Article | Published:

Expansion of oligodendrocyte progenitor cells following SIRT1 inactivation in the adult brain

Nature Cell Biology volume 15, pages 614624 (2013) | Download Citation

Abstract

Oligodendrocytes—the myelin-forming cells of the central nervous system—can be regenerated during adulthood. In adults, new oligodendrocytes originate from oligodendrocyte progenitor cells (OPCs), but also from neural stem cells (NSCs). Although several factors supporting oligodendrocyte production have been characterized, the mechanisms underlying the generation of adult oligodendrocytes are largely unknown. Here we show that genetic inactivation of SIRT1, a protein deacetylase implicated in energy metabolism, increases the production of new OPCs in the adult mouse brain, in part by acting in NSCs. New OPCs produced following SIRT1 inactivation differentiate normally, generating fully myelinating oligodendrocytes. Remarkably, SIRT1 inactivation ameliorates remyelination and delays paralysis in mouse models of demyelinating injuries. SIRT1 inactivation leads to the upregulation of genes involved in cell metabolism and growth factor signalling, in particular PDGF receptor α (PDGFRα). Oligodendrocyte expansion following SIRT1 inactivation is mediated at least in part by AKT and p38 MAPK—signalling molecules downstream of PDGFRα. The identification of drug-targetable enzymes that regulate oligodendrocyte regeneration in adults could facilitate the development of therapies for demyelinating injuries and diseases, such as multiple sclerosis.

Access optionsAccess options

Rent or Buy article

Get time limited or full article access on ReadCube.

from$8.99

All prices are NET prices.

Accessions

Primary accessions

Gene Expression Omnibus

References

  1. 1.

    & The life, death, and replacement of oligodendrocytes in the adult CNS. J. Neurochem. 107, 1–19 (2008).

  2. 2.

    & Remyelination in the CNS: from biology to therapy. Nat. Rev. Neurosci. 9, 839–855 (2008).

  3. 3.

    et al. Experimental autoimmune encephalomyelitis mobilizes neural progenitors from the subventricular zone to undergo oligodendrogenesis in adult mice. Proc. Natl Acad. Sci. USA 99, 13211–13216 (2002).

  4. 4.

    et al. Progenitor cells of the adult mouse subventricular zone proliferate, migrate and differentiate into oligodendrocytes after demyelination. Eur. J. Neurosci. 11, 4357–4366 (1999).

  5. 5.

    et al. Origin of oligodendrocytes in the subventricular zone of the adult brain. J. Neurosci. 26, 7907–7918 (2006).

  6. 6.

    , , , & Epidermal growth factor induces the progeny of subventricular zone type B cells to migrate and differentiate into oligodendrocytes. Stem Cells 27, 2032–2043 (2009).

  7. 7.

    et al. Notch1 signaling plays a role in regulating precursor differentiation during CNS remyelination. Proc. Natl Acad. Sci. USA 106, 19162–19167 (2009).

  8. 8.

    et al. Sonic hedgehog–regulated oligodendrocyte lineage genes encoding bHLH proteins in the mammalian central nervous system. Neuron 25, 317–329 (2000).

  9. 9.

    , , , & A role for platelet-derived growth factor in normal gliogenesis in the central nervous system. Cell 53, 309–319 (1988).

  10. 10.

    , , & Platelet-derived growth factor regulates oligodendrocyte progenitor numbers in adult CNS andtheir response following CNS demyelination. Mol. Cell. Neurosci. 25, 252–262 (2004).

  11. 11.

    et al. Chordin-induced lineage plasticity of adult SVZ neuroblasts after demyelination. Nat. Neurosci. 13, 541–550 (2010).

  12. 12.

    et al. Age-dependent epigenetic control of differentiation inhibitors is critical for remyelination efficiency. Nat. Neurosci. 11, 1024–1034 (2008).

