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.

  • Letter
  • Published:

Copper is required for oncogenic BRAF signalling and tumorigenesis

Subjects

Abstract

The BRAF kinase is mutated, typically Val 600→Glu (V600E), to induce an active oncogenic state in a large fraction of melanomas, thyroid cancers, hairy cell leukaemias and, to a smaller extent, a wide spectrum of other cancers1,2. BRAFV600Ephosphorylates and activates the MEK1 and MEK2 kinases, which in turn phosphorylate and activate the ERK1 and ERK2 kinases, stimulating the mitogen-activated protein kinase (MAPK) pathway to promote cancer3. Targeting MEK1/2 is proving to be an important therapeutic strategy, given that a MEK1/2 inhibitor provides a survival advantage in metastatic melanoma4, an effect that is increased when administered together with a BRAFV600Einhibitor5. We previously found that copper (Cu) influx enhances MEK1 phosphorylation of ERK1/2 through a Cu–MEK1 interaction6. Here we show decreasing the levels of CTR1 (Cu transporter 1), or mutations in MEK1 that disrupt Cu binding, decreased BRAFV600E-driven signalling and tumorigenesis in mice and human cell settings. Conversely, a MEK1–MEK5 chimaera that phosphorylated ERK1/2 independently of Cu or an active ERK2 restored the tumour growth of murine cells lacking Ctr1. Cu chelators used in the treatment of Wilson disease7decreased tumour growth of human or murine cells transformed by BRAFV600E or engineered to be resistant to BRAF inhibition. Taken together, these results suggest that Cu-chelation therapy could be repurposed to treat cancers containing the BRAFV600Emutation.

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

Figure 1: Binding of Cu to MEK1 promotes MAPK signalling and tumorigenesis by oncogenic BRAF.
Figure 2: Knockdown of CTR1 decreases MAPK signalling and tumorigenesis specifically by oncogenic BRAF.
Figure 3: Genetic ablation of Ctr1 decreases MAPK signalling and tumorigenesis and extends the lifespan in a mouse model of BrafV600E-driven lung cancer.
Figure 4: Pharmacological chelation of Cu decreases tumour growth of BRAFV600E-driven and vemurafenib-resistant tumour cells.

Similar content being viewed by others

References

  1. Davies, H. et al. Mutations of the BRAF gene in human cancer. Nature 417, 949–954 (2002)

    CAS  PubMed  ADS  Google Scholar 

  2. Forbes, S. A. et al. COSMIC: mining complete cancer genomes in the Catalogue of Somatic Mutations in Cancer. Nucleic Acids Res. 39, D945–D950 (2011)

    Article  CAS  PubMed  Google Scholar 

  3. Wan, P. T. et al. Mechanism of activation of the RAF–ERK signaling pathway by oncogenic mutations of B-RAF. Cell 116, 855–867 (2004)

    CAS  PubMed  Google Scholar 

  4. Flaherty, K. T. et al. Improved survival with MEK inhibition in BRAF-mutated melanoma. N. Engl. J. Med. 367, 107–114 (2012)

    CAS  PubMed  Google Scholar 

  5. Flaherty, K. T. et al. Combined BRAF and MEK inhibition in melanoma with BRAF V600 mutations. N. Engl. J. Med. 367, 1694–1703 (2012)

    CAS  PubMed  PubMed Central  Google Scholar 

  6. Turski, M. L. et al. A novel role for copper in Ras/mitogen-activated protein kinase signaling. Mol. Cell. Biol. 32, 1284–1295 (2012)

    CAS  PubMed  PubMed Central  Google Scholar 

  7. Ala, A., Walker, A. P., Ashkan, K., Dooley, J. S. & Schilsky, M. L. Wilson’s disease. Lancet 369, 397–408 (2007)

    CAS  PubMed  Google Scholar 

  8. Solit, D. B. et al. BRAF mutation predicts sensitivity to MEK inhibition. Nature 439, 358–362 (2006)

    CAS  PubMed  ADS  Google Scholar 

  9. Lee, J., Petris, M. J. & Thiele, D. J. Characterization of mouse embryonic cells deficient in the ctr1 high affinity copper transporter. Identification of a Ctr1-independent copper transport system. J. Biol. Chem. 277, 40253–40259 (2002)

