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:

Accumulation of 8,9-unsaturated sterols drives oligodendrocyte formation and remyelination

Abstract

Regeneration of myelin is mediated by oligodendrocyte progenitor cells—an abundant stem cell population in the central nervous system (CNS) and the principal source of new myelinating oligodendrocytes. Loss of myelin-producing oligodendrocytes in the CNS underlies a number of neurological diseases, including multiple sclerosis and diverse genetic diseases1,2,3. High-throughput chemical screening approaches have been used to identify small molecules that stimulate the formation of oligodendrocytes from oligodendrocyte progenitor cells and functionally enhance remyelination in vivo4,5,6,7,8,9,10. Here we show that a wide range of these pro-myelinating small molecules function not through their canonical targets but by directly inhibiting CYP51, TM7SF2, or EBP, a narrow range of enzymes within the cholesterol biosynthesis pathway. Subsequent accumulation of the 8,9-unsaturated sterol substrates of these enzymes is a key mechanistic node that promotes oligodendrocyte formation, as 8,9-unsaturated sterols are effective when supplied to oligodendrocyte progenitor cells in purified form whereas analogous sterols that lack this structural feature have no effect. Collectively, our results define a unifying sterol-based mechanism of action for most known small-molecule enhancers of oligodendrocyte formation and highlight specific targets to propel the development of optimal remyelinating therapeutics.

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: Imidazoles inhibit CYP51 to enhance oligodendrocyte formation.
Fig. 2: Small-molecule inhibition of CYP51, TM7SF2, or EBP enhances oligodendrocyte formation via accumulation of 8,9-unsaturated sterols.
Fig. 3: Inhibition of TM7SF2 and EBP is a unifying mechanism for many small-molecule enhancers of oligodendrocyte formation.
Fig. 4: Accumulation of 8,9-unsaturated sterols enhances remyelination in vivo and in human brain spheroids.

Similar content being viewed by others

References

  1. Goldman, S. A., Nedergaard, M. & Windrem, M. S. Glial progenitor cell-based treatment and modeling of neurological disease. Science 338, 491–495 (2012).

    Article  ADS  PubMed  PubMed Central  CAS  Google Scholar 

  2. Fancy, S. P. et al. Overcoming remyelination failure in multiple sclerosis and other myelin disorders. Exp. Neurol. 225, 18–23 (2010).

    Article  PubMed  CAS  Google Scholar 

  3. Franklin, R. J. & Ffrench-Constant, C. Remyelination in the CNS: from biology to therapy. Nat. Rev. Neurosci. 9, 839–855 (2008).

    Article  PubMed  CAS  Google Scholar 

  4. Najm, F. J. et al. Drug-based modulation of endogenous stem cells promotes functional remyelination in vivo. Nature 522, 216–220 (2015).

    Article  ADS  PubMed  PubMed Central  CAS  Google Scholar 

  5. Deshmukh, V. A. et al. A regenerative approach to the treatment of multiple sclerosis. Nature 502, 327–332 (2013).

    Article  ADS  PubMed  PubMed Central  CAS  Google Scholar 

  6. Mei, F. et al. Micropillar arrays as a high-throughput screening platform for therapeutics in multiple sclerosis. Nat. Med. 20, 954–960 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  7. Mei, F. et al. Identification of the kappa-opioid receptor as a therapeutic target for oligodendrocyte remyelination. J. Neurosci. 36, 7925–7935 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  8. Huang, J. K. et al. Retinoid X receptor gamma signaling accelerates CNS remyelination. Nat. Neurosci. 14, 45–53 (2011).

    Article  PubMed  CAS  Google Scholar 

  9. Gonzalez, G. A. et al. Tamoxifen accelerates the repair of demyelinated lesions in the central nervous system. Sci. Rep. 6, 31599 (2016).

    Article  ADS  PubMed  PubMed Central  CAS  Google Scholar 

  10. Lariosa-Willingham, K. D. et al. A high throughput drug screening assay to identify compounds that promote oligodendrocyte differentiation using acutely dissociated and purified oligodendrocyte precursor cells. BMC Res. Notes 9, 419 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  11. Korade, Z. et al. The effect of small molecules on sterol homeostasis: measuring 7-dehydrocholesterol in DHCR7-deficient Neuro2a cells and human fibroblasts. J. Med. Chem. 59, 1102–1115 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  12. Giera, M., Müller, C. & Bracher, F. Analysis and experimental inhibition of distal cholesterol biosynthesis. Chromatographia 78, 343–358 (2015).

    Article  CAS  Google Scholar 

  13. Giera, M., Plössl, F. & Bracher, F. Fast and easy in vitro screening assay for cholesterol biosynthesis inhibitors in the post-squalene pathway. Steroids 72, 633–642 (2007).

