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:

CD44 regulates epigenetic plasticity by mediating iron endocytosis

Abstract

CD44 is a transmembrane glycoprotein linked to various biological processes reliant on epigenetic plasticity, which include development, inflammation, immune responses, wound healing and cancer progression. Although it is often referred to as a cell surface marker, the functional regulatory roles of CD44 remain elusive. Here we report the discovery that CD44 mediates the endocytosis of iron-bound hyaluronates in tumorigenic cell lines, primary cancer cells and tumours. This glycan-mediated iron endocytosis mechanism is enhanced during epithelial–mesenchymal transitions, in which iron operates as a metal catalyst to demethylate repressive histone marks that govern the expression of mesenchymal genes. CD44 itself is transcriptionally regulated by nuclear iron through a positive feedback loop, which is in contrast to the negative regulation of the transferrin receptor by excess iron. Finally, we show that epigenetic plasticity can be altered by interfering with iron homeostasis using small molecules. This study reveals an alternative iron-uptake mechanism that prevails in the mesenchymal state of cells, which illuminates a central role of iron as a rate-limiting regulator of epigenetic plasticity.

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: CD44 mediates Hyal-dependent iron endocytosis.
Fig. 2: CD44-mediated iron endocytosis prevails in the mesenchymal state of cells.
Fig. 3: EMT is characterized by a redox signature that implicates iron.
Fig. 4: Nuclear iron is a rate-limiting regulator of epigenetic plasticity.
Fig. 5: Targeting iron homeostasis interferes with the maintenance of mesenchymal cells.
Fig. 6: Reciprocal endocytic–epigenetic regulation that involves iron.

Similar content being viewed by others

Zixuan Zhao, Xinyi Chen, … Hanry Yu

Data availability

All data are available in the manuscript or the Supplementary Information. Mass spectrometry data have been deposited at the ProteomeXchange Consortium (PRIDE Archive) with identifiers PXD011447 and PXD012862. ChIP-seq and RNA-seq data are available on the National Center for Biotechnology Information website with accession reference GSE121664. Source data are provided with this paper.

Code availability

Code employed for ChIP-seq and RNA-seq data analyses are available on Github at https://github.com/nservant/EMTiron.

References

  1. Ponta, H., Sherman, L. & Herrlich, P. A. CD44: from adhesion molecules to signalling regulators. Nat. Rev. Mol. Cell Biol. 4, 33–45 (2003).

    Article  CAS  PubMed  Google Scholar 

  2. Zöller, M. CD44: can a cancer-initiating cell profit from an abundantly expressed molecule? Nat. Rev. Cancer 11, 254–267 (2011).

    Article  PubMed  CAS  Google Scholar 

  3. Nieto, M. A., Huang, R. Y., Jackson, R. A. & Thiery, J. P. EMT: 2016. Cell 166, 21–45 (2016).

    Article  CAS  PubMed  Google Scholar 

  4. Brabletz, T., Kalluri, R., Nieto, M. A. & Weinberg, R. A. EMT in cancer. Nat. Rev. Cancer 18, 128–134 (2018).

    Article  CAS  PubMed  Google Scholar 

  5. Pastushenko, I. et al. Identification of the tumour transition states occurring during EMT. Nature 556, 463–468 (2018).

    Article  CAS  PubMed  Google Scholar 

  6. Günthert, U. et al. A new variant of glycoprotein CD44 confers metastatic potential to rat carcinoma cells. Cell 65, 13–24 (1991).

    Article  PubMed  Google Scholar 

  7. Aruffo, A., Stamenkovic, I., Melnick, M., Underhill, C. B. & Seed, B. CD44 is the principal cell surface receptor for hyaluronate. Cell 61, 1303–1313 (1990).

    Article  CAS  PubMed  Google Scholar 

  8. Hua, Q., Knudson, C. B. & Knudson, W. Internalization of hyaluronan by chondrocytes occurs via receptor-mediated endocytosis. J. Cell Sci. 106, 365–375 (1993).

    Article  CAS  PubMed  Google Scholar 

  9. Bartolazzi, A., Peach, R., Aruffo, A. & Stamenkovic, I. Interaction between CD44 and hyaluronate is directly implicated in the regulation of tumor development. J. Exp. Med. 180, 53–66 (1994).