  13. 13.

    et al. Sirtuin 2, a mammalian homolog of yeast silent information regulator-2 longevity regulator, is an oligodendroglial protein that decelerates cell differentiation through deacetylating α-tubulin. J. Neurosci. 27, 2606–2616 (2007).

  14. 14.

    The logic linking protein acetylation and metabolism. Cell Metab. 14, 151–153 (2011).

  15. 15.

    , , & Redox state is a central modulator of the balance between self-renewal and differentiation in a dividing glial precursor cell. Proc. Natl Acad. Sci. USA 97, 10032–10037 (2000).

  16. 16.

    et al. Sirt1 contributes critically to the redox-dependent fate of neural progenitors. Nat. Cell Biol. 10, 385–394 (2008).

  17. 17.

    , & Nutrient availability regulates SIRT1 through a forkhead-dependent pathway. Science 306, 2105–2108 (2004).

  18. 18.

    et al. SIRT1 promotes the central adaptive response to diet restriction through activation of the dorsomedial and lateral nuclei of the hypothalamus. J. Neurosci. 30, 10220–10232 (2010).

  19. 19.

    , , , & Neuronal SIRT1 regulates endocrine and behavioral responses to calorie restriction. Genes Dev. 23, 2812–2817 (2009).

  20. 20.

    , , , & Sirt1 protects against high-fat diet-induced metabolic damage. Proc. Natl Acad. Sci. USA 105, 9793–9798 (2008).

  21. 21.

    et al. Specific SIRT1 activation mimics low energy levels and protects against diet-induced metabolic disorders by enhancing fat oxidation. Cell Metab. 8, 347–358 (2008).

  22. 22.

    et al. SirT1 gain of function increases energy efficiency and prevents diabetes in mice. Cell Metab. 8, 333–341 (2008).

  23. 23.

    et al. SIRT1 improves insulin sensitivity under insulin-resistant conditions by repressing PTP1B. Cell Metab. 6, 307–319 (2007).

  24. 24.

    et al. SIRT1 transgenic mice show phenotypes resembling calorie restriction. Aging Cell 6, 759–767 (2007).

  25. 25.

    et al. Nutrient control of glucose homeostasis through a complex of PGC-1α and SIRT1. Nature 434, 113–118 (2005).

  26. 26.

    et al. Sirt1 mediates neuroprotection from mutant huntingtin by activation of the TORC1 and CREB transcriptional pathway. Nat. Med. 18, 519–165 (2011).

  27. 27.

    , , & SIRT1 suppresses β-amyloid production by activating the α-secretase gene ADAM10. Cell 142, 320–332 (2010).

  28. 28.

    et al. SIRT1 Protects against α-synuclein aggregation by activating molecular chaperones. J. Neurosci. 32, 124–132 (2012).

  29. 29.

    et al. SIRT1 deacetylase protects against neurodegeneration in models for Alzheimer’s disease and amyotrophic lateral sclerosis. EMBO J. 26, 3169–3179 (2007).

  30. 30.

    et al. A novel pathway regulates memory and plasticity via SIRT1 and miR-134. Nature 466, 1105–1109 (2010).

  31. 31.

    et al. SIRT1 is essential for normal cognitive function and synaptic plasticity. J. Neurosci. 30, 9695–9707 (2010).

  32. 32.

    et al. Stress-dependent regulation of FOXO transcription factors by the SIRT1 deacetylase. Science 303, 2011–2015 (2004).

  33. 33.

    , , , & SIRT1 activation confers neuroprotection in experimental optic neuritis. Invest. Ophthalmol. Vis. Sci. 48, 3602–3609 (2007).

  34. 34.

    et al. Sirt1 overexpression in neurons promotes neurite outgrowth and cell survival through inhibition of the mTOR signaling. J. Neurosci. Res. 89, 1723–1736 (2011).

  35. 35.

    et al. SIRT1 regulates apoptosis and Nanog expression in mouse embryonic stem cells by controlling p53 subcellular localization. Cell Stem Cell 2, 241–251 (2008).

