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:

Lymph protects metastasizing melanoma cells from ferroptosis

Abstract

Cancer cells, including melanoma cells, often metastasize regionally through the lymphatic system before metastasizing systemically through the blood1,2,3,4; however, the reason for this is unclear. Here we show that melanoma cells in lymph experience less oxidative stress and form more metastases than melanoma cells in blood. Immunocompromised mice with melanomas derived from patients, and immunocompetent mice with mouse melanomas, had more melanoma cells per microlitre in tumour-draining lymph than in tumour-draining blood. Cells that metastasized through blood, but not those that metastasized through lymph, became dependent on the ferroptosis inhibitor GPX4. Cells that were pretreated with chemical ferroptosis inhibitors formed more metastases than untreated cells after intravenous, but not intralymphatic, injection. We observed multiple differences between lymph fluid and blood plasma that may contribute to decreased oxidative stress and ferroptosis in lymph, including higher levels of glutathione and oleic acid and less free iron in lymph. Oleic acid protected melanoma cells from ferroptosis in an Acsl3-dependent manner and increased their capacity to form metastatic tumours. Melanoma cells from lymph nodes were more resistant to ferroptosis and formed more metastases after intravenous injection than did melanoma cells from subcutaneous tumours. Exposure to the lymphatic environment thus protects melanoma cells from ferroptosis and increases their ability to survive during subsequent metastasis through the blood.

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

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

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

Fig. 1: Melanoma cells in lymph experience less oxidative stress than melanoma cells in blood.
Fig. 2: Melanoma cells undergo increased ferroptosis in blood compared to lymph.
Fig. 3: Oleic acid levels are higher in lymph and in melanoma cells from lymph, and oleic acid protects against ferroptosis.

Similar content being viewed by others

Data availability

Uncropped western blots are provided in Supplementary Fig. 1. All other data are available from the corresponding author upon request. Source data are provided with this paper.

References

  1. Sleeman, J., Schmid, A. & Thiele, W. Tumor lymphatics. Semin. Cancer Biol. 19, 285–297 (2009).

    CAS  PubMed  Google Scholar 

  2. Alitalo, A. & Detmar, M. Interaction of tumor cells and lymphatic vessels in cancer progression. Oncogene 31, 4499–4508 (2012).

    CAS  PubMed  Google Scholar 

  3. Leong, S. P. et al. Cutaneous melanoma: a model to study cancer metastasis. J. Surg. Oncol. 103, 538–549 (2011).

    PubMed  Google Scholar 

  4. Willis, R. A. The Spread of Tumours in the Human Body 3rd edn (Butterworths, 1973).

  5. Naxerova, K. et al. Origins of lymphatic and distant metastases in human colorectal cancer. Science 357, 55–60 (2017).

    CAS  PubMed  PubMed Central  ADS  Google Scholar 

  6. Gundem, G. et al. The evolutionary history of lethal metastatic prostate cancer. Nature 520, 353–357 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  7. Sanborn, J. Z. et al. Phylogenetic analyses of melanoma reveal complex patterns of metastatic dissemination. Proc. Natl Acad. Sci. USA 112, 10995–11000 (2015).

    CAS  PubMed  ADS  Google Scholar 

  8. McFadden, D. G. et al. Genetic and clonal dissection of murine small cell lung carcinoma progression by genome sequencing. Cell 156, 1298–1311 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  9. Pereira, E. R. et al. Lymph node metastases can invade local blood vessels, exit the node, and colonize distant organs in mice. Science 359, 1403–1407 (2018).

    CAS  PubMed  PubMed Central  ADS  Google Scholar 

  10. Brown, M. et al. Lymph node blood vessels provide exit routes for metastatic tumor cell dissemination in mice. Science 359, 1408–1411 (2018).

    CAS  PubMed  ADS  Google Scholar 

  11. Morton, D. L. et al. Final trial report of sentinel-node biopsy versus nodal observation in melanoma. N. Engl. J. Med. 370, 599–609 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  12. Leiter, U. et al. Complete lymph node dissection versus no dissection in patients with sentinel lymph node biopsy positive melanoma (DeCOG-SLT): a multicentre, randomised, phase 3 trial. Lancet Oncol. 17, 757–767 (2016).

