Skip to main content

Advertisement

Log in

MeCP2 deficiency is associated with reduced levels of tubulin acetylation and can be restored using HDAC6 inhibitors

Journal of Molecular Medicine Aims and scope Submit manuscript

Abstract

Rett syndrome (RTT) is a severe neurodevelopmental disorder, predominantly caused by loss of function mutations in the X-linked methyl-CpG-binding protein 2 (MECP2) gene. Despite the genetic cause being known in the majority of cases, the pathophysiology of the neurological phenotype of RTT is largely unknown. Tubulin and the microtubule network play an essential role in neuronal function whereby the acetylation state of microtubules dictates the efficiency of neuronal migration and differentiation, synaptic targeting and molecular motor trafficking of mRNA, high-energy mitochondria and brain-derived neurotrophic factor (BDNF)-containing vesicles. Recent reports have shown perturbations in tubulin and microtubule dynamics in MeCP2-deficient cells, suggesting a link between the aberrations of these cellular entities and the neurobiology of RTT. We have interrogated the functional state of the microtubule network in fibroblasts derived from two patients with RTT as well as cortical neurons from a RTT mouse model and observed a reduction in acetylated α-tubulin and an increase in the tubulin-specific deacetylase, histone deacetylase 6 (HDAC6). Furthermore, we show that inhibition of HDAC6 by Tubastatin A can restore tubulin acetylation levels. We also demonstrate microtubule instability in the RTT patient fibroblasts in response to nocodazole, which is progressively ameliorated in a mutation-dependent manner by Tubastatin A. We conclude that Tubastatin A is capable of counteracting the microtubule defects observed in MeCP2-deficient cells, which could in turn lead to the restoration of molecular trafficking along the microtubules and thus could be a potentially new therapeutic option for RTT.

Key message

  • Cells from MeCP2-deficient cells show reduced levels of acetylated α-tubulin.

  • Cells from two patients and a RTT mouse model have increased levels of HDAC6 but not sirtuin 2 (SIRT2).

  • Inhibition of HDAC6 by Tubastatin A increases the in vitro acetylation of α-tubulin.

  • Inhibition of HDAC6 by Tubastatin A does not increase MECP2 expression.

  • Cells from two patients show microtubule instability, which is ameliorated by Tubastatin A.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Institutional subscriptions

Fig. 1
Fig. 2
Fig. 3
Fig. 4
Fig. 5

Similar content being viewed by others

References

  1. Xu WS, Parmigiani RB, Marks PA (2007) Histone deacetylase inhibitors: molecular mechanisms of action. Oncogene 26:5541–5552

    Article  CAS  PubMed  Google Scholar 

  2. Akella JS, Wloga D, Kim J, Starostina NG, Lyons-Abbott S, Morrissette NS, Dougan ST, Kipreos ET, Gaertig J (2010) MEC-17 is an alpha-tubulin acetyltransferase. Nature 467:218–222

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  3. Hubbert C, Guardiola A, Shao R, Kawaguchi Y, Ito A, Nixon A, Yoshida M, Wang XF, Yao TP (2002) HDAC6 is a microtubule-associated deacetylase. Nature 417:455–458

    Article  CAS  PubMed  Google Scholar 

  4. Bali P, Pranpat M, Bradner J, Balasis M, Fiskus W, Guo F, Rocha K, Kumaraswamy S, Boyapalle S, Atadja P et al (2005) Inhibition of histone deacetylase 6 acetylates and disrupts the chaperone function of heat shock protein 90: a novel basis for antileukemia activity of histone deacetylase inhibitors. J Biol Chem 280:26729–26734

    Article  CAS  PubMed  Google Scholar 

  5. Kovacs JJ, Murphy PJ, Gaillard S, Zhao X, Wu JT, Nicchitta CV, Yoshida M, Toft DO, Pratt WB, Yao TP (2005) HDAC6 regulates Hsp90 acetylation and chaperone-dependent activation of glucocorticoid receptor. Mol Cell 18:601–607

    Article  CAS  PubMed  Google Scholar 

  6. Dompierre JP, Godin JD, Charrin BC, Cordelieres FP, King SJ, Humbert S, Saudou F (2007) Histone deacetylase 6 inhibition compensates for the transport deficit in Huntington’s disease by increasing tubulin acetylation. J Neurosci Offi J Soc Neurosci 27:3571–3583

    Article  CAS  Google Scholar 

  7. Janke C, Kneussel M (2010) Tubulin post-translational modifications: encoding functions on the neuronal microtubule cytoskeleton. Trends Neurosci 33:362–372

    Article  CAS  PubMed  Google Scholar 

  8. Reed NA, Cai D, Blasius TL, Jih GT, Meyhofer E, Gaertig J, Verhey KJ (2006) Microtubule acetylation promotes kinesin-1 binding and transport. Curr Biol CB 16:2166–2172

