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.

  • Review Article
  • Published:

Clinical and biological progress over 50 years in Rett syndrome

Key Points

  • In the 50 years since its description by Andreas Rett, we have witnessed an explosion of knowledge about Rett syndrome (RTT) in relation to its genetic basis and clinical characteristics, and their interrelationships

  • Initially, the diagnosis of RTT was based solely on clinical criteria, but identification of its genetic cause has revolutionized this process, while presenting new challenges as we enter the era of next-generation sequencing

  • Mutations in the methyl-CpG-binding protein 2 (MECP2) gene were found to be causative of RTT, accounting for fundamentally altered neurobiological pathways, and providing the stimulus to identify pathways that can be manipulated therapeutically

  • The type of MECP2 mutation is associated with clinical severity, and influences many aspects of the phenotype, including functional abilities, onset of scoliosis, bone health, and sleep disturbances

  • Considerable progress has been made in understanding the natural history of RTT, leading to improvement in clinical management in selected areas, and changes in attitudes and allocation of health-care resources have increased life expectancy

  • The advancement in knowledge about RTT has been dependent on global efforts to study this disorder, including the establishment of database infrastructures, the input of advocacy groups, and the development of international collaborations

Abstract

In the 50 years since Andreas Rett first described the syndrome that came to bear his name, and is now known to be caused by a mutation in the methyl-CpG-binding protein 2 (MECP2) gene, a compelling blend of astute clinical observations and clinical and laboratory research has substantially enhanced our understanding of this rare disorder. Here, we document the contributions of the early pioneers in Rett syndrome (RTT) research, and describe the evolution of knowledge in terms of diagnostic criteria, clinical variation, and the interplay with other Rett-related disorders. We provide a synthesis of what is known about the neurobiology of MeCP2, considering the lessons learned from both cell and animal models, and how they might inform future clinical trials. With a focus on the core criteria, we examine the relationships between genotype and clinical severity. We review current knowledge about the many comorbidities that occur in RTT, and how genotype may modify their presentation. We also acknowledge the important drivers that are accelerating this research programme, including the roles of research infrastructure, international collaboration and advocacy groups. Finally, we highlight the major milestones since 1966, and what they mean for the day-to-day lives of individuals with RTT and their families.

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

Figure 1
Figure 2: The MECP2 gene and Rett syndrome.
Figure 3: Rett syndrome severity and age at diagnosis by mutation type.
Figure 4: Functional abilities and mutation type in Rett syndrome.
Figure 5: Comorbidities and mutation type in Rett syndrome.
Figure 6: Therapeutic strategies for Rett syndrome.

Similar content being viewed by others

References

  1. Rett, A. On a unusual brain atrophy syndrome in hyperammonemia in childhood. Wien. Med. Wochenschr. 116, 723–726 (in German) (1966).

    CAS  PubMed  Google Scholar 

  2. Hagberg, B., Aicardi, J., Dias, K. & Ramos, O. A progressive syndrome of autism, dementia, ataxia, and loss of purposeful hand use in girls: Rett's syndrome: report of 35 cases. Ann. Neurol. 14, 471–479 (1983). The initial clinical description of 35 cases of Rett syndrome in the English-speaking literature.

    Article  CAS  PubMed  Google Scholar 

  3. Fehr, S., Downs, J., Bebbington, A. & Leonard, H. Atypical presentations and specific genotypes are associated with a delay in diagnosis in females with Rett syndrome. Am. J. Med. Genet. A 152A, 2535–2542 (2010).

    Article  PubMed  Google Scholar 

  4. Hagberg, B., Goutières, F., Hanefeld, F., Rett, A. & Wilson, J. Rett syndrome: criteria for inclusion and exclusion. Brain Dev. 7, 372–373 (1985).

    Article  CAS  PubMed  Google Scholar 

  5. [No authors listed.] Diagnostic criteria for Rett syndrome. The Rett Syndrome Diagnostic Criteria Work Group. Ann. Neurol. 23, 425–428 (1988).