    CAS  PubMed  Google Scholar 

  10. Bertinato, J. & L’Abbe, M. R. Copper modulates the degradation of copper chaperone for Cu,Zn superoxide dismutase by the 26 S proteosome. J. Biol. Chem. 278, 35071–35078 (2003)

    CAS  PubMed  Google Scholar 

  11. Puig, S., Lee, J., Lau, M. & Thiele, D. J. Biochemical and genetic analyses of yeast and human high affinity copper transporters suggest a conserved mechanism for copper uptake. J. Biol. Chem. 277, 26021–26030 (2002)

    CAS  PubMed  Google Scholar 

  12. Bridgewater, J. D., Lim, J. & Vachet, R. W. Using metal-catalyzed oxidation reactions and mass spectrometry to identify amino acid residues within 10 Å of the metal in Cu-binding proteins. J. Am. Soc. Mass Spectrom. 17, 1552–1559 (2006)

    CAS  PubMed  Google Scholar 

  13. Fischmann, T. O. et al. Crystal structures of MEK1 binary and ternary complexes with nucleotides and inhibitors. Biochemistry 48, 2661–2674 (2009)

    CAS  PubMed  Google Scholar 

  14. Huang, W., Kessler, D. S. & Erikson, R. L. Biochemical and biological analysis of Mek1 phosphorylation site mutants. Mol. Biol. Cell 6, 237–245 (1995)

    CAS  PubMed  PubMed Central  Google Scholar 

  15. Nakamura, K., Uhlik, M. T., Johnson, N. L., Hahn, K. M. & Johnson, G. L. PB1 domain-dependent signaling complex is required for extracellular signal-regulated kinase 5 activation. Mol. Cell. Biol. 26, 2065–2079 (2006)

    CAS  PubMed  PubMed Central  Google Scholar 

  16. English, J. M. et al. Contribution of the ERK5/MEK5 pathway to Ras/Raf signaling and growth control. J. Biol. Chem. 274, 31588–31592 (1999)

    CAS  PubMed  Google Scholar 

  17. Roberts, P. J. & Der, C. J. Targeting the Raf-MEK-ERK mitogen-activated protein kinase cascade for the treatment of cancer. Oncogene 26, 3291–3310 (2007)

    CAS  PubMed  Google Scholar 

  18. Levin-Salomon, V., Kogan, K., Ahn, N. G., Livnah, O. & Engelberg, D. Isolation of intrinsically active (MEK-independent) variants of the ERK family of mitogen-activated protein (MAP) kinases. J. Biol. Chem. 283, 34500–34510 (2008)

    CAS  PubMed  PubMed Central  Google Scholar 

  19. Chu, Y., Solski, P. A., Khosravi-Far, R., Der, C. J. & Kelly, K. The mitogen-activated protein kinase phosphatases PAC1, MKP-1, and MKP-2 have unique substrate specificities and reduced activity in vivo toward the ERK2 sevenmaker mutation. J. Biol. Chem. 271, 6497–6501 (1996)

    CAS  PubMed  Google Scholar 

  20. Turski, M. L. & Thiele, D. J. New roles for copper metabolism in cell proliferation, signaling, and disease. J. Biol. Chem. 284, 717–721 (2009)

    CAS  PubMed  PubMed Central  Google Scholar 

  21. Tsai, C. Y., Finley, J. C., Ali, S. S., Patel, H. H. & Howell, S. B. Copper influx transporter 1 is required for FGF, PDGF and EGF-induced MAPK signaling. Biochem. Pharmacol. 84, 1007–1013 (2012)

    CAS  PubMed  PubMed Central  Google Scholar 

  22. Nose, Y., Kim, B. E. & Thiele, D. J. Ctr1 drives intestinal copper absorption and is essential for growth, iron metabolism, and neonatal cardiac function. Cell Metab. 4, 235–244 (2006)

    CAS  PubMed  Google Scholar 

  23. Dankort, D. et al. A new mouse model to explore the initiation, progression, and therapy of BRAFV600E-induced lung tumors. Genes Dev. 21, 379–384 (2007)

    CAS  PubMed  PubMed Central  Google Scholar 

  24. Solit, D. B. & Rosen, N. Resistance to BRAF inhibition in melanomas. N. Engl. J. Med. 364, 772–774 (2011)

    CAS  PubMed  Google Scholar 

  25. Wagle, N. et al. Dissecting therapeutic resistance to RAF inhibition in melanoma by tumor genomic profiling. J. Clin. Oncol. 29, 3085–3096 (2011)

