1, 603–619 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  89. Liu, Y. et al. Tumor exosomal RNAs promote lung pre-metastatic niche formation by activating alveolar epithelial tLR3 to recruit neutrophils. Cancer Cell 30, 243–256 (2016).

    Article  PubMed  Google Scholar 

  90. Hiratsuka, S. et al. The S100A8–serum amyloid A3–TLR4 paracrine cascade establishes a pre-metastatic phase. Nat. Cell Biol. 10, 1349–1355 (2008).

    Article  CAS  PubMed  Google Scholar 

  91. Morrissey, S. M. et al. Tumor-derived exosomes drive immunosuppressive macrophages in a pre-metastatic niche through glycolytic dominant metabolic reprogramming. Cell Metab. 33, 2040–2058 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  92. Hoshino, A. et al. Tumour exosome integrins determine organotropic metastasis. Nature 527, 329–335 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  93. Geiger, T. et al. Use of stable isotope labeling by amino acids in cell culture as a spike-in standard in quantitative proteomics. Nat. Protoc. 6, 147–157 (2011).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank P. Carmeliet (VIB-KU Leuven) for providing shRNA against Cpt1a, D. Nittner (VIB Histology and Imaging Core) for histology services and J. Lamote (VIB FACS Core) for providing advice and expertise for flow cytometry experiments. We thank T. Killian and V. van Hoef (VIB Bioinformatics Core) for their help with the RNA-seq analysis and L. Tora for advice on ChIP-seq analysis. In addition, we thank S. El Kharraz and A. Farag for performing blinded analyses of several H&E staining experiments shown in this manuscript. We acknowledge all physicians from the Multidisciplinary Breast Center Leuven (especially P. Neven, A. Smeets, I. Nevelsteen and K. Punie) for their clinical contribution to the UPTIDER and/or CHEMOREL cohorts. The authors also acknowledge the patients who accepted to donate tissues postmortem in the context of the UPTIDER program, as well as the entire multidisciplinary UPTIDER team. Figure 7h was created with BioRender.com. In terms of funding, P.A.M. is supported by a Marie Sklodowska-Curie Actions individual fellowship and has received Beug Foundation funding. Y.L. has received a Chinese Scholarship Council fellowship. A.C. is a Boehringer Ingelheim PhD fellow. G.D., J.F.G., T.G. and F.R. are Research Foundation – Flanders (FWO) PhD and senior postdoctoral fellows. G.D. has previously received a Kom op tegen Kanker and J.F.G. a FWO junior fellowship. X.-Z.L. is an EMBO fellow and has received funding from the Gilead Foundation. H.F.A. has received funding from Stiching Tegen Kanker and King Baudouin funds. V.B. received funding from Fondation Nelia et Amadeo Barletta, Switzerland, 12 Rounds contre le Cancer and Université de Paris. A.M. received doctoral funding from Fondation Nelia et Amadeo Barletta, Switzerland. E.N. and I.M. received Francis Crick Institute core funding from Cancer Research UK (CRUK) (FC001112), Medical Research Council (FC001112) and the Wellcome Trust (FC001112) and the European Research Council (ERC) CoG-2020-725492. X.L. is an EMBO postdoctoral fellow. O.M.-B. is supported by a 12T1217N project by FWO at the program under the Marie Sklodowska-Curie grant agreement no. 665501. G.F. is a recipient of a postdoctoral mandate of the University Hospitals Leuven (KOOR). M.M. was supported by an ERC Consolidator grant (ImmunoFit; no. 773208). S.Z. acknowledges funding from CRUK (A17196), Stand Up to Cancer campaign for CRUK (A29800) and Breast Cancer Now (2019AugPR1307). The laboratory of T.G.P.G. is supported by the Barbara and Wilfried Mohr Foundation. V.P. acknowledges funding from a Wallenberg Academy Fellowship (KAW 2016.0123), the Swedish Research Council (VR 2020-01480), the Ragnar Söderberg Foundation) and Karolinska Institutet (SciLifeLab and KI funds). Research of the laboratory of C.D. is supported by an ERC Consolidator grant (FATLAS, 101003153), by the Fondation Cancer Luxembourg (FC/2018/07), the Belgian Foundation Against Cancer (C/2020/1441, PhD grant to S.L.) and the Funds Nadine de Beauffort (PhD grants to K.V.B. and M.D.S.). The UPTIDER program is supported by a grant from the University Hospitals from Leuven (KOOR 2021), as well as a C1 grant (14/21/114) from KU Leuven. S.M.F. acknowledges funding from the ERC under Consolidator grant agreement no. 771486–MetaRegulation, FWO Projects (G0B4122N), Beug Foundation, Fonds Baillet Latour, KU Leuven FTBO/Internal Funding/CELSA, and Stichting tegen Kanker.