    Article  PubMed  CAS  Google Scholar 

  14. Mir, F. & Le Breton, G. C. A novel nuclear signaling pathway for thromboxane A2 receptors in oligodendrocytes: evidence for signaling compartmentalization during differentiation. Mol. Cell. Biol. 28, 6329–6341 (2008).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  15. Jachak, G. R. et al. Silicon incorporated morpholine antifungals: design, synthesis, and biological evaluation. ACS Med. Chem. Lett. 6, 1111–1116 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  16. Zhang, L. et al. Selective targeting of mutant adenomatous polyposis coli (APC) in colorectal cancer. Sci. Transl. Med. 8, 361ra140 (2016).

    Article  PubMed  CAS  Google Scholar 

  17. DeBrabander, J., Shay, J. W., Wang, W., Nijhawan, D. & Theodoropoulos, P. Targeting emopamil binding protein (EBP) with small molecules that induce an abnormal feedback response by lowering endogenous cholesterol biosynthesis. US patent application US 2016/0313302 A1 (2016).

  18. Saher, G. et al. Therapy of Pelizaeus-Merzbacher disease in mice by feeding a cholesterol-enriched diet. Nat. Med. 18, 1130–1135 (2012).

    Article  PubMed  CAS  Google Scholar 

  19. Byskov, A. G., Andersen, C. Y. & Leonardsen, L. Role of meiosis activating sterols, MAS, in induced oocyte maturation. Mol. Cell. Endocrinol. 187, 189–196 (2002).

    Article  PubMed  CAS  Google Scholar 

  20. Grøndahl, C. Oocyte maturation. Basic and clinical aspects of in vitro maturation (IVM) with special emphasis of the role of FF-MAS. Dan. Med. Bull. 55, 1–16 (2008).

    PubMed  Google Scholar 

  21. Canfrán-Duque, A. et al. Atypical antipsychotics alter cholesterol and fatty acid metabolism in vitro. J. Lipid Res. 54, 310–324 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  22. Moebius, F. F. et al. Pharmacological analysis of sterol delta8-delta7 isomerase proteins with [3H]ifenprodil. Mol. Pharmacol. 54, 591–598 (1998).

    Article  PubMed  CAS  Google Scholar 

  23. Gylling, H. et al. Tamoxifen and toremifene lower serum cholesterol by inhibition of delta 8-cholesterol conversion to lathosterol in women with breast cancer. J. Clin. Oncol. 13, 2900–2905 (1995).

    Article  PubMed  CAS  Google Scholar 

  24. Bechler, M. E., Byrne, L. & Ffrench-Constant, C. CNS myelin sheath lengths are an intrinsic property of oligodendrocytes. Curr. Biol. 25, 2411–2416 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  25. Lee, S. et al. A culture system to study oligodendrocyte myelination processes using engineered nanofibers. Nat. Methods 9, 917–922 (2012).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  26. Mi, S. et al. Promotion of central nervous system remyelination by induced differentiation of oligodendrocyte precursor cells. Ann. Neurol. 65, 304–315 (2009).

    Article  PubMed  CAS  Google Scholar 

  27. Madhavan, M. et al. Induction of myelinating oligodendrocytes in human cortical spheroids. Nat. Methods https://doi.org/10.1038/s41592-018-0081-4 (2018).

  28. Miron, V. E. et al. Statin therapy inhibits remyelination in the central nervous system. Am. J. Pathol. 174, 1880–1890 (2009).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  29. Klopfleisch, S. et al. Negative impact of statins on oligodendrocytes and myelin formation in vitro and in vivo. J. Neurosci. 28, 13609–13614 (2008).

    Article  PubMed  CAS  Google Scholar 

  30. Saher, G. et al. High cholesterol level is essential for myelin membrane growth. Nat. Neurosci. 8, 468–475 (2005).

    Article  PubMed  CAS  Google Scholar 

  31. Godefroi, E. F., Heeres, J., Van Cutsem, J. & Janssen, P. A. The preparation and antimycotic properties of derivatives of 1-phenethylimidazole. J. Med. Chem. 12, 784–791 (1969).

    Article  PubMed  CAS  Google Scholar 

  32. Giera, M., Renard, D., Plössl, F. & Bracher, F. Lathosterol side chain amides: a new class of human lathosterol oxidase inhibitors. Steroids 73, 299–308 (2008).

    Article  PubMed  CAS  Google Scholar 

  33. Najm, F. J. et al. Rapid and robust generation of functional oligodendrocyte progenitor cells from epiblast stem cells. Nat. Methods 8, 957–962 (2011).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  34. Miller, T. E. et al. Transcription elongation factors represent in vivo cancer dependencies in glioblastoma. Nature 547, 355–359 (2017).

    Article  ADS  PubMed  PubMed Central  CAS  Google Scholar 

  35. Honda, A. et al. Highly sensitive quantification of key regulatory oxysterols in biological samples by LC-ESI-MS/MS. J. Lipid Res. 50, 350–357 (2009).

    Article  PubMed  CAS  Google Scholar 

  36. Warrilow, A. G., Parker, J. E., Kelly, D. E. & Kelly, S. L. Azole affinity of sterol 14α-demethylase (CYP51) enzymes from Candida albicans and Homo sapiens. Antimicrob. Agents Chemother. 57, 1352–1360 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  37. Folch, J., Lees, M. & Sloane Stanley, G. H. A simple method for the isolation and purification of total lipides from animal tissues. J. Biol. Chem. 226, 497–509 (1957).