    CAS  PubMed  PubMed Central  Google Scholar 

  26. Bridgewater, J. D. & Vachet, R. W. Metal-catalyzed oxidation reactions and mass spectrometry: the roles of ascorbate and different oxidizing agents in determining Cu-protein-binding sites. Anal. Biochem. 341, 122–130 (2005)

    CAS  PubMed  Google Scholar 

  27. Kelly, S. M., Jess, T. J. & Price, N. C. How to study proteins by circular dichroism. Biochim. Biophys. Acta 1751, 119–139 (2005)

    CAS  PubMed  Google Scholar 

  28. Niesen, F. H., Berglund, H. & Vedadi, M. The use of differential scanning fluorimetry to detect ligand interactions that promote protein stability. Nature Protocols 2, 2212–2221 (2007)

    CAS  PubMed  Google Scholar 

  29. Augustine, C. K. et al. Sorafenib, a multikinase inhibitor, enhances the response of melanoma to regional chemotherapy. Mol. Cancer Ther. 9, 2090–2101 (2010)

    CAS  PubMed  Google Scholar 

  30. Lampson, B. L. et al. Targeting eNOS in pancreatic cancer. Cancer Res. 72, 4472–4482 (2012)

    CAS  PubMed  PubMed Central  Google Scholar 

  31. O’Hayer, K. M. & Counter, C. M. A genetically defined normal human somatic cell system to study Ras oncogenesis in vivo and in vitro. Methods Enzymol. 407, 637–647 (2006)

    PubMed  Google Scholar 

  32. Boehm, J. S. et al. Integrative genomic approaches identify IKBKE as a breast cancer oncogene. Cell 129, 1065–1079 (2007)

    CAS  PubMed  Google Scholar 

  33. O’Hayer, K. M., Brady, D. C. & Counter, C. M. ELR+ CXC chemokines and oncogenic Ras-mediated tumorigenesis. Carcinogenesis 30, 1841–1847 (2009)

    PubMed  PubMed Central  Google Scholar 

  34. Zipfel, P. A. et al. Ral activation promotes melanomagenesis. Oncogene 29, 4859–4864 (2010)

    CAS  PubMed  PubMed Central  Google Scholar 

  35. Scholl, F. A., Dumesic, P. A. & Khavari, P. A. Mek1 alters epidermal growth and differentiation. Cancer Res. 64, 6035–6040 (2004)

    CAS  PubMed  Google Scholar 

  36. Johannessen, C. M. et al. COT drives resistance to RAF inhibition through MAP kinase pathway reactivation. Nature 468, 968–972 (2010)

    CAS  PubMed  PubMed Central  ADS  Google Scholar 

  37. Bric, A. et al. Functional identification of tumor-suppressor genes through an in vivo RNA interference screen in a mouse lymphoma model. Cancer Cell 16, 324–335 (2009)

    CAS  PubMed  PubMed Central  Google Scholar 

  38. Kahsai, A. W. et al. Multiple ligand-specific conformations of the β2-adrenergic receptor. Nature Chem. Biol. 7, 692–700 (2011)

    CAS  Google Scholar 

  39. Nobles, K. N. et al. Distinct phosphorylation sites on the β2-adrenergic receptor establish a barcode that encodes differential functions of beta-arrestin. Sci. Signal. 4, ra51 (2011)

    CAS  PubMed  PubMed Central  Google Scholar 

  40. 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)

    CAS  PubMed  Google Scholar 

  41. Haas, W. et al. Optimization and use of peptide mass measurement accuracy in shotgun proteomics. Mol. Cell. Proteomics 5, 1326–1337 (2006)

    CAS  PubMed  Google Scholar 

  42. Hamad, N. M. et al. Distinct requirements for Ras oncogenesis in human versus mouse cells. Genes Dev. 16, 2045–2057 (2002)

    CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank M. McMahon, C. Cronin, E. Johnson, A. Stewart, D. S. Tyler and D. G. Kirsch for reagents, and C. Cronin, L. E. Crose, A. M. Jaeger, M. A. Luftig, E. Johnson, D. F. Kashatus, B. L. Lampson, J. P. Madigan, N. I. Nicely, Y. Nose, C. W. Pemble, N. L. K. Pershing, A. Stewart and J. D. Weyandt for technical support, discussions and/or review of the manuscript. This work was supported by National Institutes of Health grants CA178145 (D.C.B.), HL075443 (Proteomic Core K.X.), DK074192 (D.J.T.), CA094184 and CA172104 (C.M.C.), the Structural Genomics Consortium (Welcome Trust 092809/Z/10/Z), FP7 grant 278568 ‘PRIMES’ (S.K. and A.C.), the Stewart Trust (C.M.C.), the Edward Spiegel Fund of the Lymphoma Foundation (C.M.C.), and donations made in memory of Linda Woolfenden (C.M.C.).

Author information

Authors and Affiliations

Authors

Contributions

Experiments were performed by D.C.B., M.S.C., M.L.T., G.A.H., X.Y. and K.X. All authors contributed to the study design. The manuscript was written by D.C.B. and C.M.C. with contributions by all authors.

Corresponding author

Correspondence to Christopher M. Counter.

Ethics declarations

Competing interests

A provisional patent application (D.C.B., M.L.T., D.J.T. and C.M.C.) has been filed regarding the use of Cu chelators for the treatment of BRAF and RAS mutation-positive cancers and a phase I melanoma trial investigating a copper chelator has been registered with clinicaltrials.gov (NCT02068079) with one of the authors serving as a co-investigator (C.M.C.).

Extended data figures and tables

Extended Data Figure 1 CuSO4stimulates MEK1/2 kinase activity in vitro.

a, c, Detection of the amount of in vitro phosphorylated (P) recombinant GST-tagged kinase-inactive ERK2K54R protein by recombinant GST-tagged MEK1 in the presence, when indicated, of 2.5 molar equivalents of CuSO4 (Cu), AgNO3 (Ag), FeNH2SO4 (Fe), NiSO4 (Ni) or ZnSO4 (Zn) (a) or recombinant GST-tagged MEK2 in the presence, when indicated, of 2.5 μM CuSO4 and/or 50 μM TTM (c). Total (T) levels of ERK2, MEK1 and MEK2 serve as loading controls. b, Immunoblot detection of the amount of recombinant GST-tagged MEK2 protein bound to a resin charged with (Cu) or without (−) Cu. Input serves as a loading control. Gel images are representative of two technical replicates.

Extended Data Figure 2 Genetic ablation of Ctr1 decreases BRAFV600E-mediated cell growth and tumorigenesis.

a, Cell growth, as measured by staining with crystal violet, of BRAFV600E-transformed Ctr1+/+ (black circles) or Ctr1−/− (red squares) MEFs (plated in sextuplicate) over a period of three days. Representative of three independent experiments using the same cells. b, c, Representative resected tumours (scale bar, 1 cm) at 20 days after injection (b) and Kaplan–Meier analysis of percentage of mice with tumour volume at least 1.0 cm3 versus time (c) of mice (n = 8) injected with BRAFV600E-transformed Ctr1+/+ (black line) or Ctr1−/− (red line) MEFs. Results were compared using a one-tailed unpaired t-test (a) or a Mantel–Cox test (c). Four asterisks, P < 0.0001.

Extended Data Figure 3 Identification of Cu-binding mutants of MEK1 that decrease ERK1/2 phosphorylation.

a, Immunoblot detection of the amount of HA-tagged wild-type MEK1 and an example of one MEK1 mutant tested (H188A) that bound to a Cu-charged resin. Input serves as a loading control. b, Immunoblot detection of the amount of phosphorylated (P) and/or total (T) ERK1/2 or HA-MEK1 protein in Ctr1+/+ MEFs stably expressing HA-tagged wild-type MEK1 or an example of one MEK1 mutant tested (H188A). c, Summary of whether the indicated MEK1 point mutants did (YES) or did not (NO) show a decrease in binding to the Cu-charged resin or show a decrease in the levels of phosphorylated (P) ERK1/2 when stably expressed in Ctr1+/+ MEFs. Gel images are representative of two technical replicates.

Extended Data Figure 4 Amino acids in MEK1 identified to be oxidized by the MCO reaction followed by MS/MS.