  6. Hagberg, B., Hanefeld, F., Percy, A. & Skjeldal, O. An update on clinically applicable diagnostic criteria in Rett syndrome. Comments to Rett Syndrome Clinical Criteria Consensus Panel Satellite to European Paediatric Neurology Society Meeting, Baden Baden, Germany, 11 September 2001. Eur. J. Paediatr. Neurol. 6, 293–297 (2002).

    Article  PubMed  Google Scholar 

  7. Neul, J. L. et al. Rett syndrome: revised diagnostic criteria and nomenclature. Ann. Neurol. 68, 944–950 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  8. Naidu, S., Murphy, M., Moser, H. W. & Rett, A. Rett syndrome — natural history in 70 cases. Am. J. Med. Genet. Suppl. 1, 61–72 (1986).

    Article  CAS  PubMed  Google Scholar 

  9. Kerr, A. M. & Stephenson, J. B. Rett's syndrome in the west of Scotland. Br. Med. J. (Clin. Res. Ed.) 291, 579–582 (1985).

    Article  CAS  Google Scholar 

  10. Hagberg, B. & Witt-Engerström, I. Rett syndrome: a suggested staging system for describing impairment profile with increasing age towards adolescence. Am. J. Med. Genet. Suppl. 1, 47–59 (1986).

    Article  CAS  PubMed  Google Scholar 

  11. Amir, R. E. et al. Rett syndrome is caused by mutations in X-linked MECP2, encoding methyl-CpG-binding protein 2. Nat. Genet. 23, 185–188 (1999). This study established that Rett syndrome is a genetic disorder caused by mutations in the MECP2 gene.

    Article  CAS  PubMed  Google Scholar 

  12. Schanen, C. & Francke, U. A severely affected male born into a Rett syndrome kindred supports X-linked inheritance and allows extension of the exclusion map. Am. J. Hum. Genet. 63, 267–269 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Sirianni, N., Naidu, S., Pereira, J., Pillotto, R. F. & Hoffman, E. P. Rett syndrome: confirmation of X-linked dominant inheritance, and localization of the gene to Xq28. Am. J. Hum. Genet. 63, 1552–1558 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Amir, R. E. et al. Influence of mutation type and X chromosome inactivation on Rett syndrome phenotypes. Ann. Neurol. 47, 670–679 (2000).

    Article  CAS  PubMed  Google Scholar 

  15. Bienvenu, T. et al. MECP2 mutations account for most cases of typical forms of Rett syndrome. Hum. Mol. Genet. 9, 1377–1384 (2000).

    Article  CAS  PubMed  Google Scholar 

  16. Hoffbuhr, K. et al. MeCP2 mutations in children with and without the phenotype of Rett syndrome. Neurology 56, 1486–1495 (2001).

    Article  CAS  PubMed  Google Scholar 

  17. Huppke, P., Held, M., Hanefeld, F., Engel, W. & Laccone, F. Influence of mutation type and location on phenotype in 123 patients with Rett syndrome. Neuropediatrics 33, 63–68 (2002).

    Article  CAS  PubMed  Google Scholar 

  18. Cheadle, J. P. et al. Long-read sequence analysis of the MECP2 gene in Rett syndrome patients: correlation of disease severity with mutation type and location. Hum. Mol. Genet. 9, 1119–1129 (2000). One of the most important of the early genotype–phenotype studies, this joint UK–Australian collaboration identified MECP2 mutations in 80% of typical Rett syndrome cases. Each of the eight recurrent missense and nonsense mutations, which account for almost two-thirds of the mutations seen in Rett syndrome, were represented. Using a simple phenotype score, this study showed that missense mutations generally had milder effects than truncating mutations.

    Article  CAS  PubMed  Google Scholar 

  19. Dragich, J., Houwink-Manville, I. & Schanen, C. Rett syndrome: a surprising result of mutation in MECP2. Hum. Mol. Genet. 9, 2365–2375 (2000).