Author information

Authors and Affiliations

Authors

Contributions

Conception and design was carried out by P.A.M., G.D. and S.-M.F. Development of methodology was carried out by P.A.M., G.D., A.C., M.P. and C.R.D. Acquisition of data (including providing animals, acquiring and managing patient samples and providing facilities) was conducted by P.A.M., G.D., Y.L., A.C., E.N., A.M., M.P., J.V.E., I.V., D.B., X.L., H.F.A., M.D., N.R., X.S., T.G., M.D.S., S.L., F.R., S.H., Y.L., Q.W., E.K., C.C.A., V.G., G.F., J.-C.M., D.L., M.M., V.P., S.R.Z., J.C., J.S., H.W., U.B.-D., V.B., C.D., T.G.P.G. and I.M. Analysis and interpretation of data (for example, statistical analysis, biostatistics and computational analysis) was conducted by P.A.M., J.F.G., F.C.A., M.P., O.M.-B., Q.W., K.J.L., F.R., B.B., S.D., A.A., V.P. and S.-M.F. Writing, review and/or revision of the manuscript was carried out by P.A.M. and S.-M.F. Administrative, technical or material support (reporting or organizing data and constructing databases) was the responsibility of P.A.M., D.B., M.P. and J.V.E. Study supervision was carried out by S.-M.F.

Corresponding author

Correspondence to Sarah-Maria Fendt.

Ethics declarations

Competing interests

S.-M.F. has received funding from Bayer, Merck, Gilead, Black Belt Therapeutics and Alesta Therapeutics, has consulted for Fund+ and is on the advisory board of Alesta Therapeutics. T.G.P.G. has consulted for Boehringer Ingelheim. All other authors declare no competing interests.

Peer review

Peer review information

Nature Cancer thanks Andrew Hoy and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

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 High fat diet enhances several fatty acids, while during pre-metastatic niche formation only palmitate increases.

a. Fraction of free fatty acids (gray) over total fatty acid content (black) of the liver interstitial fluid of healthy BALB/c mice (n = 7). b. Mouse weight gain upon high fat or control diet over the course of the experiment. Data are presented as mean ± SEM (n =60 mice). Mixed-effects analysis with Sidak’s multiple comparisons. c. Palmitate and oleate abundance in the lung interstitial fluid of BALB/c mice after 16 weeks on control (CD) or high fat (HFD) diet (n = 11 mice). Data are presented as mean ± SEM of absolute concentration measured by mass spectrometry. Unpaired two-tailed t-tests with Welch correction. d. Palmitate and oleate abundance in the liver interstitial fluid of BALB/c mice after 16 weeks on CD or HFD diet (n = 8 mice). Data are presented as mean ± SEM of absolute concentration measured by mass spectrometry. Unpaired two-tailed t-tests with Welch correction. e. Schematic illustration for experimental pre-metastatic niche formation procedure. CM, control media; TCM, tumor conditioned media; i.v. intravenous. f. Changes in gene expression in lung populations upon TCM injection relative to CM injections, for genes whose upregulation has been previously linked to pre-metastatic niche formation, such as S100a8, S100a9, Mmp988,89 and Tlr3 and Cxcl2 in lung alveolar type II cells88, Tlr4 and Saa3 in lung endothelial cells and macrophages90; Slc2a1, Pdk1, and Ldha in macrophages91, and S100 genes in lung fibroblasts92. The color scale denotes log2 fold changes in TCM vs. CM. g. Relative glucose concentration in the lung interstitial fluid of healthy BALB/c mice exposed to control media (n = 10 mice) or tumor condition media (n = 8 mice). Data are presented as mean ± SEM. Unpaired two-tailed t-tests with Welch correction. h. Granulocytes population (which includes neutrophils) present in the lungs after induction of pre-metastatic niche formation using tumor conditioned media or control media. Data are presented as mean ± SEM (n = 3 mice). Unpaired two-tailed t-tests with Welch correction. i. Palmitate and oleate abundance in the lung (n = 16) and liver (n≥7) interstitial fluid of BALB/c mice injected with control media (CM) or 4T1 tumor conditioned media (TCM) (3 weeks, 3 times/week). Data are presented as mean ± SEM. Unpaired two-tailed t-tests with Welch correction. j. Palmitate concentration in the liver interstitial fluid of healthy (n = 11 mice) or 4T1 tumor-bearing (PT) (n = 19 mice) BALB/c mice. Data are presented as mean ± SEM.