Representative annotated MS/MS fragmentation spectra for five indicated MEK1-derived peptides containing oxidized residues highlighted in red: a, MEK1H87 and MEK1M94; b, MEK1H100; c, MEK1H188; d, MEK1M230; e, MEK1H239. The peak heights are the relative abundances of the corresponding fragmentation ions, with the annotation of the identified matched amino-terminus-containing ions (b ions) in blue and the carboxy-terminus-containing ions (y ions) in red. For clarity, only the major identified peaks are labelled. f, Amino-acid sequence of human MEK1 with the peptides identified by MS/MS underlined (red, trypsin digest; blue, chymotrypsin digest). Amino acids oxidized only in the presence of H2O2 in one to three independent MCO reactions are denoted in red. Boxes enclose amino acids that when mutated to alanine decreased both the binding of MEK1 to a Cu-charged resin and the phosphorylation of cellular ERK2 (from Extended Data Fig. 3c).

Extended Data Figure 5 Alignment of the amino-acid sequences of MEK1, MEK2, MEK5 and MEK1–MEK5.

The amino-acid sequences of human MEK1, MEK2, MEK5 and the MEK1–MEK5 chimaeric protein (without the DD mutation) aligned using Clustal W. Black letters, amino acids; coloured letters, the four amino acids mutated in MEK1CBM to decrease Cu binding (blue, conserved between MEK1, MEK2 and MEK5; red, conserved only between MEK1 and MEK2). Dashes indicate gaps in the alignment.

Extended Data Figure 6 Protein purification and biochemical analysis of wild-type and CBM versions of MEK1.

a, Coomassie blue detection of the amount of wild-type or CBM mutant purified recombinant GST-tagged MEK1 protein in the absence or presence of precision protease for cleavage of GST. bd, Circular dichroism spectra at increasing wavelengths (b), thermal denaturation monitored at 222 nm at increasing temperature (c), and differential scanning fluorimetry at increasing temperature (d, left) and the average estimated melting temperature (d, right) of purified recombinant MEK1WT (black circles and line) and MEK1CBM (red squares and line). Data are representative of two technical replicates.

Extended Data Figure 7 Tumorigenic growth of NRAS mutation-positive human melanoma cancer cell lines on knockdown of CTR1.

a, b, RT–PCR detection of the amount of endogenous CTR1 and GAPDH mRNA (a) and mean tumour volume (±s.e.m.) versus time (b) of mice (n = 3) injected with the NRAS mutation-positive (NRASQ61L) human melanoma cell lines DM598 and DM792 stably infected with a retrovirus expressing either a scramble (SCRAM) shRNA (black circles) or CTR1 shRNA (red squares). Results were compared using a one-tailed unpaired t-test (b). Four asterisks, P < 0.0001.

Extended Data Figure 8 Detection of Cre-mediated recombination and weight measurements of AdCre-treated Ctr1+/+versus Ctr1flox/floxBP mice.

a, PCR detection of BrafCA/+, Trp53flox/flox and Ctr1flox/flox recombined alleles from matched tail samples (T) and lung tumour cell lines (C) generated from indicated genotypes. Alleles are indicated by arrowheads as follows: black, WT; red, flox; blue, null; orange, BrafCA; green, BrafV600E. b, Box-and-whiskers plot of weight of Ctr1+/+ versus Ctr1flox/flox BP mice (n = 30) one month after intranasal treatment with AdCre. Results were compared using a one-tailed unpaired t-test (b). Four asterisks, P < 0.0001.

Extended Data Figure 9 TTM does not decrease the weight of mice with tumours.

Mean weight (±s.e.m.) over time of mice (n = 4) injected with BRAFV600E-transformed MEFs and treated with vehicle (black circles) or TTM (red squares).

Extended Data Figure 10 Graphical representation of Cu regulation of BRAFV600E-mediated signalling and tumorigenesis.

Inactivation of the signalling pathway is denoted in grey and dashed lines, gain-of-function mutations are denoted in green, and loss-of-function mutations are denoted in red.

PowerPoint slides

Rights and permissions

Reprints and permissions

About this article

Cite this article

Brady, D., Crowe, M., Turski, M. et al. Copper is required for oncogenic BRAF signalling and tumorigenesis. Nature 509, 492–496 (2014). https://doi.org/10.1038/nature13180

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nature13180

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