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:

MFSD12 mediates the import of cysteine into melanosomes and lysosomes

Abstract

Dozens of genes contribute to the wide variation in human pigmentation. Many of these genes encode proteins that localize to the melanosome—the organelle, related to the lysosome, that synthesizes pigment—but have unclear functions1,2. Here we describe MelanoIP, a method for rapidly isolating melanosomes and profiling their labile metabolite contents. We use this method to study MFSD12, a transmembrane protein of unknown molecular function that, when suppressed, causes darker pigmentation in mice and humans3,4. We find that MFSD12 is required to maintain normal levels of cystine—the oxidized dimer of cysteine—in melanosomes, and to produce cysteinyldopas, the precursors of pheomelanin synthesis made in melanosomes via cysteine oxidation5,6. Tracing and biochemical analyses show that MFSD12 is necessary for the import of cysteine into melanosomes and, in non-pigmented cells, lysosomes. Indeed, loss of MFSD12 reduced the accumulation of cystine in lysosomes of fibroblasts from patients with cystinosis, a lysosomal-storage disease caused by inactivation of the lysosomal cystine exporter cystinosin7,8,9. Thus, MFSD12 is an essential component of the cysteine importer for melanosomes and lysosomes.

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: MelanoIP enables the rapid isolation of pure melanosomes.
Fig. 2: MFSD12 is necessary to maintain melanosomal cystine levels and produce cysteinyldopas.
Fig. 3: MFSD12 is necessary to maintain lysosomal cystine and cysteine levels.
Fig. 4: MFSD12 is necessary and probably sufficient for the import of cysteine into melanosomes and lysosomes.

Similar content being viewed by others

Data availability

Fig. 3a and Extended Data Fig. 3a were generated from FANTOM5 expression data, accessed via the Human Protein Atlas20,21 from https://www.proteinatlas.org/about/assays+annotation#fantom. Raw values from this accession are included in the source data for Fig. 3a and Extended Data Fig. 3a. Unique biological materials in the form of plasmids available from Addgene. Unique biological materials in the form of cell lines are available from the authors by request. Source data are provided with this paper.

References

  1. Basrur, V. et al. Proteomic analysis of early melanosomes: identification of novel melanosomal proteins. J. Proteome Res. 2, 69–79 (2003).

    Article  ADS  CAS  Google Scholar 

  2. Sturm, R. A. Molecular genetics of human pigmentation diversity. Hum. Mol. Genet. 18 (R1), R9–R17 (2009).

    Article  CAS  Google Scholar 

  3. Adhikari, K. et al. A GWAS in Latin Americans highlights the convergent evolution of lighter skin pigmentation in Eurasia. Nat. Commun. 10, 358 (2019).

    Article  ADS  Google Scholar 

  4. Crawford, N. G. et al. Loci associated with skin pigmentation identified in African populations. Science 358, eaan8433 (2017).

    Article  Google Scholar 

  5. D’Alba, L. & Shawkey, M. D. Melanosomes: biogenesis, properties, and evolution of an ancient organelle. Physiol. Rev. 99, 1–19 (2019).

    Article  Google Scholar 

  6. Prota, G. Melanins and Melanogenesis (Academic, 1992).

  7. Gahl, W. A., Bashan, N., Tietze, F., Bernardini, I. & Schulman, J. D. Cystine transport is defective in isolated leukocyte lysosomes from patients with cystinosis. Science 217, 1263–1265 (1982).

    Article  ADS  CAS  Google Scholar 

  8. Jonas, A. J., Smith, M. L. & Schneider, J. A. ATP-dependent lysosomal cystine efflux is defective in cystinosis. J. Biol. Chem. 257, 13185–13188 (1982).

    CAS  PubMed  Google Scholar 

  9. Town, M. et al. A novel gene encoding an integral membrane protein is mutated in nephropathic cystinosis. Nat. Genet. 18, 319–324 (1998).

    Article  CAS  Google Scholar 

  10. Watabe, H., Kushimoto, T., Valencia, J. C. & Hearing, V. J. in Current Protocols in Cell Biology (eds J. S. Bonifacino et al.) (John Wiley & Sons, 2005).

  11. Abu-Remaileh, M. et al. Lysosomal metabolomics reveals V-ATPase- and mTOR-dependent regulation of amino acid efflux from lysosomes. Science 358, 807–813 (2017).