Source data

Extended Data Fig. 2 High fat diet moderately impacts gene expression but increases the fraction of lung resident alveolar type II cells.

a. UMAP plots for the scRNA-seq data corresponding to lungs preconditioned with control media (CM) and tumor conditioned media (TCM), or control diet (CD) and high fat diet (HFD). Color-coded based on GSVA-based marker scores for gene sets corresponding to alveolar type I (AT1) and II (AT2) marker genes. Identified clusters are indicated within black circles. Marker scores are scaled to the range 0–1 for each market set (see Methods). b. scRNA-seq-based gene expression vs. cell type and diet condition profiles for known marker for AT1 and AT2 cells and lipid-related genes indicated on the left-hand side. Scaled expression levels are indicated by the color scale, where \(\overline {CP100k}\) denotes the average gene expression level (in counts per 100k reads) over all cells of a given type in each condition, and \(\overline {CP100k}\) the average of the latter over all cell types and condition media. The areas of the circles represent the percentage of cells with non-zero expression of each gene among all cells of each type and in each dietary condition. CD, control diet; HFD, high fat diet. c. Fractions of cells corresponding to AT2 cells, among all cells present in the lung of mice exposed to control (CD) or high fat (HFD) diet, as determined from scRNA-seq data.

Extended Data Fig. 3 Lung metastases show an increase in lipid species enriched in palmitoyl acyl chains typically found in the pulmonary surfactant.

a. Fatty acid composition of the lipid classes phosphatidylcholine, phosphatidylglycerol and phosphatidylethanolamine in breast primary tumor and lung metastasis tissues from BALB/c mice orthotopically injected with 4T1 breast cancer cells. Data are presented as mean ± SEM (n = 6 mice). Unpaired nonparametric two-tailed Mann–Whitney U-tests. b. Intracellular oleate abundance from mouse (4T1, EMT6.5) and human (MCF10A H-RasV12, MCF7) breast cancer cells cultured on soft-agar (3D) or attached (2D) conditions. Data are presented as mean ± SEM (n = 3 and 4 biological replicates). c. Palmitate uptake measured by 13C16-Palmitate intracellular incorporation in 3D spheroids (n = 5 biological replicates) and 2D (n = 6 and 4 biological replicates) cultured breast cancer cells after 5 d of incubation with BSA-conjugated 13C16-Palmitic acid. Data are presented as mean ± SEM. Unpaired two-tailed t-tests with Welch correction. d. Fraction on newly synthesized fatty acids estimated by fatty acid isotopomer spectral analysis based on mass isotopomer distribution (MID) of 13C6-glucose incorporation in 3D spheroids and 2D cultured breast cancer cells in the presence of extra palmitate (75μm) for 5 d. Data are presented as mean ± 95% confidence interval (n = 4 biological replicates). Unpaired two-tailed t-tests with Welch correction. e. Intracellular palmitate and oleate abundance from 3D spheroids mouse (4T1) and human (MCF7) breast cancer cells. Data are presented as mean ± SEM (n = 4 biological replicates). One-way ANOVA with Holm-Sidak’s multiple comparison test. f. Representative pictures of 4T1 spheroids cultured in 10% FBS, or 10% FBS in the presence of palmitate (75 μm) or oleate (116 μm). Scale bar, 0.5 μm. A representative of n = 3 experiments is shown. g. Relative proliferation of 2D cultured 4T1 cells (with or without extra palmitate) normalized to condition without extra palmitate. Data are presented as mean ± SEM (n = 6 biological replicates). h. Representative pictures of 4T07 spheroids cultured in 10% FBS with or without extra palmitate (75 μm). Scale bar, 0.5 μm. A representative of n = 3 experiments is shown. i-j. 3D spheroids growth of 4T1 cells upon palmitate + oleate sup (i) and stearate (j) supplementation compared to control or palmitate supplementation, represented by the average spheroids area of >100 spheroids. Data are presented as mean ± SEM (n = 4 and 5 biological replicates). One-way ANOVA with Holm-Sidak’s multiple comparison test.