    PubMed  Google Scholar 

  13. Faries, M. B. et al. Completion dissection or observation for sentinel-node metastasis in melanoma. N. Engl. J. Med. 376, 2211–2222 (2017).

    PubMed  PubMed Central  Google Scholar 

  14. Wallace, A. C. & Hollenberg, N. K. The transplantability of tumours by intravenous and intralymphatic routes. Br. J. Cancer 19, 338–342 (1965).

    CAS  PubMed  PubMed Central  Google Scholar 

  15. Hoshida, T. et al. Imaging steps of lymphatic metastasis reveals that vascular endothelial growth factor-C increases metastasis by increasing delivery of cancer cells to lymph nodes: therapeutic implications. Cancer Res. 66, 8065–8075 (2006).

    CAS  PubMed  Google Scholar 

  16. Lee, C. K. et al. Tumor metastasis to lymph nodes requires YAP-dependent metabolic adaptation. Science 363, 644–649 (2019).

    CAS  PubMed  ADS  Google Scholar 

  17. Pascual, G. et al. Targeting metastasis-initiating cells through the fatty acid receptor CD36. Nature 541, 41–45 (2017).

    CAS  ADS  Google Scholar 

  18. Ma, Q. et al. Unexpected contribution of lymphatic vessels to promotion of distant metastatic tumor spread. Sci. Adv. 4, eaat4758 (2018).

    CAS  PubMed  PubMed Central  ADS  Google Scholar 

  19. Vanharanta, S. & Massagué, J. Origins of metastatic traits. Cancer Cell 24, 410–421 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  20. Piskounova, E. et al. Oxidative stress inhibits distant metastasis by human melanoma cells. Nature 527, 186–191 (2015).

    CAS  PubMed  PubMed Central  ADS  Google Scholar 

  21. Le Gal, K. et al. Antioxidants can increase melanoma metastasis in mice. Sci. Transl. Med. 7, 308re8 (2015).

    PubMed  Google Scholar 

  22. Dixon, S. J. & Stockwell, B. R. The hallmarks of ferroptosis. Annu. Rev. Cancer Biol. 3, 35–54 (2019).

    Google Scholar 

  23. Wang, W. et al. CD8+ T cells regulate tumour ferroptosis during cancer immunotherapy. Nature 569, 270–274 (2019).

    CAS  PubMed  PubMed Central  ADS  Google Scholar 

  24. Zhang, Y. et al. Imidazole ketone erastin induces ferroptosis and slows tumor growth in a mouse lymphoma model. Cell Chem. Biol. 26, 623–633 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  25. Wu, J. et al. Intercellular interaction dictates cancer cell ferroptosis via NF2–YAP signalling. Nature 572, 402–406 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  26. Viswanathan, V. S. et al. Dependency of a therapy-resistant state of cancer cells on a lipid peroxidase pathway. Nature 547, 453–457 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  27. Quintana, E. et al. Human melanoma metastasis in NSG mice correlates with clinical outcome in patients. Sci. Transl. Med. 4, 159ra149 (2012).

    PubMed  PubMed Central  Google Scholar 

  28. Meeth, K., Wang, J. X., Micevic, G., Damsky, W. & Bosenberg, M. W. The YUMM lines: a series of congenic mouse melanoma cell lines with defined genetic alterations. Pigment Cell Melanoma Res. 29, 590–597 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  29. Hangai-Hoger, N., Cabrales, P., Briceño, J. C., Tsai, A. G. & Intaglietta, M. Microlymphatic and tissue oxygen tension in the rat mesentery. Am. J. Physiol. Heart Circ. Physiol. 286, H878–H883 (2004).

    CAS  PubMed  Google Scholar 

  30. Tasdogan, A. et al. Metabolic heterogeneity confers differences in melanoma metastatic potential. Nature 577, 115–120 (2020).

    CAS  PubMed  ADS  Google Scholar 

  31. Friedmann Angeli, J.P. et al. Inactivation of the ferroptosis regulator Gpx4 triggers acute renal failure in mice. Nat Cell Biol 16, 1180–1191 (2014).

    CAS  PubMed  Google Scholar 

  32. Seiler, A. et al. Glutathione peroxidase 4 senses and translates oxidative stress into 12/15-lipoxygenase dependent- and AIF-mediated cell death. Cell Metab. 8, 237–248 (2008).