    PubMed  CAS  Google Scholar 

  38. Pink, J. J. & Jordan, V. C. Models of estrogen receptor regulation by estrogens and antiestrogens in breast cancer cell lines. Cancer Res. 56, 2321–2330 (1996).

    PubMed  CAS  Google Scholar 

  39. Labarca, C. & Paigen, K. A simple, rapid, and sensitive DNA assay procedure. Anal. Biochem. 102, 344–352 (1980).

    Article  PubMed  CAS  Google Scholar 

  40. Bosch, D. G. et al. NR2F1 mutations cause optic atrophy with intellectual disability. Am. J. Hum. Genet. 94, 303–309 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

Download references

Acknowledgements

This work was supported by National Institutes of Health grant NS095280 (R.H.M., P.J.T), Conrad N. Hilton Foundation Pilot Innovator in MS Award (D.J.A.), Mallinckrodt Foundation Grant Award (D.J.A.), Mt. Sinai Health Care Foundation, philanthropic support from the Peterson, Fakhouri, Long, Goodman, Geller, Judge, and Weidenthal families, and unrestricted support from the CWRU School of Medicine. Z.H., M.S.E., K.C.A., Z.S.N., and J.L.S. were supported by the CWRU Medical Scientist Training Program (NIH T32 GM007250). Z.H. was also supported by NIH TL1 TR000441. Additional support was provided by the Small-Molecule Drug Development, Proteomics, and Translational Research Shared Resources of the Case Comprehensive Cancer Center (P30 CA043703). We acknowledge use of the Leica SP8 confocal microscope in the Light Microscopy Imaging Facility at CWRU made available through the Office of Research Infrastructure (NIH-ORIP) Shared Instrumentation Grant (S10 OD016164). We thank M. Drumm, T. Miller, B. Karl, O. Iyoha-Bello, J. Pink., P. Conrad, R. Lee, X. Li, D. Schlatzer, K. Polak, Janssen Pharmaceutica N.V., CXR Biosciences, ThermoFisher, Avanti Polar Lipids, and the P. Scacheri laboratory for technical assistance and discussion.

Author information

Authors and Affiliations

Authors

Contributions

Z.H., D.A., M.S.E., M.M., Z.S.N., K.C.A., H.E.S., M.A.T., and D.J.A. evaluated the effects of small molecules and genetic manipulations on oligodendrocyte formation in vitro. Z.H., D.A., I.B., M.A.T., F.B., and D.J.A. performed and analysed sterol profiling experiments in OPCs in vitro. D.C.F., Y.F., P.J.T., and D.J.A. performed high-throughput screening. Z.H., I.B., H.E.S., E.G., M.M., M.K., R.H.M., P.J.T., and D.J.A. evaluated the in vivo efficacy of small molecules on remyelination and sterol levels. Z.H. and J.L.S. profiled nuclear hormone receptors. Z.H., M.M., and Z.S.N. performed experiments on human cortical spheroids. J.J., W.K.W., M.G., and F.B. synthesized and purified sterol reagents. Z.H., D.A., P.J.T. and D.J.A. analysed all data and wrote the manuscript. All authors provided intellectual input, edited and approved the final manuscript.

Corresponding author

Correspondence to Drew J. Adams.

Ethics declarations

Competing interests

D.J.A., P.J.T., Z.H., D.A., M.S.E. and R.H.M. are inventors on patents and patent applications that relate to this work and have been licensed to Convelo Therapeutics, Inc., which seeks to develop remyelinating therapeutics. D.J.A. and P.J.T. hold equity in Convelo Therapeutics, Inc. and receive consulting income from Convelo Therapeutics, Inc. After resubmission of this work, D.C.F. became an employee of Convelo Therapeutics, Inc.

Additional information

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 Expanded cholesterol synthesis pathway diagram.

The cascade cyclization of squalene epoxide, catalysed by lanosterol synthase (LSS), provides the first sterol, lanosterol. Processing of lanosterol to cholesterol can proceed via the Kandutsch–Russell and/or Bloch pathways, which use the same enzymes and process substrates that vary only in the presence or absence of the C24 double bond. Intermediates in blue have been confirmed in our GC–MS-based sterol profiling assay using authentic standards. Sterol 14-reductase activity in mouse is shared by two genes, TM7SF2 and LBR. Consistent with past reports21, inhibition of sterol 14-reductase activity can lead to accumulation of the expected upstream intermediate (FF-MAS) or 14-dehydrozymostenol, also known as cholesta-8,14-dien-3-β-ol. Green indicates enzyme targets and small molecules whose inhibition promotes oligodendrocyte formation.