Source data

Extended Data Fig. 4 CPT1a expression is upregulated in metastases and is associated with poor prognosis in breast cancer patients.

a. Mitochondrial mass represented by mean relative fluorescence (MFI) of MitoTracker in 3D spheroids 4T1 cells growing in the absence or presence of palmitate or oleate for 5 d. MFI is shown relative to the level of MitoTracker fluorescence of 10% FBS condition. Data are presented as mean ± SD (n = 4 biological replicates). b-c. Differentially expressed genes in (b) 4T1 cells cultured in 3D spheroids or 2D monolayer in the presence of extra palmitate, or (c) in 3D spheroid 4T1 in the presence or absence of extra palmitate (10% FBS + palmitate or 10% FBS). Calculated differences in gene expression are presented by plotting the negative log10 of false discovery rate (Y-axis) against the log2 fold change of gene expression (X-axis). Each dot represents an individual gene. In red, genes belong to the GEO term lipid metabolic process (GO:0006629). The highest-ranking (top 10) overexpressed genes or genes above the established cutoff are annotated. d. CPT1a expression in 3D spheroid 4T1 cells growing for 5 d in the presence of additional palmitate, oleate and stearate. A representative image of n = 3 experiments is shown. e. Forest plot depicting the hazard ratio (HR) (x-axis) and corresponding 95% confidence intervals (denoted by error bars) for overall survival of patients with primary breast cancer according to CPT1A expression from two different cohorts: TCGA (n = 1,221 patients) and METABRIC (n = 1,904 patients). Cox proportional hazards models were applied, controlling for age, tumor stage (cTNM-staging system), and tumor subtype in both datasets. Panel complementary to Fig. 3i. f. 3D spheroids growth of MCF7 cells upon CPT1a knockdown compared to scrambled control upon palmitate supplementation (75 μm) represented by the average spheroids area of >100 spheroids. Data are presented as mean ± SEM (n = 4 and 5 biological replicates). One-way ANOVA with Tukey’s multiple comparison test. g. 3D spheroids growth (5 d) of 4T1 cells upon CPT1a inhibition using etomoxir (50 μm) in the presence of extra palmitate (75 μm) represented by the average spheroids area of >100 spheroids. Data are presented as mean ± SEM (n = 4 and 8 biological replicates). One-way ANOVA with Tukey’s multiple comparison test. h. 3D spheroids number per well of 4T1 cells upon palmitate supplementation (75 μm), CPT1a genetic inhibition performed by shRNA (shCpt1a) and CRISPR (sgCpt1a), and upon metabolic rescue by acetate (5mM). Data are presented as mean ± SEM (n = 6, 8, 6, 5, 4, and 8 biological replicates respectively). One-way ANOVA with Tukey’s multiple comparison test.

Source data

Extended Data Fig. 5 Representative lung and liver H&E staining and primary tumor weight upon CPT1a and KAT2a loss.

a. Representative pictures of tissue from the lung of mice injected with 4T1 (m.f.) and EMT6.5 (i.v.) upon genetic inhibition of CPT1a compared to non-targeting sg/shRNA as a control, based on H&E staining. Arrowheads indicate metastasis tissue. Arrows are not shown in the liver pictures. Scale bars, 2 mm. b. Final primary tumor weight (grams) from individual breast tumors upon genetic inhibition of CPT1A (n = 19 and 12 mice) and KAT2A (n = 11 mice) or control (n = 13 and 11 mice) in the 4T1 model (m.f.). Data are presented as mean ± SEM. One-way ANOVA with Dunnett’s multiple comparison test.