  36. 36.

    et al. Sirtuin 1 regulation of developmental genes during differentiation of stem cells. Proc. Natl Acad. Sci. USA 107, 13736–13741 (2010).

  37. 37.

    et al. miRNAs regulate SIRT1 expression during mouse embryonic stem cell differentiation and in adult mouse tissues. Aging 2, 415–431 (2010).

  38. 38.

    et al. Histone deacetylase SIRT1 modulates neuronal differentiation by its nuclear translocation. Proc. Natl Acad. Sci. USA 105, 15599–15604 (2008).

  39. 39.

    et al. Sir2 regulates skeletal muscle differentiation as a potential sensor of the redox state. Mol. Cell. 12, 51–62 (2003).

  40. 40.

    et al. SIRT1 deficiency compromises mouse embryonic stem cell hematopoietic differentiation, and embryonic and adult hematopoiesis in the mouse. Blood 117, 440–450 (2011).

  41. 41.

    et al. Cells previously identified as retinal stem cells are pigmented ciliary epithelial cells. Proc. Natl Acad. Sci. USA 106, 6685–6690 (2009).

  42. 42.

    , , , & Prox1 is required for granule cell maturation and intermediate progenitor maintenance during brain neurogenesis. PLoS Biol. 8, e1000460 (2010).

  43. 43.

    & Six3 is required for ependymal cell maturation. Development 138, 5291–5300 (2011).

  44. 44.

    et al. Pten deletion causes mTorc1-dependent ectopic neuroblast differentiation without causing uniform migration defects. Development 139, 3422–3431 (2012).

  45. 45.

    et al. Developmental defects and p53 hyperacetylation in Sir2 homolog (SIRT1)-deficient mice. Proc. Natl Acad. Sci. USA 100, 10794–10799 (2003).

  46. 46.

    , , , & A global double-fluorescent Cre reporter mouse. Genesis 45, 593–605 (2007).

  47. 47.

    , , & NG2-expressing glial progenitor cells: an abundant and widespread population of cycling cells in the adult rat CNS. Mol. Cell. Neurosci. 24, 476–488 (2003).

  48. 48.

    & Experimental disseminated encephalomyelitis in white mice. J. Exp. Med. 90, 213–224 (1949).

  49. 49.

    & Experimental allergic encephalomyelitis in inbred and outbred mice. J. Immunol. 110, 139–143 (1973).

  50. 50.

    , & A myelin oligodendrocyte glycoprotein peptide induces typical chronic experimental autoimmune encephalomyelitis in H-2b mice: fine specificity and T cell receptor V β expression of encephalitogenic T cells. Eur. J. Immunol. 25, 1951–1959 (1995).

  51. 51.

    & Relationship of iron to oligodendrocytes and myelination. Glia 17, 83–93 (1996).

  52. 52.

    , , , & Platelet-derived growth factor promotes division and motility and inhibits premature differentiation of the oligodendrocyte/type-2 astrocyte progenitor cell. Nature 333, 560–562 (1988).

  53. 53.

    , & Purification and characterization of adult oligodendrocyte precursor cells from the rat optic nerve. J. Neurosci. 18, 4627–4636 (1998).

  54. 54.

    , , & Transcriptional silencing and longevity protein Sir2 is an NAD-dependent histone deacetylase. Nature 403, 795–800 (2000).

  55. 55.

    , , , & PDGF and FGF-2 signaling in oligodendrocyte progenitor cells: regulation of proliferation and differentiation by multiple intracellular signaling pathways. Mol. Cell. Neurosci. 15, 314–329 (2000).

  56. 56.

    , , & Mechanisms of regulation of oligodendrocyte development by p38 mitogen-activated protein kinase. J. Neurosci. 30, 11011–11027 (2010).