    Article  ADS  CAS  Google Scholar 

  12. Chen, W. W., Freinkman, E., Wang, T., Birsoy, K. & Sabatini, D. M. Absolute quantification of matrix metabolites reveals the dynamics of mitochondrial metabolism. Cell 166, 1324–1337 (2016).

    Article  CAS  Google Scholar 

  13. Ray, G. J. et al. A PEROXO-tag enables rapid isolation of peroxisomes from human cells. iScience 23, 101109 (2020).

    Article  ADS  CAS  Google Scholar 

  14. Bruder, J. M. et al. Melanosomal dynamics assessed with a live-cell fluorescent melanosomal marker. PLoS ONE 7, e43465 (2012).

    Article  ADS  CAS  Google Scholar 

  15. Diment, S., Eidelman, M., Rodriguez, G. M. & Orlow, S. J. Lysosomal hydrolases are present in melanosomes and are elevated in melanizing cells. J. Biol. Chem. 270, 4213–4215 (1995).

    Article  CAS  Google Scholar 

  16. Bissig, C., Rochin, L. & van Niel, G. PMEL amyloid fibril formation: the bright steps of pigmentation. Int. J. Mol. Sci. 17, 1438 (2016).

    Article  Google Scholar 

  17. Reddy, V. S., Shlykov, M. A., Castillo, R., Sun, E. I. & Saier, M. H. Jr. The major facilitator superfamily (MFS) revisited. FEBS J. 279, 2022–2035 (2012).

    Article  CAS  Google Scholar 

  18. Bloom, J. L. & Falconer, D. S. ‘Grizzled’, a mutant in linkage group X of the mouse. Genet. Res. 7, 159–167 (1966).

    Article  Google Scholar 

  19. Potterf, S. B. et al. Cysteine transport in melanosomes from murine melanocytes. Pigment Cell Res. 12, 4–12 (1999).

    Article  CAS  Google Scholar 

  20. Kawaji, H., Kasukawa, T., Forrest, A., Carninci, P. & Hayashizaki, Y. The FANTOM5 collection, a data series underpinning mammalian transcriptome atlases in diverse cell types. Sci. Data 4, 170113 (2017).

    Article  CAS  Google Scholar 

  21. Uhlén, M. et al. Tissue-based map of the human proteome. Science 347, 1260419 (2015).

    Article  Google Scholar 

  22. Pisoni, R. L., Acker, T. L., Lisowski, K. M., Lemons, R. M. & Thoene, J. G. A cysteine-specific lysosomal transport system provides a major route for the delivery of thiol to human fibroblast lysosomes: possible role in supporting lysosomal proteolysis. J. Cell Biol. 110, 327–335 (1990).

    Article  CAS  Google Scholar 

  23. Gahl, W. A., Thoene, J. G. & Schneider, J. A. Cystinosis. N. Engl. J. Med. 347, 111–121 (2002).

    Article  Google Scholar 

  24. Oshima, R. G., Rhead, W. J., Thoene, J. G. & Schneider, J. A. Cystine metabolism in human fibroblasts. Comparison of normal, cystinotic, and gamma-glutamylcysteine synethetase-deficient cells. J. Biol. Chem. 251, 4287–4293 (1976).

    CAS  PubMed  Google Scholar 

  25. Sato, H., Tamba, M., Ishii, T. & Bannai, S. Cloning and expression of a plasma membrane cystine/glutamate exchange transporter composed of two distinct proteins. J. Biol. Chem. 274, 11455–11458 (1999).

    Article  CAS  Google Scholar 

  26. Wilbrandt, W. & Rosenberg, T. The concept of carrier transport and its corollaries in pharmacology. Pharmacol. Rev. 13, 109–183 (1961).

    CAS  PubMed  Google Scholar 

  27. Behnke, J., Eskelinen, E.-L., Saftig, P. & Schröder, B. Two dileucine motifs mediate late endosomal/lysosomal targeting of transmembrane protein 192 (TMEM192) and a C-terminal cysteine residue is responsible for disulfide bond formation in TMEM192 homodimers. Biochem. J. 434, 219–231 (2011).

    Article  CAS  Google Scholar 

  28. Lloyd, J. B. Disulphide reduction in lysosomes. The role of cysteine. Biochem. J. 237, 271–272 (1986).

    Article  CAS  Google Scholar 

  29. Mego, J. L. Role of thiols, pH and cathepsin D in the lysosomal catabolism of serum albumin. Biochem. J. 218, 775–783 (1984).

