Skip to main content

Advertisement

Log in

Elevated MeCP2 in Mice Causes Neurodegeneration Involving Tau Dysregulation and Excitotoxicity: Implications for the Understanding and Treatment of MeCP2 Triplication Syndrome

  • Published:
Molecular Neurobiology Aims and scope Submit manuscript

Abstract

Expression of MeCP2 must be carefully regulated as a reduction or increase results in serious neurological disorders. We are studying transgenic mice in which the MeCP2 gene is expressed at about three times higher than the normal level. Male MeCP2-Tg mice, but not female mice, suffer motor and cognitive deficits and die at 18–20 weeks of age. MeCP2-Tg mice display elevated GFAP and Tau expression within the hippocampus and cortex followed by neuronal loss in these brain regions. Loss of Purkinje neurons, but not of granule neurons in the cerebellar cortex is also seen. Exposure of cultured cortical neurons to either conditioned medium from astrocytes (ACM) derived from male MeCP2-Tg mice or normal astrocytes in which MeCP2 is expressed at elevated levels promotes their death. Interestingly, ACM from male, but not female MeCP2-Tg mice, displays this neurotoxicity reflecting the gender selectivity of neurological symptoms in mice. Male ACM, but not female ACM, contains highly elevated levels of glutamate, and its neurotoxicity can be prevented by MK-801, indicating that it is caused by excitotoxicity. Based on the close phenotypic resemblance of MeCP2-Tg mice to patients with MECP2 triplication syndrome, we suggest for the first time that the human syndrome is a neurodegenerative disorder resulting from astrocyte dysfunction that leads to Tau-mediated excitotoxic neurodegeneration. Loss of cortical and hippocampal neurons may explain the mental retardation and epilepsy in patients, whereas ataxia likely results from the loss of Purkinje neurons.

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.

Fig. 1
Fig. 2
Fig. 3
Fig. 4
Fig. 5
Fig. 6
Fig. 7
Fig. 8
Fig. 9
Fig. 10
Fig. 11
Fig. 12
Fig. 13

Similar content being viewed by others

References

  1. Adkins NL, Georgel PT (2011) MeCP2: Structure and function. Biochem Cell Biol (Can) 89:1–11

    Article  CAS  Google Scholar 

  2. Guy J, Cheval H, Selfridge J, Bird A (2011) The role of MeCP2 in the brain. Annu Rev Cell Dev Biol (U S) 27:631–652

    Article  CAS  Google Scholar 

  3. Hite KC, Adams VH, Hansen JC (2009) Recent advances in MeCP2 structure and function. Biochem Cell Biol (Can) 87:219–227

    Article  CAS  Google Scholar 

  4. Ausio J, Martinez de Paz A, Esteller M (2014) MeCP2: The long trip from a chromatin protein to neurological disorders. Trends Mol Med (Engl) 20:487–498

    Article  CAS  Google Scholar 

  5. Chahrour M, Zoghbi HY (2007) The story of rett syndrome: From clinic to neurobiology. Neuron (U S) 56:422–437

    Article  CAS  Google Scholar 

  6. Lombardi LM, Baker SA, Zoghbi HY (2015) MECP2 disorders: From the clinic to mice and back. J Clin Invest (U S) 125:2914–2923

    Article  Google Scholar 

  7. Peters SU, Hundley RJ, Wilson AK, Carvalho CM, Lupski JR, Ramocki MB (2013) Brief report: Regression timing and associated features in MECP2 duplication syndrome. J Autism Dev Disord 43:2484–2490

    Article  CAS  Google Scholar 

  8. Ramocki MB, Tavyev YJ, Peters SU (2010) The MECP2 duplication syndrome. Am J Med Genet A (U S) 152A:1079–1088

    Article  Google Scholar 

  9. Van Esch H (2012) MECP2 duplication syndrome. Mol Syndromol 2:128–136

    PubMed  Google Scholar 

  10. Tang B, Becanovic K, Desplats PA, Spencer B, Hill AM, Connolly C, Masliah E, Leavitt BR et al (2012a) Forkhead box protein p1 is a transcriptional repressor of immune signaling in the CNS: Implications for transcriptional dysregulation in Huntington disease. Hum Mol Genet (Engl) 21:3097–3111