    Article  CAS  Google Scholar 

  9. Chen S, Owens GC, Makarenkova H, Edelman DB (2010) HDAC6 regulates mitochondrial transport in hippocampal neurons. PLoS One 5:e10848

    Article  PubMed Central  PubMed  Google Scholar 

  10. Webster DR, Borisy GG (1989) Microtubules are acetylated in domains that turn over slowly. J Cell Sci 92(Pt 1):57–65

    PubMed  Google Scholar 

  11. Matsuyama A, Shimazu T, Sumida Y, Saito A, Yoshimatsu Y, Seigneurin-Berny D, Osada H, Komatsu Y, Nishino N, Khochbin S et al (2002) In vivo destabilization of dynamic microtubules by HDAC6-mediated deacetylation. EMBO J 21:6820–6831

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  12. Tran AD, Marmo TP, Salam AA, Che S, Finkelstein E, Kabarriti R, Xenias HS, Mazitschek R, Hubbert C, Kawaguchi Y et al (2007) HDAC6 deacetylation of tubulin modulates dynamics of cellular adhesions. J Cell Sci 120:1469–1479

    Article  CAS  PubMed  Google Scholar 

  13. Kapoor S, Panda D (2012) Kinetic stabilization of microtubule dynamics by indanocine perturbs EB1 localization, induces defects in cell polarity and inhibits migration of MDA-MB-231 cells. Biochem Pharmacol 83:1495–1506

    Article  CAS  PubMed  Google Scholar 

  14. Palazzo A, Ackerman B, Gundersen GG (2003) Cell biology: tubulin acetylation and cell motility. Nature 421:230

    Article  CAS  PubMed  Google Scholar 

  15. Henriques AG, Vieira SI, da Cruz ESEF, da Cruz ESOA (2010) Abeta promotes Alzheimer’s disease-like cytoskeleton abnormalities with consequences to APP processing in neurons. J Neurochem 113:761–771

    Article  CAS  PubMed  Google Scholar 

  16. Li G, Jiang H, Chang M, Xie H, Hu L (2011) HDAC6 alpha-tubulin deacetylase: a potential therapeutic target in neurodegenerative diseases. J Neurol Sci 304:1–8

    Article  CAS  PubMed  Google Scholar 

  17. d’Ydewalle C, Krishnan J, Chiheb DM, Van Damme P, Irobi J, Kozikowski AP, Vanden Berghe P, Timmerman V, Robberecht W, Van Den Bosch L (2011) HDAC6 inhibitors reverse axonal loss in a mouse model of mutant HSPB1-induced Charcot-Marie-Tooth disease. Nat Med 17:968–974

    Article  PubMed  Google Scholar 

  18. Kao DI, Aldridge GM, Weiler IJ, Greenough WT (2010) Altered mRNA transport, docking, and protein translation in neurons lacking fragile X mental retardation protein. Proceedings of the National Academy of Sciences of the United States of America 107: 15601–15606. DOI 10.1073/pnas.1010564107

  19. Williamson SL, Christodoulou J (2006) Rett syndrome: new clinical and molecular insights. Eur J Hum Genet EJHG 14:896–903

    Article  CAS  Google Scholar 

  20. Armstrong DD, Dunn K, Antalffy B (1998) Decreased dendritic branching in frontal, motor and limbic cortex in Rett syndrome compared with trisomy 21. J Neuropathol Exp Neurol 57:1013–1017

    Article  CAS  PubMed  Google Scholar 

  21. Bauman ML, Kemper TL, Arin DM (1995) Microscopic observations of the brain in Rett syndrome. Neuropediatrics 26:105–108

    Article  CAS  PubMed  Google Scholar 

  22. Asaka Y, Jugloff DG, Zhang L, Eubanks JH, Fitzsimonds RM (2006) Hippocampal synaptic plasticity is impaired in the Mecp2-null mouse model of Rett syndrome. Neurobiol Dis 21:217–227

    Article  CAS  PubMed  Google Scholar 

  23. Guy J, Gan J, Selfridge J, Cobb S, Bird A (2007) Reversal of neurological defects in a mouse model of Rett syndrome. Science 315:1143–1147

    Article  CAS  PubMed  Google Scholar 

  24. Moretti P, Levenson JM, Battaglia F, Atkinson R, Teague R, Antalffy B, Armstrong D, Arancio O, Sweatt JD, Zoghbi HY (2006) Learning and memory and synaptic plasticity are impaired in a mouse model of Rett syndrome. J Neurosci Off J Soc Neurosci 26:319–327

    Article  CAS  Google Scholar 

  25. Delepine C, Nectoux J, Bahi-Buisson N, Chelly J, Bienvenu T (2013) MeCP2 deficiency is associated with impaired microtubule stability. FEBS Lett 587:245–253