    CAS  PubMed  Google Scholar 

  33. Doll, S. et al. ACSL4 dictates ferroptosis sensitivity by shaping cellular lipid composition. Nat. Chem. Biol. 13, 91–98 (2017).

    CAS  PubMed  Google Scholar 

  34. Kayden, H. J., Karmen, A. & Dumont, A. Alterations in the fatty acid composition of human lymph and serum lipoproteins by single feedings. J. Clin. Invest. 42, 1373–1381 (1963).

    CAS  PubMed  PubMed Central  Google Scholar 

  35. Magtanong, L. et al. Exogenous monounsaturated fatty acids promote a ferroptosis-resistant cell state. Cell Chem. Biol. 26, 420–432 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  36. Spitz, D. R., Kinter, M. T., Kehrer, J. P. & Roberts, R. J. The effect of monosaturated and polyunsaturated fatty acids on oxygen toxicity in cultured cells. Pediatr. Res. 32, 366–372 (1992).

    CAS  PubMed  Google Scholar 

  37. Bowden, J. A. et al. Harmonizing lipidomics: NIST interlaboratory comparison exercise for lipidomics using SRM 1950-metabolites in frozen human plasma. J. Lipid Res. 58, 2275–2288 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  38. Fisher, E. A. & Ginsberg, H. N. Complexity in the secretory pathway: the assembly and secretion of apolipoprotein B-containing lipoproteins. J. Biol. Chem. 277, 17377–17380 (2002).

    CAS  PubMed  Google Scholar 

  39. Tang, Y., Zhou, J., Hooi, S. C., Jiang, Y. M. & Lu, G. D. Fatty acid activation in carcinogenesis and cancer development: essential roles of long-chain acyl-CoA synthetases. Oncol. Lett. 16, 1390–1396 (2018).

    PubMed  PubMed Central  Google Scholar 

  40. Chen, W. C. et al. Systematic analysis of gene expression alterations and clinical outcomes for long-chain acyl-coenzyme A synthetase family in cancer. PLoS ONE 11, e0155660 (2016).

    PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  42. Doll, S. et al. FSP1 is a glutathione-independent ferroptosis suppressor. Nature 575, 693–698 (2019).

    CAS  PubMed  ADS  Google Scholar 

  43. Bersuker, K. et al. The CoQ oxidoreductase FSP1 acts parallel to GPX4 to inhibit ferroptosis. Nature 575, 688–692 (2019).

    CAS  PubMed  PubMed Central  ADS  Google Scholar 

  44. Veglia, F. et al. Fatty acid transport protein 2 reprograms neutrophils in cancer. Nature 569, 73–78 (2019).

    CAS  PubMed  PubMed Central  ADS  Google Scholar 

  45. Kassan, A. et al. Acyl-CoA synthetase 3 promotes lipid droplet biogenesis in ER microdomains. J. Cell Biol. 203, 985–1001 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  46. Agathocleous, M. et al. Ascorbate regulates haematopoietic stem cell function and leukaemogenesis. Nature 549, 476–481 (2017).

    PubMed  PubMed Central  ADS  Google Scholar 

  47. Hu, Y. & Smyth, G. K. ELDA: extreme limiting dilution analysis for comparing depleted and enriched populations in stem cell and other assays. J. Immunol. Methods 347, 70–78 (2009).

    CAS  PubMed  Google Scholar 

  48. Chong, J. W. D.S. and Xia, J. Using MetaboAnalyst 4.0 for comprehensive and integrative metabolomics data analysis. Curr. Protoc. Bioinformatics 68, e86 (2019).

    PubMed  Google Scholar 

Download references

Acknowledgements

S.J.M. is a Howard Hughes Medical Institute Investigator, the Mary McDermott Cook Chair in Pediatric Genetics, the Kathryn and Gene Bishop Distinguished Chair in Pediatric Research, the director of the Hamon Laboratory for Stem Cells and Cancer, and a Cancer Prevention and Research Institute of Texas Scholar. The research was supported by the Cancer Prevention and Research Institute of Texas (RP170114 and RP180778) and by the National Institutes of Health (NIH; U01 CA228608). A.T. was supported by the Leopoldina Fellowship (LPDS 2016-16) from the German National Academy of Sciences and the Fritz Thyssen Foundation. B.S. was supported by a Ruth L. Kirschstein National Research Service Award Postdoctoral Fellowship from the National Heart, Lung, and Blood Institute (F32 HL139016). M.L.M. and D.R.S. were supported by NIH grants P01 (CA217797) and P30 (CA086862). We thank M. Dellinger for comments on the manuscript; N. Meireles for collecting human melanomas; M. Nitcher for mouse colony management; the BioHPC (High Performance Computing) core for data storage; and N. Loof and the Moody Foundation Flow Cytometry Facility.