    Article  CAS  Google Scholar 

  11. Lugtenberg D, Kleefstra T, Oudakker AR, Nillesen WM, Yntema HG, Tzschach A, Raynaud M, Rating D et al (2009) Structural variation in Xq28: MECP2 duplications in 1% of patients with unexplained XLMR and in 2% of male patients with severe encephalopathy. Eur J Hum Genet (Engl) 17:444–453

    Article  CAS  Google Scholar 

  12. Shimada S, Okamoto N, Ito M, Arai Y, Momosaki K, Togawa M, Maegaki Y, Sugawara M et al (2013) MECP2 duplication syndrome in both genders. Brain Dev (Neth) 35:411–419

    Article  Google Scholar 

  13. Van Esch H, Bauters M, Ignatius J, Jansen M, Raynaud M, Hollanders K, Lugtenberg D, Bienvenu T 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 (U S) 77:442–453

    Article  Google Scholar 

  14. 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 (Engl) 13:2679–2689

    Article  CAS  Google Scholar 

  15. Jaworski T, Kugler S, Van Leuven F (2010) Modeling of tau-mediated synaptic and neuronal degeneration in alzheimer's disease. Int J Alzheimers Dis (Engl) 2010:1–10. https://doi.org/10.4061/2010/573138

    Article  Google Scholar 

  16. Jaworski T, Dewachter I, Lechat B, Croes S, Termont A, Demedts D, Borghgraef P, Devijver H et al (2009) AAV-tau mediates pyramidal neurodegeneration by cell-cycle re-entry without neurofibrillary tangle formation in wild-type mice. PLoS One (U S) 4:e7280

    Article  Google Scholar 

  17. Min SW et al (2015) Critical role of acetylation in tau-mediated neurodegeneration and cognitive deficits. Nat Med (U S) 21:1154–1162

    Article  CAS  Google Scholar 

  18. Wang L, Colodner KJ, Feany MB (2011) Protein misfolding and oxidative stress promote glial-mediated neurodegeneration in an alexander disease model. J Neurosci (U S) 31:2868–2877

    Article  CAS  Google Scholar 

  19. Bardai FH, Price V, Zaayman M, Wang L, D'Mello SR (2012) Histone deacetylase-1 (HDAC1) is a molecular switch between neuronal survival and death. J Biol Chem (U S) 287:35444–35453

    Article  CAS  Google Scholar 

  20. Bardai FH, Verma P, Smith C, Rawat V, Wang L, D'Mello SR (2013) Disassociation of histone deacetylase-3 from normal huntingtin underlies mutant huntingtin neurotoxicity. J Neurosci (U S) 33:11833–11838

    Article  CAS  Google Scholar 

  21. Norwood J, Franklin JM, Sharma D, D'Mello SR (2014) Histone deacetylase 3 is necessary for proper brain development. J Biol Chem (U S) 289:34569–34582

    Article  Google Scholar 

  22. Majdzadeh N, Wang L, Morrison BE, Bassel-Duby R, Olson EN, D'Mello SR (2008) HDAC4 inhibits cell-cycle progression and protects neurons from cell death. Dev Neurobiol (U S) 68:1076–1092

    Article  CAS  Google Scholar 

  23. Chen HM, Wang L, D'Mello SR (2008) A chemical compound commonly used to inhibit PKR, {8-(imidazol-4-ylmethylene)-6H-azolidino[5,4-g] benzothiazol-7-one}, protects neurons by inhibiting cyclin-dependent kinase. Eur J Neurosci (Fr) 28:2003–2016

    Article  Google Scholar 

  24. Tang SS, Fernandez D, Lazarou LP, Singh R, Fallon P (2012b) MECP2 triplication in 3 brothers - a rarely described cause of familial neurological regression in boys. Eur J Paediatr Neurol (Engl) 16:209–212