    Article  CAS  PubMed  Google Scholar 

  20. Cuddapah, V. A. et al. Methyl-CpG-binding protein 2, (MECP2) mutation type is associated with disease severity in Rett syndrome. J. Med. Genet. 51, 152–158 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Erlandson, A. et al. Multiplex ligation-dependent probe amplification (MLPA) detects large deletions in the MECP2 gene of Swedish Rett syndrome patients. Genet. Test. 7, 329–332 (2003).

    Article  CAS  PubMed  Google Scholar 

  22. Hardwick, S. A. et al. Delineation of large deletions of the MECP2 gene in Rett syndrome patients, including a familial case with a male proband. Eur. J. Hum. Genet. 15, 1218–1229 (2007).

    Article  CAS  PubMed  Google Scholar 

  23. Armstrong, D., Dunn, J. K., Antalffy, B. & Trivedi, R. Selective dendritic alterations in the cortex of Rett syndrome. J. Neuropathol. Exp. Neurol. 54, 195–201 (1995).

    Article  CAS  PubMed  Google Scholar 

  24. Marchetto, M. C. et al. A model for neural development and treatment of Rett syndrome using human induced pluripotent stem cells. Cell 143, 527–539 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Yazdani, M. et al. Disease modeling using embryonic stem cells: MeCP2 regulates nuclear size and RNA synthesis in neurons. Stem Cells 30, 2128–2139 (2012).

    Article  CAS  PubMed  Google Scholar 

  26. Livide, G. et al. GluD1 is a common altered player in neuronal differentiation from both MECP2-mutated and CDKL5-mutated iPS cells. Eur. J. Hum. Genet. 23, 195–201 (2015).

    Article  CAS  PubMed  Google Scholar 

  27. Williams, E. C. et al. Mutant astrocytes differentiated from Rett syndrome patients-specific iPSCs have adverse effects on wild-type neurons. Hum. Mol. Genet. 23, 2968–2980 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Cheung, A. Y., Horvath, L. M., Carrel, L. & Ellis, J. X-chromosome inactivation in Rett syndrome human induced pluripotent stem cells. Front. Psychiatry 3, 24 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Guy, J., Hendrich, B., Holmes, M., Martin, J. E. & Bird, A. A mouse Mecp2-null mutation causes neurological symptoms that mimic Rett syndrome. Nat. Genet. 27, 322–326 (2001).

    Article  CAS  PubMed  Google Scholar 

  30. Chen, R. Z., Akbarian, S., Tudor, M. & Jaenisch, R. Deficiency of methyl-CpG binding protein-2 in CNS neurons results in a Rett-like phenotype in mice. Nat. Genet. 27, 327–331 (2001). References 29 and 30 described neurological deficits in Mecp2 -knockout mice, establishing a model system for studying Rett syndrome.

    Article  CAS  PubMed  Google Scholar 

  31. Gemelli, T. et al. Postnatal loss of methyl-CpG binding protein 2 in the forebrain is sufficient to mediate behavioral aspects of Rett syndrome in mice. Biol. Psychiatry 59, 468–476 (2006).

    Article  CAS  PubMed  Google Scholar 

  32. Chao, H. T. et al. Dysfunction in GABA signalling mediates autism-like stereotypies and Rett syndrome phenotypes. Nature 468, 263–269 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Fyffe, S. L. et al. Deletion of Mecp2 in Sim1-expressing neurons reveals a critical role for MeCP2 in feeding behavior, aggression, and the response to stress. Neuron 59, 947–958 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Wang, X., Lacza, Z., Sun, Y. E. & Han, W. Leptin resistance and obesity in mice with deletion of methyl-CpG-binding protein 2 (MeCP2) in hypothalamic pro-opiomelanocortin (POMC) neurons. Diabetologia 57, 236–245 (2014).

    Article  CAS  PubMed  Google Scholar 

  35. Samaco, R. C. et al. Loss of MeCP2 in aminergic neurons causes cell-autonomous defects in neurotransmitter synthesis and specific behavioral abnormalities. Proc. Natl Acad. Sci. USA 106, 21966–21971 (2009).