Author information

Authors and Affiliations

Authors

Contributions

J.M.U. and S.J.M. conceived the project, and designed and interpreted experiments. J.M.U. performed most of the experiments. V.R. and A.T. helped J.M.U. to perform the in vivo tumorigenesis assays. E.C.M. performed the western blot analyses and assisted with flow cytometry. B.S., A.T. and Z.G. helped to design and generate CRISPR gene-targeting constructs for Gpx4 and Acsl3. M.L.M. and D.R.S. advised on the interpretation of antioxidant levels in blood versus lymph. A.B.D. provided patient melanoma samples and expertise on the clinical behaviour of melanomas. T.P.M. and M.S.M.-S. developed methods for lipidomic profiling and performed all of the mass spectrometric analysis of melanoma specimens. Z.Z. performed statistical analyses. J.M.U. and S.J.M. wrote the manuscript.

Corresponding author

Correspondence to Sean J. Morrison.

Ethics declarations

Competing interests

S.J.M. is an advisor for Frequency Therapeutics and Protein Fluidics as well as a stockholder in G1 Therapeutics and Mereo Biopharma.

Additional information

Peer review information Nature thanks Martin Bergo, Marcus Conrad, Sarah-Maria Fendt and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

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

Extended data figures and tables

Extended Data Fig. 1 Representative flow cytometry gates for the isolation of melanoma cells from blood and lymph and representative bioluminescence images of visceral organs to quantify the metastatic disease burden.

Related to Table 1, Figs. 1, 2, 3. ad, Flow cytometry plots showing the gating strategies used to identify human melanoma cells in the blood (a) or lymph (b) of NSG mice or mouse melanoma cells in the blood (c) or lymph (d) of C57BL mice. In all cases, cells were gated on forward scatter area versus side height (FSC-H vs. FSC-A) to exclude red blood cells and cell clumps. Mouse haematopoietic and endothelial cells were excluded by gating out cells that stained positively for anti-mouse CD45, CD31, or Ter119. Human melanoma cells were selected by including cells that stained positively for HLA-ABC and mouse melanoma cells were selected by including cells that stained positively for CD146. Melanoma cells were also identified in these studies based on DsRed, which was stably expressed in all melanomas along with luciferase. eg, Representative bioluminescence imaging of visceral organs dissected from a negative control mouse (e), and mice transplanted with luciferase-expressing human (f) or mouse (g) melanomas. h, Evan’s blue dye was injected into a subcutaneous melanoma to expose the tumour-draining blood (white arrow) and lymphatic (black arrow) vessels. i, Inefficiently metastasizing human melanomas were transplanted subcutaneously into NSG mice and the number of melanoma cells per microlitre of blood and tumour-draining lymph were determined (n = 4 or 5 mice per melanoma from two independent experiments). Statistical significance was assessed using a Kruskal–Wallis test (*P < 0.05, ***P < 0.001). Exact P values are provided in the source data files.

Source data

Extended Data Fig. 2 The effect of liproxstatin-1 on the growth of subcutaneous tumours and the effect of other inhibitors on metastasis.