    Article  Google Scholar 

  25. Molyneaux BJ, Arlotta P, Menezes JR, Macklis JD (2007) Neuronal subtype specification in the cerebral cortex. Nat Rev Neurosci (Engl) 8:427–437

    Article  CAS  Google Scholar 

  26. Cerminara NL, Lang EJ, Sillitoe RV, Apps R (2015) Redefining the cerebellar cortex as an assembly of non-uniform purkinje cell microcircuits. Nat Rev Neurosci (Engl) 16:79–93

    Article  CAS  Google Scholar 

  27. Maragakis NJ, Rothstein JD (2006) Mechanisms of disease: Astrocytes in neurodegenerative disease. Nat Clin Pract Neurol (Engl) 2:679–689

    Article  CAS  Google Scholar 

  28. Bjornsen LP, Eid T, Holmseth S, Danbolt NC, Spencer DD, de Lanerolle NC (2007) Changes in glial glutamate transporters in human epileptogenic hippocampus: Inadequate explanation for high extracellular glutamate during seizures. Neurobiol Dis (U S) 25:319–330

    Article  CAS  Google Scholar 

  29. Robel S, Sontheimer H (2016) Glia as drivers of abnormal neuronal activity. Nat Neurosci (U S) 19:28–33

    Article  CAS  Google Scholar 

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

    Article  Google Scholar 

  31. 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 (U S) 90:990–998

    Article  CAS  Google Scholar 

  32. Papanikolopoulou K, Skoulakis EM (2011) The power and richness of modelling tauopathies in drosophila. Mol Neurobiol (U S) 44:122–133

    Article  CAS  Google Scholar 

  33. Sun XY, Tuo QZ, Liuyang ZY, Xie AJ, Feng XL, Yan X, Qiu M, Li S et al (2016) Extrasynaptic NMDA receptor-induced tau overexpression mediates neuronal death through suppressing survival signaling ERK phosphorylation. Cell Death Dis (Engl) 7:e2449

    Article  Google Scholar 

  34. Wittmann CW, Wszolek MF, Shulman JM, Salvaterra PM, Lewis J, Hutton M, Feany MB (2001a) Tauopathy in drosophila: Neurodegeneration without neurofibrillary tangles. Science (U S) 293:711–714

    Article  CAS  Google Scholar 

  35. Friez MJ, Jones JR, Clarkson K, Lubs H, Abuelo D, Bier JA, Pai S, Simensen R et al (2006) Recurrent infections, hypotonia, and mental retardation caused by duplication of MECP2 and adjacent region in Xq28. Pediatrics (U S) 118:e1687–e1695

    Google Scholar 

  36. Lubs H, Abidi F, Bier JA, Abuelo D, Ouzts L, Voeller K, Fennell E, Stevenson RE et al (1999) XLMR syndrome characterized by multiple respiratory infections, hypertelorism, severe CNS deterioration and early death localizes to distal Xq28. Am J Med Genet (U S) 85:243–248

    Article  CAS  Google Scholar 

  37. Reardon W, Donoghue V, Murphy AM, King MD, Mayne PD, Horn N, Birk Moller L (2010) Progressive cerebellar degenerative changes in the severe mental retardation syndrome caused by duplication of MECP2 and adjacent loci on Xq28. Eur J Pediatr (Germany) 169:941–949

    Article  Google Scholar 

  38. Jiang M, Ash RT, Baker SA, Suter B, Ferguson A, Park J, Rudy J, Torsky SP et al (2013) Dendritic arborization and spine dynamics are abnormal in the mouse model of MECP2 duplication syndrome. J Neurosci (U S) 33:19518–19533

    Article  Google Scholar 

  39. Ben-Shachar S, Chahrour M, Thaller C, Shaw CA, Zoghbi HY (2009) Mouse models of MeCP2 disorders share gene expression changes in the cerebellum and hypothalamus. Hum Mol Genet (Engl) 18:2431–2442

    Article  CAS  Google Scholar 

  40. Chahrour M, Jung SY, Shaw C, Zhou X, Wong ST, Qin J, Zoghbi HY (2008) MeCP2, a key contributor to neurological disease, activates and represses transcription. Science (U S) 320:1224–1229