    Article  CAS  PubMed  Google Scholar 

  26. Nectoux J, Florian C, Delepine C, Bahi-Buisson N, Khelfaoui M, Reibel S, Chelly J, Bienvenu T (2012) Altered microtubule dynamics in Mecp2-deficient astrocytes. J Neurosci Res. doi:10.1002/jnr.23001

    PubMed  Google Scholar 

  27. Roux JC, Zala D, Panayotis N, Borges-Correia A, Saudou F, Villard L (2012) Modification of Mecp2 dosage alters axonal transport through the Huntingtin/Hap1 pathway. Neurobiol Dis 45:786–795

    Article  CAS  PubMed  Google Scholar 

  28. Butler K, Kalin J, Brochier C, Vistoli G, Langley B, Kozikowski AP (2010) Rational design and simple chemistry yield a superior, neuroprotective HDAC6 inhibitor, tubastatin A. J Am Chem Soc 132:10842

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  29. Asthana J, Kapoor S, Mohan R, Panda D (2013) Inhibition of HDAC6 deacetylase activity increases its binding with microtubules and suppresses microtubule dynamic instability in MCF-7 cells. J Biol Chem 288:22516–22526

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  30. Xu X, Kozikowski AP, Pozzo-Miller L (2014) A selective histone deacetylase-6 inhibitor improves BDNF trafficking in hippocampal neurons from Mecp2 knockout mice: implications for Rett syndrome. Front Cell Neurosci 8:68

    PubMed Central  PubMed  Google Scholar 

  31. Goffin D, Allen M, Zhang L, Amorim M, Wang IT, Reyes AR, Mercado-Berton A, Ong C, Cohen S, Hu L et al (2011) Rett syndrome mutation MeCP2 T158A disrupts DNA binding, protein stability and ERP responses. Nat Neurosci 15:274–283

    Article  PubMed Central  PubMed  Google Scholar 

  32. Scaife RM (2006) Microtubule disassembly and inhibition of mitosis by a novel synthetic pharmacophore. J Cell Biochem 98:102–114

    Article  CAS  PubMed  Google Scholar 

  33. Schmittgen TD, Livak KJ (2008) Analyzing real-time PCR data by the comparative C(T) method. Nat Protoc 3:1101–1108

    Article  CAS  PubMed  Google Scholar 

  34. Klauck SM, Lindsay S, Beyer KS, Splitt M, Burn J, Poustka A (2002) A mutation hot spot for nonspecific X-linked mental retardation in the MECP2 gene causes the PPM-X syndrome. Am J Hum Genet 70:1034–1037

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  35. Van Esch H, Bauters M, Ignatius J, Jansen M, Raynaud M, Hollanders K, Lugtenberg D, Bienvenu T, Jensen LR, Gecz J et al (2005) Duplication of the MECP2 region is a frequent cause of severe mental retardation and progressive neurological symptoms in males. Am J Hum Genet 77:442–453

    Article  PubMed Central  PubMed  Google Scholar 

  36. Collins AL, Levenson JM, Vilaythong AP, Richman R, Armstrong DL, Noebels JL, David Sweatt J, Zoghbi HY (2004) Mild overexpression of MeCP2 causes a progressive neurological disorder in mice. Hum Mol Genet 13:2679–2689

    Article  CAS  PubMed  Google Scholar 

  37. del Gaudio D, Fang P, Scaglia F, Ward PA, Craigen WJ, Glaze DG, Neul JL, Patel A, Lee JA, Irons M et al (2006) Increased MECP2 gene copy number as the result of genomic duplication in neurodevelopmentally delayed males. Genet Med Off J Am Coll Med Genet 8:784–792

    Google Scholar 

Download references

Acknowledgments

We thank Associate Professor James Eubanks of the University of Toronto, for many valuable discussions and Dr Zhaolan Zhou of the University of Pennsylvania, for supplying us with the initial breeding stock of the Mecp2 T158A mouse model. This work was supported by the Rett Syndrome Association of New South Wales, Rett Syndrome Australian Research Fund, Rett Syndrome Association of Australia, International Rett Syndrome Foundation and Shire Human Genetic Therapies Inc (Lexington, MA, USA).

Conflict of interest

The authors declare that there are no commercial or other conflicts of interest in connection with this research.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to John Christodoulou.

Electronic supplementary material

Below is the link to the electronic supplementary material.

Supplementary Table S1

Statistical analysis for Fig. 4 (Tubastatin A protects microtubules from severe nocodazole-induced depolymerisation). Statistical analysis was conducted on the computational counts of relative intensity levels of polymerised microtubules of patient and control fibroblasts treated in Fig. 4. (PDF 153 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Gold, W.A., Lacina, T.A., Cantrill, L.C. et al. MeCP2 deficiency is associated with reduced levels of tubulin acetylation and can be restored using HDAC6 inhibitors. J Mol Med 93, 63–72 (2015). https://doi.org/10.1007/s00109-014-1202-x

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s00109-014-1202-x

Keywords

Navigation