Extended Data Fig. 2 CYP51 is the functional target by which imidazole antifungals enhance oligodendrocyte formation.

a, Azole molecules with varying degrees of potency for mammalian CYP51 inhibition. Throughout, green labels indicate molecules considered active, while red labels indicate inactive molecules. b, Percentage of MBP+ oligodendrocytes generated from a second, independent derivation of OPCs (OPC-1) at 72 h following treatment with the indicated concentrations of azoles. n = 4 wells per condition except DMSO (n = 24), with >1,000 cells analysed per well. c, GC–MS-based quantification of lanosterol levels in a second derivation of OPCs (OPC-1) treated for 24 h with the indicated azoles at 2.5 μM. n = 2 wells per condition. d, e, GC–MS-based quantification of cholesterol levels in OPCs (OPC-5 and OPC-1) treated for 24 h with the indicated azoles at 2.5 μM. n = 2 wells per condition. f, g, GC–MS-based quantification of lanosterol levels in OPCs (OPC-5, OPC-1) treated for 24 h with the indicated doses of ketoconazole. n = 2 wells per condition. Concentrations shown in f and g mirror those shown in b and Fig. 1c. h, Percentage of MBP+ oligodendrocytes generated from mouse primary OPCs at 72 h following treatment with the indicated imidazole antifungals at 3 μM. n = 4 wells per condition, with >1,000 cells analysed per well. i, GC–MS-based quantification of lanosterol levels in mouse primary OPCs treated for 24 h with the indicated imidazole antifungals at 3 μM. n = 2 wells per condition. j, Assessment of oligodendrocyte formation using an alternative image quantification metric, fold increase in total neurite length. Re-analysis of data shown in Fig. 1c. n = 4 wells per condition except DMSO (n = 24), with >1,000 cells analysed per well. k, Percentage of oligodendrocytes generated from OPCs at 72 h following treatment with ketoconazole (2.5 μM) as measured by PLP1 immunostaining. Left, OPC-5; right, OPC-1. n = 8 wells per condition, with >1,000 cells analysed per well. l, LC–MS-based quantification of lanosterol levels in OPC-5 cells treated for 24 h with ketoconazole at 2.5 μM. n = 2 wells per condition. m, CYP51 mRNA levels measured by RT–qPCR following 96-h treatment with non-targeting or CYP51-targeting pools of cell-permeable siRNAs. n = 2 wells per condition. n, GC–MS-based quantification of lanosterol levels in OPC-1 cells treated for 96 h with the indicated pooled siRNA reagents. n = 2 wells per condition. o, Percentage of MBP+ oligodendrocytes generated from a second, independent batch of OPCs (OPC-1) at 72 h following treatment with the indicated reagents. n = 3 wells per condition, with >1,000 cells analysed per well. p, Percentage of MBP+ oligodendrocytes generated from an independent derivation of OPCs at 72 h following treatment with exogenous lanosterol. n = 4 wells per condition except DMSO and ketoconazole (n = 8), with >1,000 cells analysed per well. q, Representative images of OPC-5 cells treated for 72 h with the indicated siRNA reagents and lanosterol. Nuclei are labelled with DAPI (blue), and oligodendrocytes are indicated by immunostaining for MBP (green). Scale bar, 100 μm. All bar graphs indicate mean ± s.d.; b, d, h, i, k, l, o and p are representative of two independent experiments, and all findings have been confirmed in a second independent derivation of OPCs (Fig. 1).

Source Data

Extended Data Fig. 3 Effect of small-molecule inhibition of the cholesterol biosynthesis pathway on enhancing oligodendrocyte formation.

a, GC–MS-based quantification of sterol levels in OPCs (OPC-5) treated for 24 h with the indicated inhibitors of cholesterol biosynthesis. Left, cholesterol; right, desmosterol. n = 2 wells per condition. Inhibitors were used at the following doses unless otherwise noted: mevastatin, ketoconazole, MGI-39, 2.5 μM; YM53601, 2 μM; Ro 48-8071, amorolfine, TASIN-1, 100 nM; AY9944, 200 nM. b, GC–MS-based quantification of sterol levels in a second derivation of OPCs (OPC-1). Left, cholesterol; right, desmosterol. n = 2 wells per condition. c, GC–MS-based quantification of the sterol intermediates expected to accumulate following treatment of OPCs with the indicated inhibitors of cholesterol biosynthesis for 24 h. n = 2 wells per condition. d, GC–MS-based quantification of the sterol intermediates expected to accumulate following treatment of a second derivation of OPCs (OPC-1) with the indicated inhibitors of cholesterol biosynthesis for 24 h. n = 2 wells per condition. In c and d, no accumulation of other sterol intermediates indicative of off-target effects within the cholesterol pathway were observed (see Source Data). e, Representative images of OPC-5 cells treated for 72 h with the indicated small molecules. All treatments are at the highest concentration shown in Fig. 2b. Scale bar, 100 μm. f, Percentage of MBP+ oligodendrocytes generated from a second batch of OPCs (OPC-1) at 72 h following treatment with the indicated cholesterol pathway inhibitors. n = 4 wells per condition, except DMSO, n = 24, with >1,000 cells analysed per well. g, Percentage of MBP+ oligodendrocytes generated from mouse primary OPCs at 72 h following treatment with the indicated cholesterol pathway inhibitors at 300 nM. n = 4 wells per condition, except DMSO, n = 8, with >1,000 cells analysed per well. h, GC–MS-based quantification of sterol intermediate levels in mouse primary OPCs treated for 24 h with the indicated inhibitors of cholesterol biosynthesis at 300 nM. Left, 14-dehydrozymostenol levels following treatment with amorolfine; right, zymostenol levels following treatment with TASIN-1. n = 2 wells per condition. i, j, GC–MS-based quantification of sterol intermediate levels in OPC-5 (i) and OPC-1 (j) cells treated for 24 h with the indicated doses of inhibitors of cholesterol biosynthesis. Left, 14-dehydrozymostenol levels following treatment with amorolfine; right, zymostenol levels following treatment with TASIN-1. n = 2 wells per condition. Concentrations shown in i mirror those shown in f. All bar graphs indicate mean ± s.d., and a, c, eh are representative of two independent experiments.