    Article  CAS  Google Scholar 

  41. Samaco RC, Mandel-Brehm C, McGraw CM, Shaw CA, McGill BE, Zoghbi HY (2012) Crh and Oprm1 mediate anxiety-related behavior and social approach in a mouse model of MECP2 duplication syndrome. Nat Genet (U S) 44:206–211

    Article  CAS  Google Scholar 

  42. Messing A, Brenner M (2003) Alexander disease: GFAP mutations unify young and old. Lancet Neurol (Engl) 2:75

    Article  Google Scholar 

  43. Messing A, Brenner M, Feany MB, Nedergaard M, Goldman JE (2012) Alexander disease. J Neurosci (U S) 32:5017–5023

    Article  CAS  Google Scholar 

  44. Yoshida T, Nakagawa M (2012) Clinical aspects and pathology of alexander disease, and morphological and functional alteration of astrocytes induced by GFAP mutation. Neuropathology (Aust) 32:440–446

    Article  Google Scholar 

  45. Hagemann TL, Connor JX, Messing A (2006) Alexander disease-associated glial fibrillary acidic protein mutations in mice induce Rosenthal fiber formation and a white matter stress response. J Neurosci (U S) 26:11162–11173

    Article  CAS  Google Scholar 

  46. Jany PL, Hagemann TL, Messing A (2013) GFAP expression as an indicator of disease severity in mouse models of alexander disease. ASN Neuro (Engl) 5:e00109

    Google Scholar 

  47. Tanaka KF, Takebayashi H, Yamazaki Y, Ono K, Naruse M, Iwasato T, Itohara S, Kato H et al (2007) Murine model of alexander disease: Analysis of GFAP aggregate formation and its pathological significance. Glia (U S) 55:617–631

    Article  Google Scholar 

  48. Minkel HR, Anwer TZ, Arps KM, Brenner M, Olsen ML (2015) Elevated GFAP induces astrocyte dysfunction in caudal brain regions: A potential mechanism for hindbrain involved symptoms in type II alexander disease. Glia (U S) 63:2285–2297

    Article  Google Scholar 

  49. Sosunov AA, Guilfoyle E, Wu X, McKhann GM 2nd, Goldman JE (2013) Phenotypic conversions of “protoplasmic” to “reactive” astrocytes in alexander disease. J Neurosci (U S) 33:7439–7450

    Article  CAS  Google Scholar 

  50. Tian R, Wu X, Hagemann TL, Sosunov AA, Messing A, McKhann GM, Goldman JE (2010) Alexander disease mutant glial fibrillary acidic protein compromises glutamate transport in astrocytes. J Neuropathol Exp Neurol (U S) 69:335–345

    Article  CAS  Google Scholar 

  51. Ilieva H, Polymenidou M, Cleveland DW (2009) Non-cell autonomous toxicity in neurodegenerative disorders: ALS and beyond. J Cell Biol (U S) 187:761–772

    Article  CAS  Google Scholar 

  52. McGann JC, Lioy DT, Mandel G (2012) Astrocytes conspire with neurons during progression of neurological disease. Curr Opin Neurobiol (Engl) 22:850–858

    Article  CAS  Google Scholar 

  53. Sun M, Chen L (2015) Studying tauopathies in drosophila: A fruitful model. Exp Neurol 274:52–57

    Article  CAS  Google Scholar 

  54. Boehm J (2013) A ‘danse macabre’: Tau and fyn in STEP with amyloid beta to facilitate induction of synaptic depression and excitotoxicity. Eur J Neurosci (Fr) 37:1925–1930

    Article  Google Scholar 

  55. Couratier P, Lesort M, Terro F, Dussartre C, Hugon J (1996) NMDA antagonist blockade of AT8 tau immunoreactive changes in neuronal cultures. Fundam Clin Pharmacol (Engl) 10:344–349

    Article  CAS  Google Scholar 

  56. Esclaire F, Lesort M, Blanchard C, Hugon J (1997) Glutamate toxicity enhances tau gene expression in neuronal cultures. J Neurosci Res (U S) 49:309–318