Source data

Extended Data Fig. 6 Breast cancer spheroids in the presence of additional palmitate rely on CPT1a for acetyl-CoA production and for sustaining palmitate-induced 3D growth.

a. Invasive ability in a 3D matrix of 4T1 cells upon CPT1a knockout (sgCpt1a) compared to control (Scrambled) cells. Invasion was assessed by measuring the invasive area of cancer cells stained with calcein green. Representative images are depicted in the left panel (scale bar, 500 μm), and quantification in the right panel. Each dot represents a different, randomly selected microscopy field (n = 5 field). b. Migratory ability of 4T1 upon CPT1a knockout (sgCpt1a) compared to control (Scrambled) cells. Migration was assessed by analyzing the total cells migrated through transwells coated with endothelial cells. Blue, DAPI nuclear staining. Representative images are depicted in the left panel (scale bar, 500 μm), and quantification in the right panel. Each dot represents a different, randomly selected microscopy field (n = 5 field). c. Relatives changes in acetyl-CoA abundance in human MCF10A H-RasV12 and MCF7 breast cancer spheroids transduced with a lentiviral vector with shRNA against CPT1A (knockdown) compared to scrambled control sequences in the presence of extra palmitate. Data are presented as mean ± SEM (n = 4 biological replicates). Unpaired two-tailed t-tests with Welch correction. d-e. Relatives changes in acetyl-CoA abundance in mouse 4T1 breast cancer spheroids transduced with a lentiviral vector with RNA against Cpt1a (c, knockdown and d, knockout) compared to non-targeting sh/sgRNA control in the presence or absence of extra palmitate. Data are presented as mean ± SEM (n = 4 biological replicates). One-way ANOVA with Dunnett’s multiple comparison test or two-tailed unpaired Student’s t-test. f. 3D spheroids growth (5 d) of 4T1 cells upon palmitate supplementation (75 μm), CPT1a genetic inhibition (shCpt1a) and metabolic rescue with octanoate (130 μm) compared to non-targeting shRNA control, represented by the average of spheroids area of >100 spheroids. Data are presented as mean ± SEM (n = 4 biological replicates). One-way ANOVA with Dunnett’s multiple comparison test. g. Schematic representation of the palmitate flux into the mitochondria via CPT1A and the ACLY-dependent export of the mitochondrial acetyl-CoA pool to the cytosol via citrate. h. Intracellular levels of ATP in CPT1a knockout and control 4T1 3D spheroids cultured for 5 d in medium containing extra palmitate (75 μm) or acetate as metabolic rescue (5 mM). Data are presented as mean ± SEM (n = 8 and 4 biological replicates). i. Heatmap display of the log2 transformed ratios obtained for the indicated histone acetylation for CPT1a knockdown and control 4T1 3D spheroids cultured for 5 d in medium containing extra palmitate (75 μm). Relative abundances ratios, light/SILAC heavy, were obtained with the SILAC (Stable Isotope Labeling with Amino acids in Cell culture) internal standard strategy93.

Source data

Extended Data Fig. 7 CPT1a deletion reduces NF-κB signaling pathway but does not affect p65 DNA binding.

a. Top 10 enriched pathways (p<0.05) obtained from gene set enrichment analysis (GSEA) of 4T1 spheroids upon CPT1a inhibition (sgCPT1a), in the presence of the extra palmitate (75 μm) or acetate as metabolic rescue (5 mM) using the Hallmark gene set from Molecular Signatures Database (MSigDB). NES, normalized enrichment score. b. Relative expression of genes implicated in invasion and metastasis, and that are known to be regulated via NF-κB activation in 4T1 spheroids. Fold change is calculated from normalized raw counts (RNA-sequencing) of CPT1a knockout and non-targeting sgRNA control 4T1 3D spheroids cultured for 5 d in medium containing extra palmitate (75 μm) or acetate (5 mM). Data are presented as mean ± SD (n = 3 biological replicates). Multiple testing correction with false discovery rate (FDR) estimation. c. Relative expression of genes implicated in invasion and metastasis, and that are known to be regulated via NF-κB activation in MCF10A H-RasV12 spheroids. Fold changes are calculated for CPT1a knockdown and non-targeting sgRNA control MCF10A H-RasV12 3D spheroids cultured for 5 d in medium containing extra palmitate (75 μm) or acetate (5 mM) and are normalized to gene expression in control cells. Data are presented as mean ± SEM (n = 3 biological replicates). One-way ANOVA with Dunnett’s multiple comparison test. d. Upstream regulator analysis performed using Ingenuity Pathway Analysis using the differential gene expression of CPT1A knockout (sgCpt1a) 4T1 3D spheroids in the presence or absence of acetate (metabolic rescue, 5 mM) as input. Activation score of the top 30 upstream regulators (left column) was compared to those predicted for the differential gene expression of CPT1A knockout versus control conditions (in the presence of extra palmitate). Genes related to the activation of the NF-κB pathway are framed. Asterisks represent the overlap P value calculated using one-sided Fisher’s Exact Test (****P<0.0001). e. GSEA enrichment plots comparing the gene expression profiles in 4T1 3D spheroids transduced with a lentiviral vector containing sgCpt1a or sgScrambled as a control (left panel) and sgCpt1a 4T1 3D spheroids cultured with or without acetate (right panel). NES, normalized enrichment score; the P value indicates the significance of the enrichment score (permutation test). f. Total p65 binding to DNA measured by electrophoretic mobility shift assay (EMSA). Arrow indicates the position of the NF-κB containing complex. A representative of n = 3 experiments is shown.