Source Data

Extended Data Fig. 4 Effect of independent chemical-genetic and genetic modulators of CYP51, sterol 14 reductase and EBP on oligodendrocyte formation and cholesterol biosynthesis.

a, d, g, Percentage of MBP+ oligodendrocytes generated from two independent derivation of OPCs at 72 h following treatment with the indicated concentrations of medroxyprogesterone acetate (a), 2-methyl ketoconazole (d) or TASIN-449 (g). n = 4 wells per condition, except DMSO, n = 12 in a, d. In g, for OPC-5, n = 4 except DMSO, n = 7; for OPC-1, n = 3 except DMSO, n = 6. b, e, h, GC–MS-based quantification of sterol levels in two independent derivations of OPCs treated for 24 h with medroxyprogesterone acetate at 10 μM (b), 2-methyl ketoconazole at 2.5 μM (e) and TASIN-449 at the indicated concentrations (h). n = 2 wells per condition. c, f, Rat CYP51 enzymatic activity following treatment with varying concentrations of medroxyprogesterone acetate (c) and 2-methyl ketoconazole (f) as measured by LC–MS-based quantification of the CYP51 product FF-MAS. n = 2 independent enzymatic assays. i, Percentage of MBP+ oligodendrocytes generated from OPCs (OPC-5) infected with lentivirus expressing Cas9 and an independent guide RNA targeting EBP (see also Fig. 2c). Eight wells per condition, with >1,000 cells analysed per well. Two-tailed Student’s t-test, *P = 0.0009. j, Functional validation of CRISPR-based targeting of EBP with a second sgRNA using GC–MS-based quantification of zymostenol levels. n = 2 wells per condition. k, EBP mRNA levels measured by RT–qPCR in OPCs (OPC-5) infected with lentivirus expressing Cas9 and either of two guide RNAs targeting EBP. One well per condition, with results validated in an independent experiment. l, Representative images of the oligodendrocyte formation assay shown in Fig. 2c. Nuclei are labelled with DAPI (blue), and oligodendrocytes are indicated by immunostaining for MBP (green). Scale bar, 100 μm. All bar graphs indicate mean ± s.d., and a, d, g, i, k are representative of two independent experiments.

Source Data

Extended Data Fig. 5 Effect of 8,9-unsaturated sterols on oligodendrocyte formation.