Related to Figs. 1, 2. a, Addition of 2-mercaptoethanol to M405 melanoma cells isolated from subcutaneous tumours or the blood blunted the increase in ROS levels in melanoma cells from the blood. b, c, Human (b) or mouse (c) melanomas were treated in culture with erastin and/or deferoxamine. dh, Treatment of mice with liproxstatin-1 had little or no effect on the growth of subcutaneous tumours formed by human (df) or mouse (g, h) melanomas. i, Pretreatment of inefficiently metastasizing human melanoma cells with Liproxstatin-1 did not significantly affect metastatic disease burden after intravenous injection into NSG mice. j, Mouse melanomas were pretreated with autophagy (3-MA), apoptosis (ZVAD), or necroptosis (GSK′872) inhibitors then injected intravenously into C57BL mice and metastatic disease was assessed 1 month later by bioluminescence imaging. The number of replicates is indicated in each panel and the number of independent experiments is shown in Supplementary Data, ‘Statistics and reproducibility’. All data represent mean ± s.d. Statistical significance was assessed using a correlated-samples two-way ANOVA followed by Sidak’s multiple comparisons adjustment (a), two-way ANOVA (i) followed by Tukey’s multiple comparison adjustment (b), Kruskal–Wallis tests followed by Dunn’s multiple comparisons adjustment (c (Y1.7), j), one-way ANOVA followed by Tukey’s multiple comparisons adjustment (c (Y3.3)), or nparLD tests (dh). No statistically significant differences were observed in df, i or j. For all panels, statistical tests were two-sided where applicable and *P < 0.05, **P < 0.01, ***P < 0.001. Exact P values are provided in the source data files.

Source data

Extended Data Fig. 3 The effect of Gpx4 deletion on the survival and proliferation of mouse melanomas in culture.

Related to Fig. 2. a, Western blot analysis of GPX4 in parental or Gpx4-deleted mouse melanomas (representative of 2 independent experiments). b, Western blot analysis of GPX4 in efficiently and inefficiently metastasizing melanomas from patients as well as normal mouse brain and liver tissue. Actin was used as a loading control (representative of 2 independent experiments). Uncropped western blots are in Supplementary Fig. 1. c, d, Gpx4-deficiency did not significantly affect the growth of melanoma cells cultured in low oxygen (c) but did significantly reduce the growth of some melanomas cultured at atmospheric oxygen levels (d) (n = 6 replicate cultures per melanoma; data reflect one representative experiment of two conducted). e, Lipid ROS levels in melanoma cells from subcutaneous tumours formed by Gpx4-deleted or parental control melanomas cells (n = 6 mice per melanoma in two independent experiments). fi, Growth of primary subcutaneous tumours (f, g) and frequency of circulating melanoma cells in the blood (h, i) of NSG mice transplanted with parental or Gpx4-deleted melanomas (n = 4 or 5 mice per melanoma per experiment from two independent experiments). All data represent mean ± s.d. Statistical significance was assessed using repeated measures two-way ANOVAs (c (Y1.7), d) or mixed-effects analysis (c (Y3.3)) followed by Dunnett’s multiple comparisons adjustment (c, d), correlated-samples two-way ANOVA (e), nparLD tests (f, g), Fisher’s LSD test (e), Welch’s one-way ANOVA followed by Dunett’s T3 multiple comparisons adjustment (h), or Kruskal–Wallis tests followed by Dunn’s multiple comparisons adjustment (i). For all panels, statistical tests were two-sided where applicable and **P < 0.01, ***P < 0.001. Exact P values are provided in the source data files.

Source data

Extended Data Fig. 4 Lipid species in plasma and lymph.

Related to Fig. 3. a, b, Lipid species that significantly differed in abundance between the plasma and lymph of NSG (a) or C57BL (b) mice (P < 0.01). c, Relative triacylglycerol content in the ApoB+ and ApoB fractions of blood plasma or lymph fluid (after cells were removed) from C57BL mice (two independent samples per treatment). d, Relative oleic acid abundance in the ApoB+ and ApoB fractions of blood plasma or lymph fluid from C57BL mice (two independent samples per treatment). Statistical significance was assessed using generalized linear modelling with log-transformed, half-min imputed data replacing zeros followed by the Benjamini-Hochberg multiple comparisons adjustment using two-sided t-statistics (a and b). The number of replicates is indicated in each panel. Each panel reflects two independent experiments. All data represent means and, when present, error bars reflect s.d. Exact P values are provided in the source data files.

Source data

Extended Data Fig. 5 ACSL3 is required for oleic acid incorporation into phospholipids and the protective effect of oleic acid against ferroptosis.