Source data

Extended Data Fig. 8 Global histone acetylation and chromatin accessibility are not consistently changed upon CPT1a and/or KAT2a loss.

a. KAT2a expression in 3D spheroid 4T1 cells growing for 5 d in the absence or presence of additional palmitate, oleate and stearate. A representative image of n = 3 experiments is shown. b. Intracellular levels of acetyl-CoA in 4T1 cells growing in 3D for 5 d in medium containing only 10% FBS, or supplemented with palmitate (75 mM), oleate (116 μm) or acetate (5 mM). Data are presented as mean ± SEM (n = 4 biological replicates). c. Heatmap of the signal intensity of H3K9ac-targeted gene loci in non-targeting RNA control, CPT1a and KAT2a knockout 4T1 3D spheroids cultured for 5 d in medium containing extra palmitate (75 μm) (n = 3 biological replicates). d. Correlation plot of H3K9 acetylation in 4T1 3D spheroids cultured in the presence of palmitate upon CPT1a inhibition with and without acetate (5 d). e. Heatmap and hierarchical clustering of top-scored downregulated genes of the NF-κB signaling pathway upon CPT1a deletion in 4T1 3D spheroids cultured in the presence of palmitate for 5 d, represented together with the expression status of the same genes upon acetate rescue and KAT2a deletion non-targeting sgRNA is used in control transfected samples (n = 3 biological replicates). f. Proliferation of 4T1 cells upon genetic inhibition of either Cpt1a or Kat2a in 2D culture measured using incucyte. Mean of growth rate ± SEM is shown (n = 6 biological replicates). One-way ANOVA with Dunnett’s multiple comparison test. g. 3D spheroids in 4T1 cells upon pharmacologic inhibition of either KAT2a using the inhibitor CPTH2 (2 μm) or CPT1A using etomoxir (50 μm) cultured for 5 d in medium with or without extra palmitate supplementation. Size quantification is represented by the average spheroids area of >100 spheroids. Data are presented as mean ± SEM (n = 3 biological replicates). One-way ANOVA with Tukey’s multiple comparison test. h. 3D spheroids number per well of 4T1 cells upon palmitate supplementation (75 μm), CPT1a or KAT2a genetic inhibition performed by CRISPR (sgCpt1a and sgKat2a) compared to non-targeting sgScrambled as a control, and upon metabolic rescue by acetate (5mM). Data are presented as mean ± SEM (n = 4 and 5 biological replicates). One-way ANOVA with Tukey’s multiple comparison test. i. Dose-response of 3D spheroid growth to the pharmacologic inhibition of KAT2a using CPTH2 inhibitor with or without extra supplementation of palmitate. Left panel, representative pictures. Right panel, spheroid size quantification is represented by the average spheroids area of >100 spheroids (n = 4 biological replicates). Two-way ANOVA with Tukey’s multiple comparison test. j. Total area and number of metastases in the lung of mice after 14 d of intravenous (i.v.) injections with 4T1 Kat2a knockout (sgKat2a, n = 8 mice) or non-targeting sgScrambled control cells (n = 10 mice) analyzed by H&E staining. Unpaired two-tailed t-tests with Welch correction.