a, Percentage of MBP+ oligodendrocytes generated from OPCs (OPC-5) at 72 h following treatment with methyl β-cyclodextrin (1 mM) for 30 min at 37 °C. n = 8 wells per condition, with >1,000 cells analysed per well. b, GC–MS-based quantification of cholesterol (left) and desmosterol (right) in OPCs (OPC-5) treated with methyl β-cyclodextrin (Me-β-CD) at 1 mM or ketoconazole at 2.5 μM. n = 2 wells per condition. c, d, Percentage of MBP+ oligodendrocytes generated from OPC-1 (c) and OPC-5 cells (d) at 72 h following treatment with the indicated purified sterol intermediates. n = 4 wells per condition, except n = 8 for DMSO and ketoconazole, with >1,000 cells analysed per well. Green text highlights metabolites that accumulate after treatments that enhance oligodendrocyte formation (Fig. 2e, Extended Data Fig. 3c). e, Percentage of MBP+ oligodendrocytes generated from OPC1 cells at 72 h following treatment with MAS-412 and MAS-414. n = 4 wells per condition, with >1,000 cells analysed per well. f, Representative images of OPC5 cells treated for 72 h with DMSO, MAS-412, or MAS-414 (3 μM). Nuclei are labelled with DAPI (blue), and oligodendrocytes are indicated by immunostaining for MBP (green). Scale bar, 100 μm. g, Percentage of MBP+ oligodendrocytes generated from OPC-1 at 72 h following treatment with 2,2-dimethyl-zymosterol. n = 4 wells per condition except DMSO (n = 12), with >1,000 cells analysed per well. h, Representative images of OPC-5 cells treated for 72 h with vehicle and 2,2-dimethyl-zymosterol (2.5 μM). Nuclei are labelled with DAPI (blue), and oligodendrocytes are indicated by immunostaining for MBP (green). Scale bar, 100 μm. i, Percentage of MBP+ oligodendrocytes generated from OPC-5 (left) and OPC-1 (right) cells at 72 h following treatment with FF-MAS or T-MAS. n = 4 wells per condition except DMSO and ketoconazole (n = 8), with >1,000 cells analysed per well. j, Percentage of MBP+ oligodendrocytes generated from OPC-5 and OPC-1 OPCs at 72 h following treatment with the indicated concentrations of cholesterol. n = 8 wells per condition, with >1,000 cells analysed per well. k, l, Percentage of MBP+ oligodendrocytes generated from OPC-5 and OPC-1 cells at 72 h following treatment with the indicated concentrations of sterols that are structurally identical aside from the presence or absence of the 8,9 double bond (structures in o). n ≥ 3 wells per condition (see dot plots as replicate values vary by condition), with >1,000 cells analysed per well. m, Percentage of MBP+ oligodendrocytes generated from OPCs (OPC-5) at 72 h following treatment with the indicated small molecules or combinations of small molecules (ketoconazole, 2.5 μM; Ro 48-8071, 11 nM; liothyronine, 3 μM). n = 3 wells per condition, except DMSO n = 11, ketoconazole n = 13, liothyronine n = 8 & liothyronine + Ro 48-8071 n = 4, with >1,000 cells analysed per well. n, GC–MS-based quantification of lanosterol levels in OPCs (OPC-5) treated for 24 h with the indicated small molecules or combinations of small molecules at concentrations stated in m. n = 2 wells per condition. o, Structures of zymostenol, 8,9-dehydrocholesterol, 5α-cholestanol, and cholesterol. p, Total cell number as measured by counting of DAPI+ nuclei in the experiment presented in m. q, r, Percentage of MBP+ oligodendrocytes generated from OPCs (OPC5 and OPC-1) at 72 h following treatment with the indicated small molecules or combinations of small molecules in two independent batches of OPCs (ketoconazole, 2.5 μM; MAS412, 5 μM). In q, n = 16 for DMSO, 8 for ketoconazole, and 4 for remaining bars. In r, n = 8 wells per condition. s, Luciferase reporter assays were used to assess whether 2,2-dimethylzymosterol (5 μM), ketoconazole (2.5 μM), and TASIN-1 (250 nM) modulate human ERα, GR, LXRβ, NFkB, NRF2, PGR, PPARδ, PPARγ, RARα, RARγ, RXRα, RXRβ, TRα, TRβ and VDR transcriptional activity in agonist mode and ERRα, RORα and RORγ in inverse-agonist mode. n = 2 wells per condition and n = 3 wells per positive control condition. t, Effects of sterols (2,2-dimethylzymosterol 5 μM, FF-MAS 10 μM) and small molecules (ketoconazole 2.5 μM, TASIN-1 100 nM) on the NR2F1-mediated activation of a NGFI-A promoter driven luciferase reporter. n = 2 wells per condition. u, Effects of 2,2-dimethylzymosterol (5 μM) on NR2C2-mediated activation of a NGFI-A promoter driven luciferase reporter in comparison to cells transfected with reporter only, untreated, or treated with a previously reported positive control (all-trans retinoic acid, ATRA, 5 μM). n = 2 wells per condition. v, LSS, DHCR7, LDLR mRNA levels measured by RT–qPCR following 24 h treatment with DMSO, mevastatin (2.5 μM), Ro 48-8071 (500 nM), ketoconazole (2.5 μM), TASIN-1 (100 nM), or amorolfine (100 nM). n = 2 wells. All bar graphs indicate mean ± s.d., and an and tv are representative of two independent experiments.

Source Data

Extended Data Fig. 6 Inhibiting CYP51, TM7SF2 and EBP is a unifying mechanism for many small-molecule enhancers of oligodendrocyte formation identified by high-throughput screening.