    Article  CAS  PubMed  Google Scholar 

  10. Zoltan-Jones, A., Huang, L., Ghatak, S. & Toole, B. P. Elevated hyaluronan production induces mesenchymal and transformed properties in epithelial cells. J. Biol. Chem. 278, 45801–45810 (2003).

    Article  CAS  PubMed  Google Scholar 

  11. Toole, B. P. Hyaluronan: from extracellular glue to pericellular cue. Nat. Rev. Cancer 4, 528–539 (2004).

    Article  CAS  PubMed  Google Scholar 

  12. Mercê, A. L., Marques Carrera, L. C., Santos Romanholi, L. K. & Lobo Recio, M. A. Aqueous and solid complexes of iron(iii) with hyaluronic acid. Potentiometric titrations and infrared spectroscopy studies. J. Inorg. Biochem. 89, 212–218 (2002).

    Article  PubMed  Google Scholar 

  13. Torti, S. V. & Torti, F. M. Iron and cancer: more ore to be mined. Nat. Rev. Cancer 13, 342–355 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Schonberg, D. L. et al. Preferential iron trafficking characterizes glioblastoma stem-like cells. Cancer Cell 28, 441–455 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Mai, T. T. et al. Salinomycin kills cancer stem cells by sequestering iron in lysosomes. Nat. Chem. 9, 1025–1033 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Basuli, D. et al. Iron addiction: a novel therapeutic target in ovarian cancer. Oncogene 36, 4089–4099 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Hangauer, M. J. et al. Drug-tolerant persister cancer cells are vulnerable to GPX4 inhibition. Nature 551, 247–250 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Morel, A. P. et al. Generation of breast cancer stem cells through epithelial–mesenchymal transition. PLoS One 3, e2888 (2008).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  20. Niwa, M., Hirayama, T., Okuda, K. & Nagasawa, H. A new class of high-contrast Fe(ii) selective fluorescent probes based on spirocyclized scaffolds for visualization of intracellular labile iron delivered by transferrin. Org. Biomol. Chem. 12, 6590–6597 (2014).

    Article  CAS  PubMed  Google Scholar 

  21. Guerquin-Kern, J. L., Wu, T. D., Quintana, C. & Croisy, A. Progress in analytical imaging of the cell by dynamic secondary ion mass spectrometry (SIMS microscopy). Biochim. Biophys. Acta 1724, 228–238 (2005).

    Article  CAS  PubMed  Google Scholar 

  22. Stuelten, C. H. et al. Complex display of putative tumor stem cell markers in the NCI60 tumor cell line panel. Stem Cells 28, 649–660 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Pantopoulos, K., Porwal, S. K., Tartakoff, A. & Devireddy, L. Mechanisms of mammalian iron homeostasis. Biochemistry 51, 5705–5724 (2012).

    Article  CAS  PubMed  Google Scholar 

  24. Muckenthaler, M. U., Rivella, S., Hentze, M. W. & Galy, B. A red carpet for iron metabolism. Cell 168, 344–361 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Chuang, C. H. et al. Molecular definition of a metastatic lung cancer state reveals a targetable CD109-Janus kinase-Stat axis. Nat. Med. 23, 291–300 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Bendris, N. et al. SNX9 promotes metastasis by enhancing cancer cell invasion via differential regulation of RhoGTPases. Mol. Biol. Cell 27, 1409–1419 (2016).

    Article  CAS  PubMed Central  Google Scholar 

  27. Feng, W., Yonezawa, M., Ye, J., Jenuwein, T. & Grummt, I. PHF8 activates transcription of rRNA genes through H3K4me3 binding and H3K9me1/2 demethylation. Nat. Struct. Mol. Biol. 17, 445–450 (2010).

    Article  CAS  PubMed  Google Scholar 

  28. Fortschegger, K. et al. PHF8 targets histone methylation and RNA polymerase II to activate transcription. Mol. Cell. Biol. 30, 3286–3298 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Liu, W. et al. PHF8 mediates histone H4 lysine 20 demethylation events involved in cell cycle progression. Nature 466, 508–512 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Qi, H. H. et al. Histone H4K20/H3K9 demethylase PHF8 regulates zebrafish brain and craniofacial development. Nature 466, 503–507 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Shao, P. et al. Histone demethylase PHF8 promotes epithelial to mesenchymal transition and breast tumorigenesis. Nucleic Acids Res. 45, 1687–1702 (2017).