    Article  CAS  PubMed  Google Scholar 

  36. Goffin, D., Brodkin, E. S., Blendy, J. A., Siegel, S. J. & Zhou, Z. Cellular origins of auditory event-related potential deficits in Rett syndrome. Nat. Neurosci. 17, 804–806 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Zhang, W., Peterson, M., Beyer, B., Frankel, W. N. & Zhang, Z. W. Loss of MeCP2 from forebrain excitatory neurons leads to cortical hyperexcitation and seizures. J. Neurosci. 34, 2754–2763 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Ward, C. S. et al. MeCP2 is critical within HoxB1-derived tissues of mice for normal lifespan. J. Neurosci. 31, 10359–10370 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Lyst, M. J. & Bird, A. Rett syndrome: a complex disorder with simple roots. Nat. Rev. Genet. 16, 261–275 (2015).

    Article  CAS  PubMed  Google Scholar 

  40. Meng, X. et al. Manipulations of MeCP2 in glutamatergic neurons highlight their contributions to Rett and other neurological disorders. eLife 5, e14199 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Cheval, H. et al. Postnatal inactivation reveals enhanced requirement for MeCP2 at distinct age windows. Hum. Mol. Genet. 21, 3806–3814 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. McGraw, C. M., Samaco, R. C. & Zoghbi, H. Y. Adult neural function requires MeCP2. Science 333, 186 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Guy, J., Gan, J., Selfridge, J., Cobb, S. & Bird, A. Reversal of neurological defects in a mouse model of Rett syndrome. Science 315, 1143–1147 (2007). This study shows that overt neurological features seen in MeCP2-deficient mice can be substantially reversed by re-expression of the protein in adult mice.

    Article  CAS  PubMed  Google Scholar 

  44. Robinson, L. et al. Morphological and functional reversal of phenotypes in a mouse model of Rett syndrome. Brain 135, 2699–2710 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  45. Ure, K. et al. Restoration of Mecp2 expression in GABAergic neurons is sufficient to rescue multiple disease features in a mouse model of Rett syndrome. eLife 5, e14198 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Shahbazian, M. D., Antalffy, B., Armstrong, D. L. & Zoghbi, H. Y. Insight into Rett syndrome: MeCP2 levels display tissue- and cell-specific differences and correlate with neuronal maturation. Hum. Mol. Genet. 11, 115–124 (2002).

    Article  CAS  PubMed  Google Scholar 

  47. Skene, P. J. et al. Neuronal MeCP2 is expressed at near histone-octamer levels and globally alters the chromatin state. Mol. Cell 37, 457–468 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Lioy, D. T. et al. A role for glia in the progression of Rett's syndrome. Nature 475, 497–500 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Nguyen, M. V. et al. Oligodendrocyte lineage cells contribute unique features to Rett syndrome neuropathology. J. Neurosci. 33, 18764–18774 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Song, C. et al. DNA methylation reader MECP2: cell type- and differentiation stage-specific protein distribution. Epigenetics Chromatin 7, 17 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Ross, P. D. et al. Exclusive expression of MeCP2 in the nervous system distinguishes between brain and peripheral Rett syndrome-like phenotypes. Hum. Mol. Genet. http://dx.doi.org/10.1093/hmg/ddw269 (2016).

  52. Ballas, N., Lioy, D. T., Grunseich, C. & Mandel, G. Non-cell autonomous influence of MeCP2-deficient glia on neuronal dendritic morphology. Nat. Neurosci. 12, 311–317 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Luikenhuis, S., Giacometti, E., Beard, C. F. & Jaenisch, R. Expression of MeCP2 in postmitotic neurons rescues Rett syndrome in mice. Proc. Natl Acad. Sci. USA 101, 6033–6038 (2004).

    Article  CAS  PubMed  Google Scholar 

  54. Kyle, S. M., Saha, P. K., Brown, H. M., Chan, L. C. & Justice, M. J. MeCP2 co-ordinates liver lipid metabolism with the NCoR1/HDAC3 corepressor complex. Hum. Mol. Genet. 25, 3029–3041 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  55. De Felice, C. et al. Unrecognized lung disease in classic Rett syndrome: a physiologic and high-resolution CT imaging study. Chest 138, 386–392 (2010).