a, Percentage of MBP+ oligodendrocytes (relative to DMSO control wells) generated from OPCs (OPC-1 derivation) at 72 h following treatment with a library of 3,000 bioactive small molecules, each at 2 μM. Each dot represents the result for one small molecule in the library. Red, imidazole antifungals; blue, clemastine; green, EPZ005687, the top novel hit molecule (Extended Data Fig. 7). b, c, Percentage of MBP+ oligodendrocytes generated from OPCs (left: OPC-5; right: OPC-1) at 72 h following treatment with ketoconazole, nine top molecules identified by bioactives screening (green), and nine randomly chosen library members (red) at a uniform dose of 5 μM. n = 4 wells per condition except DMSO and ketoconazole, n = 12 wells, with >1,000 cells analysed per well. d, GC–MS-based quantification of zymosterol, zymostenol, and 14-dehydrozymostenol levels in a second batch of OPCs treated for 24 h with the indicated screening hits and randomly chosen library members at 2 μM. n = 1; for validation in a second derivation of OPCs, see Fig. 3a. Molecules are clustered by enzyme targeted (top labels). e, Percentage of MBP+ oligodendrocytes generated from OPCs at 72 h following treatment with the indicated doses of fulvestrant, one of the top 10 HTS hits. n = 4 wells per condition except DMSO, n = 12, with >1,000 cells analysed per well. f, GC–MS-based quantification of lanosterol levels in OPCs treated for 24 h with fulvestrant at 2 μM. n = 2 wells per condition. gi, GC–MS-based quantification of metabolite levels in OPCs treated for 24 h with the indicated previously reported enhancers of oligodendrocyte formation at the following doses: benztropine, 2 μM; clemastine, 1 μM; tamoxifen, 100 nM; U50488, 5 μM; bexarotene, 1 μM; liothyronine, 3 μM. n = 2 wells per condition. j, k, Percentage of MBP+ oligodendrocytes generated from OPCs (OPC-5 left, OPC-1 right) at 72 h following treatment with the indicated previously reported enhancers of oligodendrocyte formation. n = 4 wells per condition, except DMSO n = 20 for OPC-5 and n = 12 for OPC-1, with >1,000 cells analysed per well. All doses are in μM. l, Representative images of OPCs treated for 72 h with the indicated small molecules. All treatments in l are at the highest concentration shown in j. Scale bar, 100 μm. m, Structures of muscarinic receptor antagonists used in this study. n, q, Percentage of MBP+ oligodendrocytes generated from OPCs (OPC-5: top, OPC-1: bottom) at 72 h following treatment with ketoconazole or the indicated muscarinic receptor modulators at 2 μM, the concentration used during screening. n = 4 wells per condition except DMSO and ketoconazole, n = 8, with >1,000 cells analysed per well. o, GC–MS-based quantification of three metabolite levels in OPC-5 OPCs treated for 24 h with U50488 (5 μM) or the indicated muscarinic receptor modulators (2 μM). Left, zymostenol; centre, cholesterol; right, desmosterol. n = 2 wells per condition. p, Heatmap indicating inhibition of muscarinic receptor isoforms M1, M3, and M5 by the indicated small molecules (2 μM) assayed using GeneBLAzer NFAT-bla CHO-K1 cells. n = 2 wells per condition. r, GC–MS-based quantification of three metabolite levels in OPC-1 OPCs treated for 24 h with clemastine (1 μM) or the indicated muscarinic receptor modulators at 2 μM. n = 2 wells per condition. Left, zymostenol; centre, zymosterol; right, cholesterol. Sigma H127, p-fluorohexahydro-sila-difenidol. All bar graphs indicate mean ± s.d., and b, c, e, i, j, k, n, q are representative of two independent experiments.

Source Data

Extended Data Fig. 7 Effect of selective oestrogen receptor modulators and EZH2 inhibitors on cellular EBP function and oligodendrocyte formation.

a, Structures of selective oestrogen receptor modulators used in this study. b, Effects of ospemifene and toremifene on the oestrogen-dependent growth of T47D cells. n = 3 wells per condition. c, d, Percentage of MBP+ oligodendrocytes generated from two independent batches of OPCs at 72 h following treatment with ospemifene and toremifene. n = 4 wells per condition except DMSO and ketoconazole, n = 8, with >1,000 cells analysed per well. e, Representative images of OPCs treated for 72 h with the indicated small molecules. All molecules were used at 300 nM. Scale bar, 100 μm. f, g, GC–MS-based quantification of two metabolite levels in OPCs treated for 24 h with ospemifene and toremifene at 300 nM. Left, zymostenol; right, cholesterol. n = 2 wells per condition. h, Percentage of MBP+ oligodendrocytes generated from two independent batches of OPCs at 72 h following treatment with tamoxifen and 4-hydroxytamoxifen. Left, OPC-5; right, OPC-1. n = 4 wells per condition, except DMSO, n = 6 for OPC-1 (right). i, Effects of tamoxifen and 4-hydroxytamoxifen on the oestrogen-dependent growth of T47D cells. n = 3 wells per condition. j, GC–MS-based quantification of zymostenol (left axis) and zymosterol levels (right axis) in OPC-5 and OPC-1 treated 24 h with tamoxifen and 4-hydroxytamoxifen at the indicated concentrations. n = 2 wells per condition. k, Percentage of MBP+ oligodendrocytes generated from OPCs at 72 h following treatment with the indicated structurally analogous EZH2 inhibitors. n = 4 wells per condition, except DMSO, n = 12, with >1,000 cells analysed per well. l, Percentage of MBP+ oligodendrocytes generated from a second batch of OPCs at 72 h following treatment with the indicated structurally analogous EZH2 inhibitors. n = 4 wells per condition, except DMSO, n = 12, with >1,000 cells analysed per well. m, Percentage of MBP+ oligodendrocytes generated from mouse primary OPCs at 72 h following treatment with EPZ005687. n = 4 wells per condition, except DMSO, n = 12, with >1,000 cells analysed per well. n, Structure of EPZ005687 and structurally analogous EZH2 inhibitors. o, Representative images of OPCs treated for 72 h with the indicated EZH2 inhibitors. All treatments are at 2 μM. Scale bar, 100 μm. p, GC–MS-based quantification of two sterol intermediates following treatment of OPCs with the indicated EZH2 inhibitors at 1 μM for 24 h. Left, zymostenol; right, zymosterol. n = 2 wells per condition. q, GC–MS-based quantification of two sterol intermediates following treatment of a second derivation of OPCs with the indicated EZH2 inhibitors at 1 μM for 24 h. Left, zymostenol; right, zymosterol. n = 2 wells per condition. r, GC–MS-based quantification of two sterol intermediates following treatment of mouse primary OPCs with EPZ005687 at 2 μM for 24 h. Left, zymostenol; right, zymosterol. n = 2 wells per condition. All bar graphs indicate mean ± s.d., and c, d, h, ko, r are representative of two independent experiments.