    Article  CAS  Google Scholar 

  30. Tsui, C. K. et al. CRISPR–cas9 screens identify regulators of antibody-drug conjugate toxicity. Nat. Chem. Biol. 15, 949–958 (2019).

    Article  CAS  Google Scholar 

  31. Gahl, W. A. et al. Cysteamine therapy for children with nephropathic cystinosis. N. Engl. J. Med. 316, 971–977 (1987).

    Article  CAS  Google Scholar 

  32. Thoene, J. G., Oshima, R. G., Crawhall, J. C., Olson, D. L. & Schneider, J. A. Cystinosis. Intracellular cystine depletion by aminothiols in vitro and in vivo. J. Clin. Invest. 58, 180–189 (1976).

    Article  CAS  Google Scholar 

  33. Mujahid, N. et al. A UV-independent topical small-molecule approach for melanin production in human skin. Cell Rep. 19, 2177–2184 (2017).

    Article  CAS  Google Scholar 

  34. Djoumbou-Feunang, Y. et al. CFM-ID 3.0: significantly improved ESI-MS/MS prediction and compound identification. Metabolites 9, 72 (2019).

    Article  CAS  Google Scholar 

  35. Wishart, D. S. et al. HMDB: the Human Metabolome Database. Nucleic Acids Res. 35, D521–D526 (2007).

    Article  CAS  Google Scholar 

  36. Guan, X., Hoffman, B., Dwivedi, C. & Matthees, D. P. A simultaneous liquid chromatography/mass spectrometric assay of glutathione, cysteine, homocysteine and their disulfides in biological samples. J. Pharm. Biomed. Anal. 31, 251–261 (2003).

    Article  CAS  Google Scholar 

  37. Martin, G. B. et al. Development of a mass spectrometry method for the determination of a melanoma biomarker, 5-S-cysteinyldopa, in human plasma using solid phase extraction for sample clean-up. J. Chromatogr. A 1156, 141–148 (2007).

    Article  CAS  Google Scholar 

  38. Ito, S. & Prota, G. A facile one-step synthesis of cysteinyldopas using mushroom tyrosinase. Experientia 33, 1118–1119 (1977).

    Article  CAS  Google Scholar 

  39. Graham, J. M. Isolation of lysosomes from tissues and cells by differential and density gradient centrifugation. Curr. Protoc. Cell Biol. Ch. 3, Unit 3.6 (2001).

    Google Scholar 

  40. Goldman, R. & Kaplan, A. Rupture of rat liver lysosomes mediated by l-amino acid esters. Biochim. Biophys. Acta 318, 205–216 (1973).

    Article  CAS  Google Scholar 

  41. Verdon, Q. et al. SNAT7 is the primary lysosomal glutamine exporter required for extracellular protein-dependent growth of cancer cells. Proc. Natl Acad. Sci. USA 114, E3602–E3611 (2017).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank P. Budde, M. Abu-Remaileh, W. W. Chen, L. Bar-Peled and J. G. Bryan, as well as all current members of the Sabatini laboratory for helpful insights. This work was supported by grants from the Leo Foundation (LF18057) and NIH (R01 CA103866, R01 CA129105 and R01 AI047389), fellowship support from the NIH (NRSA F31 CA228241-01) to C.H.A., Marshall Plan Foundation to A.K.T., HHMI XROP to B.C., and NSF (2016197106) to K.J.C. D.M.S. is an investigator of the Howard Hughes Medical Institute and an American Cancer Society Research Professor.

Author information

Authors and Affiliations

Authors

Contributions

C.H.A. and D.M.S. initiated the project and designed the research plan. C.H.A. performed the experiments and analysed the data with help from A.K.T., B.C. and K.J.C. The LC–MS platform was operated by S.H.C., T.K. and C.A.L., who also had a critical role in method development for LC–MS-based assays for cysteine and cysteinyldopas. C.H.A. wrote the manuscript and D.M.S. edited it.