  57. 57.

    et al. The protein kinase encoded by the Akt proto-oncogene is a target of the PDGF-activated phosphatidylinositol 3-kinase. Cell 81, 727–736 (1995).

  58. 58.

    et al. Akt-mediated survival of oligodendrocytes induced by neuregulins. J. Neurosci. 20, 7622–7630 (2000).

  59. 59.

    et al. The type III histone deacetylase Sirt1 is essential for maintenance of T cell tolerance in mice. J. Clin. Invest. 119, 3048–3058 (2009).

  60. 60.

    et al. Inhibition of SIRT1 catalytic activity increases p53 acetylation but does not alter cell survival following DNA damage. Mol. Cell. Biol. 26, 28–38 (2006).

  61. 61.

    et al. Sirtuin 2 inhibitors rescue α-synuclein-mediated toxicity in models of Parkinson’s disease. Science 317, 516–519 (2007).

  62. 62.

    et al. Class I histone deacetylase-selective novel synthetic inhibitors potently inhibit human tumor proliferation. Clin. Cancer Res. 10, 5271–5281 (2004).

  63. 63.

    & Sirtuin 1 in malignant transformation: friend or foe? Cancer Lett. 306, 10–14 (2011).

  64. 64.

    et al. MicroRNA and target protein patterns reveal physiopathological features of glioma subtypes. PLoS ONE 6, e20600 (2011).

  65. 65.

    et al. Asymmetry-defective oligodendrocyte progenitors are glioma precursors. Cancer Cell 20, 328–340 (2011).

  66. 66.

    et al. Non-stem cell origin for oligodendroglioma. Cancer Cell 18, 669–682 (2010).

  67. 67.

    et al. Mosaic analysis with double markers reveals tumor cell of origin in glioma. Cell 146, 209–221 (2011).

  68. 68.

    , , , & Oligodendrocyte progenitor cells can act as cell of origin for experimental glioma. Oncogene 28, 2266–2275 (2009).

  69. 69.

    et al. Oligodendrocyte lineage genes (OLIG) as molecular markers for human glial brain tumors. Proc. Natl Acad. Sci. USA 98, 10851–10856 (2001).

  70. 70.

    et al. Sirt1 improves healthy ageing and protects from metabolic syndrome-associated cancer. Nat. Commun. 1, 3 (2010).

  71. 71.

    et al. Reversal of paralysis and reduced inflammation from peripheral administration of β-amyloid in TH1 and TH17 versions of experimental autoimmune encephalomyelitis. Sci. Transl. Med. 4, 145ra105 (2012).

  72. 72.

    et al. FoxO3 regulates neural stem cell homeostasis. Cell Stem Cell 5, 527–539 (2009).

  73. 73.

    , , , & Functional genomic analysis of oligodendrocyte differentiation. J. Neurosci. 26, 10967–10983 (2006).

  74. 74.

    , , & Rank products: a simple, yet powerful, new method to detect differentially regulated genes in replicated microarray experiments. FEBS Lett. 573, 83–92 (2004).

Download references

Acknowledgements

We thank F. Alt for the generous gift of the Sirt1+/− and Sirt1lox/lox mice. We thank T. Palmer, T. Rando, and T. Wyss-Coray for helpful suggestions. We thank M. Winslow and J. Sage for critical review of the manuscript and discussion of the experiments. We are grateful to B. Benayoun and A. Morgan for their advice on the microarray analysis. We thank members of the A.B. laboratory for their invaluable discussion of the experiments and manuscript, in particular D. Leeman, J. Lim and A. Webb. This work was supported by NIH/NIA grants (R01 AG026648 and P01 AG036695), a California Institute for Regenerative Medicine grant, a Brain Tumour Society grant, an AFAR grant, the Glenn Foundation for Medical Research (A.B.) and the National Multiple Sclerosis Society (J.C.D., A.I. and B.A.B.—RG4059A8). V.A.R. was supported by an NSF graduate fellowship and an NINDS/NRSA graduate fellowship (5F31NS064600). E.A.P. was supported by an NSF graduate fellowship and an NIA/NRSA graduate fellowship (F31AG043232). L.S. and P.P.H were supported by the NIH (R01 NS055997), the National Multiple Sclerosis Society and the Guthy–Jackson Charitable Foundation. S.J.B. was supported by NIH P01CA096832. L.M.L.C. was supported by the Canadian Institutes of Health Research.