Related to Fig. 3. a, Western blot analysis of ACSL3 in efficiently and inefficiently metastasizing melanomas from patients as well as normal mouse brain and liver tissue. Actin was used as a loading control (representative of 2 independent experiments). b, Western blot analysis of ACSL3 in parental control and Acsl3-deleted mouse melanomas (representative of 2 independent experiments). Uncropped western blots are in Supplementary Fig. 1. cd, Relative levels of oleic acid in phospholipids from Acsl3-deleted and parental control melanomas. In some cases, wild-type (Acsl3OE) or catalytically dead mutant Acsl3 (mut. Acsl3OE) were overexpressed in YUMM1.7 (c) or YUMM3.3 (d) mouse melanomas. The number of replicates per treatment is indicated in each panel (two independent experiments). e, Lipid ROS (BODIPY-C11Oxidized/BODIPY-C11Oxidized + BODIPY-C11Non-oxidized ratio) levels in mouse melanoma cells from subcutaneous tumours in C57BL mice. The number of replicates per melanoma is indicated in each panel (two independent experiments). The data from parental controls cells in this experiment are also shown in Extended Data Fig. 3e. f, Growth of Acsl3-deleted and parental control melanomas in culture (4 replicate cultures per melanoma per experiment, representative of two independent experiments; no differences were statistically significant). g, h, Growth of Acsl3-deleted melanomas in culture with oleic acid and with or without erastin. In some cases, wild-type (Acsl3OE) or catalytically dead mutant Acsl3 (mut. Acsl3OE) were overexpressed in YUMM1.7 (g) or YUMM3.3 (h) melanomas. i, Metastatic disease burden in mice intranodally injected with Acsl3-deleted or control melanomas. All data represent mean ± s.d. Statistical significance was assessed using one-sided ANOVAs followed by Sidak’s and Dunnett’s (c, d) multiple comparisons adjustments, paired t-tests (e), repeated measures two-sided ANOVAs followed by Dunnett’s multiple comparisons adjustment (f), Kruskal–Wallis tests followed by Dunn’s multiple comparisons adjustment (g, h, i (Y3.3)), or Welch’s one-way ANOVA followed by Dunnett’s T3 multiple comparisons adjustment (i (Y1.7)). For all panels, statistical tests were two-sided where applicable and *P < 0.05, **P < 0.01, ***P < 0.001. Exact P values are provided in the source data files.

Source data

Extended Data Fig. 6 Expression of potential ferroptosis regulators by melanoma cells.

Related to Fig. 3. af, Western blot analysis of SLC7A11 (a, b), ACSL4 (c, d) and FSP1 (e, f) in efficiently and inefficiently metastasizing human melanomas (a, c, e) as well as mouse melanomas (b, d, f) (representative of two experiments). Normal mouse liver and brain, human lung, and mouse fibroblasts were sometimes included as positive or negative controls. Uncropped western blots can be found in Supplementary Fig. 1. g, FSP1 transcript levels by qRT–PCR in human melanoma cells isolated from subcutaneous tumours, blood, or lymph of xenografted mice (3 replicates (each replicate was pooled from 4 or 5 mice) per melanoma). h, RNA-sequencing analysis of the fatty acid transporters FATP1, FATP3, FATP4, and FATP5 in efficiently and inefficiently metastasizing human melanomas (2 to 3 replicates per melanoma). i, Staining with anti-CD36 or isotype control antibody in human and mouse melanomas. Data are representative of 2 tumours per melanoma. j, Elevated levels of oleic acid in lymph suppress lipid ROS accumulation and ferroptosis in metastasizing melanoma cells, increasing their ability to survive during subsequent dissemination through the blood. Image created with BioRender. No statistically significant differences were observed in g or h. All data represent mean ± s.d. Statistics were determined repeated measures two-sided ANOVA followed by Tukey’s multiple comparisons adjustment (g). Exact P values are provided in the source data files.

Source data

Extended Data Table 1 Characterization of human and mouse melanomas
Extended Data Table 2 List of lipid species analysed by mass spectrometry

Supplementary information

Supplementary Information

This file contains Supplementary Figure 1: uncropped western blot scans with size marker indications and Supplementary Data: Statistics and Reproducibility - the numbers of replicates and independent experiments for each figure panel in each figure.

Reporting Summary

Source data

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Ubellacker, J.M., Tasdogan, A., Ramesh, V. et al. Lymph protects metastasizing melanoma cells from ferroptosis. Nature 585, 113–118 (2020). https://doi.org/10.1038/s41586-020-2623-z

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41586-020-2623-z

This article is cited by

Comments

By submitting a comment you agree to abide by our Terms and Community Guidelines. If you find something abusive or that does not comply with our terms or guidelines please flag it as inappropriate.

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