Source Data

Extended Data Fig. 8 Effect of combinations of small-molecule treatments on oligodendrocyte formation, and ability of oligodendrocytes to track along and wrap electrospun microfibres after single small-molecule treatments.

a, b, Percentage of MBP+ oligodendrocytes generated from OPCs (left, OPC-1; right, OPC-5) at 72 h following treatment with the indicated combinations of liothyronine and enhancers of oligodendrocyte formation. Unless noted, the following concentrations were used: ketoconazole, 2.5 μM; benztropine, 2 μM; clemastine 2 μM; tamoxifen 200 nM; liothyronine, 3 μM. n = 4 wells per treatment condition, with >1,000 cells analysed per well. Lio, liothyronine. c, d, Percentage of MBP+ oligodendrocytes generated from OPCs at 72 h following treatment with the indicated combinations of ketoconazole and enhancers of oligodendrocyte formation. n = 4 wells per treatment condition, with >1,000 cells analysed per well. e, Representative images of OPCs treated for 72 h with the indicated small molecules. Small-molecule concentrations are as in a. Scale bar, 100 μm. f, Fold-increase in MBP+ oligodendrocytes following plating of OPCs (OPC-5) onto microfibres and treatment for 14 days with the indicated pathway modulators. n = 2 wells per condition, except DMSO, n = 4. g, In an independent experiment, OPCs (OPC-5) were plated onto microfibres, treated with small molecules for 4 days, and fixed and stained after 14 days. The extent to which MBP+ oligodendrocytes tracked along the microfibre substrate was measured. n = 2 wells per condition. h, Total DAPI+ cell number in the experiment in g. i, Representative images highlighting tracking along the microfibre substrate. Each image is a montage of four separate images within the same well. Green, MBP. Scale bar, 100 μm. j, High-resolution images of MBP+ oligodendrocytes tracking along microfibres. Green, MBP; blue, DAPI. Ketoconazole, 2.5 μM. Scale bar, 50 μm. k, Confocal imaging of OPCs seeded onto aligned microfibres and treated for 14 days with ketoconazole (2.5 μM). The plane of the cross-section is highlighted in yellow and the cross-section, in which green fluorescence appears to encircle several microfibres, is shown in the bottom panel. Green, MBP; blue, DAPI. All bar graphs indicate mean ± s.d., and ad are representative of two independent experiments.

Extended Data Fig. 9 Effect of oligodendrocyte-enhancing small molecules on sterol levels in human cells and human cortical spheroids.

a, Representative images of toluidine blue-stained sections of LPC-lesioned dorsal spinal cord from mice treated for 8 days with ifenprodil (10 mg per kg) or tamoxifen (2 mg per kg). Scale bar, 20 μm. b, GC–MS-based quantification of three metabolite levels in human glioma cells (GBM528) treated for 24 h with the indicated small molecules at the following concentrations: tamoxifen, 100 nM; clemastine, 2 μM; ifenprodil, 2 μM; ketoconazole, 2.5 μM; amorolfine, 100 nM. Left, lanosterol; centre, zymostenol; right, 14-dehydrozymostenol. n = 2 wells per condition. c, GC–MS-based quantification of three metabolite levels in two independent batches of human cortical spheroids treated for 24 h with the indicated small molecules at 2 μM. Left, lanosterol; centre, zymostenol; right, zymosterol. n = 3 spheroids per condition; representative of two independent experiments.

Source Data

Extended Data Fig. 10 Twenty-seven small molecules and nine purified 8,9-unsaturated sterols shown to enhance the formation of oligodendrocytes.

a, Schematic showing the proposed mechanism of action for enhanced oligodendrocyte formation by diverse small molecules. b, Molecules that enhance oligodendrocyte formation are grouped by enzyme inhibited (GC–MS analysis in OPCs): CYP51, top; sterol 14-reductase, centre; EBP, bottom. c, Purified 8,9-unsaturated sterols that enhance oligodendrocyte formation.

Supplementary information

Supplementary Tables

This file contains Supplementary Tables 1–3.

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

Hubler, Z., Allimuthu, D., Bederman, I. et al. Accumulation of 8,9-unsaturated sterols drives oligodendrocyte formation and remyelination. Nature 560, 372–376 (2018). https://doi.org/10.1038/s41586-018-0360-3

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41586-018-0360-3

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