Source data

Extended Data Fig. 9 Protein and RNA expression of genetically modified breast cancer cells.

a. Relative gene expression analysis of CPT1A in human (MCF10A H-RasV12 and MCF7), mouse (4T1, EO771-MCB3 and EMT6.5) breast cancer cells infected with either a control shRNA, or two different CPT1A, or Cpt1a shRNAs normalized to the control condition. Data are presented as mean ± SD (n = 3 biological replicates). Unpaired two-tailed t-tests. b. Protein expression in mouse 4T1 cancer cells infected with either a non-targeting sgScrambled as a control or two different sgRNA against Cpt1a and Kat2a gRNAs. A representative of n = 3 experiments is shown.

Source data

Extended Data Fig. 10 MS/MS validation of lipids detected in metastases by MALDI-MSI molecular imaging.

a. MS/MS spectrum of the ion at m/z 760.5842 detected from the metastatic areas within the lung tissue in positive ion mode using the timsTOF fleX. Ions supporting the assignment of PC O-18:1_16:0 are annotated with their corresponding mass accuracy. b. MS/MS spectrum of the ion at m/z 760.5842 detected from the metastatic areas within the lung tissue in positive ion mode using the timsTOF fleX. Ions supporting the assignment of PC 20:1_16:0 are annotated with their corresponding mass accuracy. c. MS/MS spectrum of the ion at m/z 760.5842 detected from the metastatic areas within the lung tissue in positive ion mode using the timsTOF fleX. Ions supporting the assignment of PC18:1_16:0 are annotated with their corresponding mass accuracy.

Supplementary information

Supplementary Information

Supplementary Fig. 1

Reporting Summary

Supplementary Tables 1–5

Supplementary Table 1. Clinical data of human participants who donated healthy lung tissue, protocol S57123 (UZ Leuven). Supplementary Table 2. Clinical data of patients from the UPTIDER program. Supplementary Table 3. Clinical data comparison between primary metastasized and non-metastasized patients in the CHEMOREL study. Supplementary Table 4. Clinical information and uni/multivariate analysis of TCGA and METABRIC datasets. Supplementary Table 5. Primers used to analyze mRNA levels by qPCR.

Source data

Source Data Fig. 1

Statistical Source Data for Fig. 1.

Source Data Fig. 2

Statistical Source Data for Fig. 2.

Source Data Fig. 3

Statistical Source Data for Fig. 3.

Source Data Fig. 3

Unprocessed western blot.

Source Data Fig. 4

Statistical Source Data for Fig. 4.

Source Data Fig. 5

Statistical Source Data for Fig. 5.

Source Data Fig. 6

Statistical Source Data for Fig. 6.

Source Data Fig. 6

Unprocessed western blot.

Source Data Fig. 7

Statistical Source Data for Fig. 7.

Source Data Fig. 7

Unprocessed western blot.

Source Data Extended Data Fig. 1

Statistical Source Data for Extended Data Fig. 1.

Source Data Extended Data Fig. 3

Statistical Source Data for Extended Data Fig. 3.

Source Data Extended Data Fig. 4

Statistical Source Data for Extended Data Fig. 4.

Source Data Extended Data Fig. 4

Unprocessed western blot.

Source Data Extended Data Fig. 5

Statistical Source Data for Extended Data Fig. 5.

Source Data Extended Data Fig. 6

Statistical Source Data for Extended Data Fig. 6.

Source Data Extended Data Fig. 7

Statistical Source Data for Extended Data Fig. 7.

Source Data Extended Data Fig. 8

Statistical Source Data for Extended Data Fig. 8.

Source Data Extended Data Fig. 9

Statistical Source Data for Extended Data Fig. 9.

Source Data Extended Data Fig. 8

Unprocessed western blot.

Source Data Extended Data Fig. 9

Unprocessed western blot.

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

Altea-Manzano, P., Doglioni, G., Liu, Y. et al. A palmitate-rich metastatic niche enables metastasis growth via p65 acetylation resulting in pro-metastatic NF-κB signaling. Nat Cancer 4, 344–364 (2023). https://doi.org/10.1038/s43018-023-00513-2

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s43018-023-00513-2

This article is cited by

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