    Article  CAS  PubMed  Google Scholar 

  32. Carey, B. W., Finley, L. W., Cross, J. R., Allis, C. D. & Thompson, C. B. Intracellular alpha-ketoglutarate maintains the pluripotency of embryonic stem cells. Nature 518, 413–416 (2015).

    Article  CAS  PubMed  Google Scholar 

  33. Bullock, G. C. et al. Iron control of erythroid development by a novel aconitase-associated regulatory pathway. Blood 116, 97–108 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Li, X., Egervari, G., Wang, Y., Berger, S. L. & Lu, Z. Regulation of chromatin and gene expression by metabolic enzymes and metabolites. Nat. Rev. Mol. Cell Biol. 19, 563–578 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Smith, A. G., Carthew, P., Francis, J. E., Edwards, R. E. & Dinsdale, D. Characterization and accumulation of ferritin in hepatocyte nuclei of mice with iron overload. Hepatology 12, 1399–1405 (1990).

    Article  CAS  PubMed  Google Scholar 

  36. Yu, L. et al. Structural insights into a novel histone demethylase PHF8. Cell Res. 20, 166–173 (2010).

    Article  CAS  PubMed  Google Scholar 

  37. McDonald, O. G., Wu, H., Timp, W., Doi, A. & Feinberg, A. P. Genome-scale epigenetic reprogramming during epithelial-to-mesenchymal transition. Nat. Struct. Mol. Biol. 18, 867–874 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Zhao, Q. Y. et al. Global histone modification profiling reveals the epigenomic dynamics during malignant transformation in a four-stage breast cancer model. Clin. Epigenetics 8, 34 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  39. McDonald, O. G. et al. Epigenomic reprogramming during pancreatic cancer progression links anabolic glucose metabolism to distant metastasis. Nat. Genet. 49, 367–376 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Gerry, C. J. & Schreiber, S. L. Chemical probes and drug leads from advances in synthetic planning and methodology. Nat. Rev. Drug Discov. 17, 333–352 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Cañeque, T., Müller, S. & Rodriguez, R. Visualizing biologically active small molecules in cells using click chemistry. Nat. Rev. Chem. 2, 202–215 (2018).

    Article  Google Scholar 

  42. Müller, S. et al. Metformin reveals a mitochondrial copper addiction of mesenchymal cancer cells. PLoS One 13, e0206764 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  43. Janzer, A. et al. Metformin and phenformin deplete tricarboxylic acid cycle and glycolytic intermediates during cell transformation and NTPs in cancer stem cells. Proc. Natl Acad. Sci. USA 111, 10574–10579 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Zilka, O. et al. On the mechanism of cytoprotection by ferrostatin-1 and liproxstatin-1 and the role of lipid peroxidation in ferroptotic cell death. ACS Cent. Sci. 3, 232–243 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Conrad, M. & Pratt, D. A. The chemical basis of ferroptosis. Nat. Chem. Biol. 15, 1137–1147 (2019).

    Article  CAS  PubMed  Google Scholar 

  46. Flavahan, W. A., Gaskell, E. & Bernstein, B. E. Epigenetic plasticity and the hallmarks of cancer. Science 357, eaal2380 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  47. Tian, X. et al. High-molecular-mass hyaluronan mediates the cancer resistance of the naked mole rat. Nature 499, 346–349 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Gerken, T. et al. The obesity-associated FTO gene encodes a 2-oxoglutarate-dependent nucleic acid demethylase. Science 318, 1469–1472 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Jia, G. et al. N6-methyladenosine in nuclear RNA is a major substrate of the obesity-associated FTO. Nat. Chem. Biol. 7, 885–887 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Greer, E. L. & Shi, Y. Histone methylation: a dynamic mark in health, disease and inheritance. Nat. Rev. Genet. 13, 343–357 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Tsai, Y. P. et al. TET1 regulates hypoxia-induced epithelial–mesenchymal transition by acting as a co-activator. Genome Biol. 15, 513 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