Author information

Author notes

    • Victoria A. Rafalski
    • , Jason C. Dugas
    •  & Lionel M. L. Chow

    Present addresses: Gladstone Institute of Neurological Disease, University of California, San Francisco, 1650 Owens Street, San Francisco, California 94158, USA (V.A.R.); Myelin Repair Foundation, 18809 Cox Avenue, Saratoga, California 95070, USA (J.C.D.); Cancer and Blood Diseases Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio 45229, USA (L.M.L.C.)

Affiliations

  1. Department of Genetics, 300 Pasteur Drive, Stanford University School of Medicine, Stanford, California 94305, USA

    • Victoria A. Rafalski
    • , Jamie O. Brett
    • , Duygu Ucar
    • , Elizabeth A. Pollina
    •  & Anne Brunet
  2. Neurosciences Program, Stanford University, Stanford, California 94305, USA

    • Victoria A. Rafalski
    • , Ben A. Barres
    •  & Anne Brunet
  3. Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, California 94305, USA

    • Peggy P. Ho
    •  & Lawrence Steinman
  4. Department of Neurobiology, Stanford University School of Medicine, Stanford, California 94305, USA

    • Jason C. Dugas
    • , Adiljan Ibrahim
    •  & Ben A. Barres
  5. Cancer Biology Program, Stanford University, Stanford, California 94305, USA

    • Elizabeth A. Pollina
    •  & Anne Brunet
  6. Department of Developmental Neurobiology, St Jude Children’s Research Hospital, Memphis, Tennessee 38105, USA

    • Lionel M. L. Chow
    •  & Suzanne J. Baker

Authors

  1. Search for Victoria A. Rafalski in:

  2. Search for Peggy P. Ho in:

  3. Search for Jamie O. Brett in:

  4. Search for Duygu Ucar in:

  5. Search for Jason C. Dugas in:

  6. Search for Elizabeth A. Pollina in:

  7. Search for Lionel M. L. Chow in:

  8. Search for Adiljan Ibrahim in:

  9. Search for Suzanne J. Baker in:

  10. Search for Ben A. Barres in:

  11. Search for Lawrence Steinman in:

  12. Search for Anne Brunet in:

Contributions

V.A.R. conceived and planned the study with the help of A.B. V.A.R. performed the experiments and wrote the paper with the help of A.B. P.P.H. and V.A.R. designed and performed the EAE experiments (Fig. 6) under the supervision of L.S. J.O.B. performed and analysed the RT–qPCR experiments (Fig. 7c and Supplementary Fig. S7a). D.U. helped with microarray analysis (Fig. 7a and Supplementary Fig. S6a and Table S1). The postnatal OPC experiments (Supplementary Fig. S5) were conceived and planned by J.C.D. and performed by A.I. under the supervision of B.A.B. E.A.P. performed the analysis of global histone acetylation (Supplementary Fig. S7b,c). L.M.L.C. and S.J.B. generated and characterized the NestinCreER mice. All authors discussed the results and commented on the manuscript.

Competing interests

The authors declare no competing financial interests.

Corresponding author

Correspondence to Anne Brunet.

Supplementary information

PDF files

  1. 1.

    Supplementary Information

    Supplementary Information

Excel files

  1. 1.

    Supplementary Table 1

    Supplementary Information

About this article

Publication history

Received

Accepted

Published

DOI

https://doi.org/10.1038/ncb2735

Further reading