Corresponding author

Correspondence to David M. Sabatini.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Peer review information Nature thanks Edmund Kunji 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 MelanoIP analysis detects changes in Tyr dependent melanosomal metabolites.

a, Schematic of melanin synthesis. The common pathway elements for eumelanin and pheomelanin synthesis have a grey backdrop. The brown and red backdrops highlight unique portions of eumelanin and pheomelanin synthesis, respectively. Enzymes proposed to catalyse each step are shown in green. Synthetic intermediates annotated and validated in biological samples in this study are in blue. b, Follow-up analysis with standard validated m/z and internal normalization of ‘proteogenic amino acids’ highlighted in untargeted metabolite profiling of wild-type and Tyr-knockout melanosomes (Fig. 1d). Amino acids are presented in order of increasing retention time (n = 3 independently prepared extracts, aP = 3.9 × 10−2, bP = 2.0 × 10−3, cP = 6.5 × 10−3, dP = 3.8 × 10−2, eP = 1.9 × 10−2). Error bars are mean ± s.e.m., P values by two-sided Student’s t-test.

Source data

Extended Data Fig. 2 In vitro synthesis and biological detection of cysteinyldopas.

a, Cysteinyldopas were synthesized according to an adapted protocol from Ito and Prota, 197738. Two species, distinguished by retention time, were generated at the expected m/z for cysteinyldopas. It has been shown that 5′-cysteinyldopa is produced in greater abundance than 2’-cysteinyldopa in this reaction. Taking MS1 peak intensity to approximate abundance, we putatively annotate the ‘Minor Isomer’ as 2’ substituted, and the ‘Major Isomer’ as 5′ substituted. b, Mirror plot of ddMS2 data comparing 2’- and 5′-cysteinyldopa in synthetic cysteinyldopas. c,d, Mirror plots of ddMS2 peaks displaying similarities in ddMS2 spectra of 2’- and 5′-cysteinyldopa species in biological samples (B16F10 extracts) and synthetic standards.

Source data

Extended Data Fig. 3 MFSD12 maintains lysosomal cystine in non-pigmented cells.

a, FANTOM5 CAGE profiling data accessed via Human Protein Atlas20,21. Six representative pigmentation genes, including MFSD12, are shown. b, Metabolite profiling of LysoIP samples from HEK 293T cells comparing lysosomes from wild-type and MFSD12-knockout cells. ‘Accumulates upon inhibition of:’ has been previously reported11 (n = 4 independently prepared metabolite extracts, aP = 7.0 × 10−4, bP = 3.0 × 10−3, cP = 4.1 × 10−2, dP = 4.2 × 10−2, eP = 1.7 × 10−4). c, Lentiviral shRNA knock-down of MFSD12 quantified via qPCR and normalized to ACTB levels (n = 3 assays on independently prepared cDNA libraries, aP = 1.97 × 10−3, bP = 3.0 × 10−3). Error bars are mean ± s.e.m., P values by two-sided Student’s t-test.

Source data

Extended Data Fig. 4 MFSD12 mediated cysteine transport is cysteine specific.

a, Test of lysosomal counter-transport. Lysosomes were purified by differential centrifugation and incubated with water or 1 mM cysteine methyl ester before washing, resuspension, and incubated for 5 min with 20 μM cysteine and trace amounts of [35S]cysteine (n = 3 independently performed assays per condition, aP = 2.5 × 10−3, bP = 3.3 × 10−2, NS = not significant). b, Lysosomal import of [14C]cystine. Lysosomes were purified by differential centrifugation and incubated for 10 min with 1 μM [14C]cystine, either untreated (Unreduced) or pre-treated with 10 mM DTT (Reduced, n = 6 independently performed assays per condition, aP = 2.1 × 10−7, bP = 1.3 × 10−6, cP = 3.8 × 10−8). c, Competition for [35S]cysteine transport. Lysosomes were purified by differential centrifugation and incubated for 10 min with 20 μM cysteine and trace amounts of [35S]cysteine with 500 μM competitor where indicated (n = 3 independently performed assays per condition, P values compare competition condition versus water control condition (red), aP = 2.7 × 10−4, bP = 2.5 × 10−4, cP = 3.2 × 10−4). Error bars are mean ± s.e.m., P values by two-sided Student’s t-test.

Source data

Supplementary information

Supplementary Figure 1

Uncropped blots for Fig 1c.

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

Adelmann, C.H., Traunbauer, A.K., Chen, B. et al. MFSD12 mediates the import of cysteine into melanosomes and lysosomes. Nature 588, 699–704 (2020). https://doi.org/10.1038/s41586-020-2937-x

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41586-020-2937-x

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: Translational Research

Sign up for the Nature Briefing: Translational Research newsletter — top stories in biotechnology, drug discovery and pharma.

Get what matters in translational research, free to your inbox weekly. Sign up for Nature Briefing: Translational Research