Download references

Acknowledgements

We thank A. Puisieux, C. Hivroz and S. Dogniaux for providing us with HMLER and primary human T cells, the PICT-IBiSA@Pasteur Imaging Facility of Institut Curie, a member of the France-BioImaging national research infrastructure for the use of microscopes and SIMS, C. Gaillet for assistance with NMR spectroscopy, J.-L. Guerquin-Kern for assistance with SIMS sample preparation, the ICP-MS platform at the Institut de Physique du Globe de Paris, G. Arras for assistance with mass spectrometry data analysis, S. Durand and G. Kroemer for providing access to the metabolomics platform and P. Legoix for NGS sample preparation. The R.R. research group is funded by the European Research Council (ERC) under the European Union’s Horizon 2020 research and innovation programme (grant agreement no. 647973), the Fondation Charles Defforey-Institut de France and Ligue Contre le Cancer (Equipe Labellisée). R.R. and D.L. are supported by Region IdF for NMR and MS infrastructures. The Institut de Physique du Globe de Paris is supported by the IPGP multidisciplinary program PARI and Paris–Region IdF (SESAME grant agreement no. 12015908). High-throughput sequencing was performed by the ICGex NGS platform of Institut Curie, supported by ANR-10-EQPX-03 (Equipex), ANR-10-INBS-09-08 (France Génomique Consortium) from the Agence Nationale de la Recherche (Investissements d’Avenir program) and by the Cancéropole IdF and the SiRIC-Curie program—SiRIC Grant (INCa-DGOS-4654).

Author information

Authors and Affiliations

Authors

Contributions

R.R. conceptualized the study and directed the research. R.R., S.M. and F.S. designed the experiments. T.C. performed NMR spectroscopy and synthesized the clickable iron chelators. A.V. synthesized the iron(ii)-specific fluorescent probes. S.M. produced knock out cell lines and performed the experiments in relation to iron endocytosis, which included western blotting, cell imaging, RNA interference, flow cytometry and ICP-MS. T.-D.W. performed SIMS imaging. A.L. performed RT-qPCR and subcellular fractionation experiments. F.S. prepared the samples for quantitative proteomics, metabolomics and next generation sequencing. B.L. and D.L. carried out quantitative proteomics. E.C.-J. and C.G. provided tumour samples and performed cell sorting. A.D., C.V. and S.B. provided assistance with the NGS library preparation. N.S. performed bioinformatics analysis. R.R., S.M. and F.S. interpreted the data and wrote the article.

Corresponding author

Correspondence to Raphaël Rodriguez.

Ethics declarations

Competing interests

R.R. is a founder, shareholder and serves on the scientific advisory board of SideROS.

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 CD44-mediates iron endocytosis in distinct cell lines and primary cells.

a, Fluorescence microscopy of RhoNox-M-positive vesicles colocalizing with internalized Hyal-FITC or TF-647 (left panels). Scale bars, 10 μm. n = 3 biologically independent experiments for MDA-MB-468, U2OS and HT1080 cell lines and n = 1 for primary cancer cells and the LNCaP cell line. Flow cytometry of CD44 (right panels). b, Fluorescence microscopy of RhoNox-M-positive vesicles colocalizing with internalized Hyal-FITC in primary human T cells (left panel). Scale bar, 2 μm. n = 3 biologically independent experiments. Flow cytometry of CD44 (right panel). Bars and error bars, mean values ± s.d.

Extended Data Fig. 2 CD44-mediated iron uptake is Hyal-dependent in distinct cell lines and primary cells.

a, Flow cytometry of RhoNox-M fluorescence in HMLER CD44 and TFRC ko clones and a MCF7 CD44 ko clone treated with Hyal. b, Flow cytometry of RhoNox-M fluorescence in primary cancer cells and primary T cells treated with Hyal. n = 1. c, Flow cytometry of RhoNox-M fluorescence in MDA-MB-468 cells treated with Hyal of varying molecular mass. d, and e, Flow cytometry of RhoNox-M in MDA-MB-468, primary lung CTC, HMLER CD44high and MCF7 cells treated with Hyal, hyaluronidase or blocking antibodies. n = 1. HMM Hyal (0.6-1 MDa) was used in a, b, d. Data representative of n = 3 biologically independent experiments for a and c.