    Article  CAS  Google Scholar 

  57. Sindou P, Couratier P, Barthe D, Hugon J (1992) A dose-dependent increase of tau immunostaining is produced by glutamate toxicity in primary neuronal cultures. Brain Res (Neth) 572:242–246

    Article  CAS  Google Scholar 

  58. Pizzi M, Valerio A, Ribola M, Spano PF, Memo M (1993) A tau antisense oligonucleotide decreases neurone sensitivity to excitotoxic injury. Neuroreport (Engl) 4:823–826

    Article  CAS  Google Scholar 

  59. Pizzi M, Valerio A, Arrighi V, Galli P, Belloni M, Ribola M, Alberici A, Spano P et al (1995) Inhibition of glutamate-induced neurotoxicity by a tau antisense oligonucleotide in primary culture of rat cerebellar granule cells. Eur J Neurosci (Fr) 7:1603–1613

    Article  CAS  Google Scholar 

  60. Cheng JS, Craft R, Yu GQ, Ho K, Wang X, Mohan G, Mangnitsky S, Ponnusamy R et al (2014) Tau reduction diminishes spatial learning and memory deficits after mild repetitive traumatic brain injury in mice. PLoS One (U S) 9:e115765

    Article  Google Scholar 

  61. Ke YD, Suchowerska AK, van der Hoven J, De Silva DM, Wu CW, van Eersel J, Ittner A, Ittner LM (2012) Lessons from tau-deficient mice. Int J Alzheimers Dis (U S) 2012:873270

    Google Scholar 

  62. Morris M, Maeda S, Vossel K, Mucke L (2011) The many faces of tau. Neuron (U S) 70:410–426

    Article  CAS  Google Scholar 

  63. Roberson ED, Scearce-Levie K, Palop JJ, Yan F, Cheng IH, Wu T, Gerstein H, Yu GQ et al (2007) Reducing endogenous tau ameliorates amyloid beta-induced deficits in an alzheimer's disease mouse model. Science (U S) 316:750–754

    Article  CAS  Google Scholar 

  64. Shipton OA, Leitz JR, Dworzak J, Acton CE, Tunbridge EM, Denk F, Dawson HN, Vitek MP et al (2011) Tau protein is required for amyloid {beta}-induced impairment of hippocampal long-term potentiation. J Neurosci (U S) 31:1688–1692

    Article  CAS  Google Scholar 

Download references

Acknowledgements

This research was supported by a grant from the National Institutes of Health (R01 NS040408). We thank Dr. Jade Franklin for making significant contributions to the research and the preparation of figures for the manuscript. We also acknowledge the contribution of Dr. Xiaoju Zou for her assistance with some of the immunohistology work described in the manuscript.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Santosh R. D’Mello.

Electronic supplementary material

Supplemental Figure 1

(A and B) Locomotor analysis at 15 weeks of age in WT and MeCP2-Tg female mice (n = 6). (C and D) Forelimb and hindlimb grip strength at 15 weeks of age in WT and MeCP2-Tg female mice (n = 6). (E) Brain Weight of 15-week-old WT and MeCP2-Tg female Mice (n = 6). (F) Body Weight of 15-week-old WT and MeCP2-Tg female mice (n = 6). (GIF 28 kb).

High Resolution Image (TIFF 191 kb).

Supplemental Figure 2

Male WT and MeCP2-Tg mice (blue), display elevated Tau that correlates to increases in MeCP2 expression (n = 11). Female WT and MeCP2-Tg mice (red) do not display the same correlation (n = 8). (GIF 14 kb).

High Resolution Image (TIFF 165 kb).

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Montgomery, K.R., Louis Sam Titus, A.S.C., Wang, L. et al. Elevated MeCP2 in Mice Causes Neurodegeneration Involving Tau Dysregulation and Excitotoxicity: Implications for the Understanding and Treatment of MeCP2 Triplication Syndrome. Mol Neurobiol 55, 9057–9074 (2018). https://doi.org/10.1007/s12035-018-1046-4

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s12035-018-1046-4

Keywords

Navigation