Extended Data Fig. 3 CD44-mediated iron endocytosis prevails in the mesenchymal state of cells.

a, Time course flow cytometry of CD44 and TfR1 at plasma membrane of MCF7 and HMLER CD44low cells treated with OSM or TGF-β as indicated. Data representative of n = 3 biologically independent experiments. b, Fluorescence microscopy of RhoNox-M-positive vesicles colocalizing with internalized Hyal-FITC or TF-647 in cells treated as indicated for 72 h. Scale bars, 10 μm. n = 3 biologically independent experiments for MCF7 cells and n = 1 for primary cells. Bars and error bars, mean values ± s.d. Unpaired t-tests, two-sided.

Extended Data Fig. 4 Quantitative analysis of proteins and metabolites in cells undergoing EMT.

a, Gene Ontology-term enrichment heatmap of proteins ranked by molecular function illustrating a bias towards proteins with oxidoreductase activity being increased in the mesenchymal cell state. n = 3 biologically independent experiments. Statistical analysis, Material and Methods. b, Western blots of selected proteins. Data representative of n = 3 biologically independent experiments. c, Heatmap of metabolites in cells treated with EGF for 60 h. n = 4 technical replicates. MDA-MB-468 cells throughout the figure and treated with EGF for 72 h unless stated otherwise.

Extended Data Fig. 5 Genome-wide analysis of histone marks in cells undergoing EMT.

a, H3K9me2 ChIP-seq profiles for selected genes. n = 3 biologically independent experiments. b, ChIP-qPCR of selected genes. n = 2 biologically independent experiments. ce, ChIP-seq profiles of H3K4me3, H3K27me3 and H3K9me3 for selected genes. n = 2 biologically independent experiments. MDA-MB-468 cells were used throughout the figure and treated with EGF for 72 h.

Extended Data Fig. 6 Pharmacological targeting of iron-regulated processes.

a, Western blots of H3K9me2 in cells co-treated with OSM and DFO or TGF-β and DFO as indicated. n = 1 for primary cells. b, Western blots of proteins whose genes are regulated by H3K9me2 in cells co-treated with OSM and DFO or TGF-β and DFO as indicated. n = 1 for primary cells. c, Molecular structure of clickable deferasirox (cDFX) (left), fluorescence microscopy of labelled cDFX and the mitochondrial component Cyt c (right). Scale bar, 10 μm. d, αKG quantification assay of MDA-MB-468 cells co-treated with EGF and deferasirox (DFX). n = 3 technical replicates. e, Western blots of CD44 and H3K9me2 in MDA-MB-468 cells co-treated with EGF and DFX. Data representative of n = 3 biologically independent experiments throughout the figure unless stated otherwise. Bars and error bars, mean values ± s.d.

Supplementary information

Supplementary Information

Materials and Methods, Supplementary References, Table 5 and original western blots.

Reporting Summary

Supplementary Table 1

Quantitative label-free proteomics.

Supplementary Table 2

Quantitative metabolomics.

Supplementary Table 3

Quantitative mass spectrometry analysis of histone marks.

Supplementary Table 4

ChIP-seq and RNA-seq analyses.

Source data

Source Data Fig. 1

Statistical Source Data.

Source Data Fig. 1

Unprocessed western blots.

Source Data Fig. 2

Statistical Source Data.

Source Data Fig. 2

Unprocessed western blots.

Source Data Fig. 3

Statistical Source Data.

Source Data Fig. 4

Unprocessed western blots.

Source Data Fig. 5

Statistical Source Data.

Source Data Fig. 5

Unprocessed western blots.

Source Data Extended Fig. 3

Statistical Source Data.

Source Data Extended Fig. 4

Unprocessed western blots.

Source Data Extended Fig. 6

Statistical Source Data

Source Data Extended Fig. 6

Unprocessed western blots.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Müller, S., Sindikubwabo, F., Cañeque, T. et al. CD44 regulates epigenetic plasticity by mediating iron endocytosis. Nat. Chem. 12, 929–938 (2020). https://doi.org/10.1038/s41557-020-0513-5

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41557-020-0513-5

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