    Article  PubMed  Google Scholar 

  56. Panighini, A. et al. Vascular dysfunction in a mouse model of Rett syndrome and effects of curcumin treatment. PLoS ONE 8, e64863 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  57. McCauley, M. D. et al. Pathogenesis of lethal cardiac arrhythmias in Mecp2 mutant mice: implication for therapy in Rett syndrome. Sci. Transl Med. 3, 113ra125 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. O'Connor, R. D., Zayzafoon, M., Farach-Carson, M. C. & Schanen, N. C. Mecp2 deficiency decreases bone formation and reduces bone volume in a rodent model of Rett syndrome. Bone 45, 346–356 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Kamal, B. et al. Biomechanical properties of bone in a mouse model of Rett syndrome. Bone 71, 106–114 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Blue, M. E. et al. Osteoblast function and bone histomorphometry in a murine model of Rett syndrome. Bone 76, 23–30 (2015).

    Article  PubMed  Google Scholar 

  61. Conti, V. et al. MeCP2 affects skeletal muscle growth and morphology through non cell-autonomous mechanisms. PLoS ONE 10, e0130183 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  63. Yasui, D. H. et al. Mice with an isoform-ablating Mecp2 exon 1 mutation recapitulate the neurologic deficits of Rett syndrome. Hum. Mol. Genet. 23, 2447–2458 (2014).

    Article  CAS  PubMed  Google Scholar 

  64. Kerr, B. et al. Transgenic complementation of MeCP2 deficiency: phenotypic rescue of Mecp2-null mice by isoform-specific transgenes. Eur. J. Hum. Genet. 20, 69–76 (2012).

    Article  CAS  PubMed  Google Scholar 

  65. Lewis, J. D. et al. Purification, sequence, and cellular localization of a novel chromosomal protein that binds to methylated DNA. Cell 69, 905–914 (1992). This paper describes the original discovery of MeCP2 as a DNA-binding protein.

    Article  CAS  PubMed  Google Scholar 

  66. Nan, X., Meehan, R. R. & Bird, A. Dissection of the methyl-CpG binding domain from the chromosomal protein MeCP2. Nucleic Acids Res. 21, 4886–4892 (1993).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Guo, W. et al. VPA alleviates neurological deficits and restores gene expression in a mouse model of Rett syndrome. PLoS ONE 9, e100215 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Mellen, M., Ayata, P., Dewell, S., Kriaucionis, S. & Heintz, N. MeCP2 binds to 5hmC enriched within active genes and accessible chromatin in the nervous system. Cell 151, 1417–1430 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Spruijt, C. G. et al. Dynamic readers for 5-(hydroxy)methylcytosine and its oxidized derivatives. Cell 152, 1146–1159 (2013).

    Article  CAS  PubMed  Google Scholar 

  70. Kriaucionis, S. & Heintz, N. The nuclear DNA base 5-hydroxymethylcytosine is present in Purkinje neurons and the brain. Science 324, 929–930 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Lyst, M. J. et al. Rett syndrome mutations abolish the interaction of MeCP2 with the NCoR/SMRT co-repressor. Nat. Neurosci. 16, 898–902 (2013). This paper describes the interaction of MeCP2 with NCOR–SMRT, and shows that this interaction is abolished by Rett syndrome-causing mutations in this region.

    Article  CAS  PubMed  Google Scholar 

  72. Baker, S. A. et al. An AT-hook domain in MeCP2 determines the clinical course of Rett syndrome and related disorders. Cell 152, 984–996 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Heckman, L. D., Chahrour, M. H. & Zoghbi, H. Y. Rett-causing mutations reveal two domains critical for MeCP2 function and for toxicity in MECP2 duplication syndrome mice. eLife 3, e02676 (2014).

    Article  CAS  PubMed Central  Google Scholar 

  74. Bird, A. DNA methylation patterns and epigenetic memory. Genes Dev. 16, 6–21 (2002).

    Article  CAS  PubMed  Google Scholar 

  75. Chahrour, M. et al. MeCP2, a key contributor to neurological disease, activates and represses transcription. Science 320, 1224–1229 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Nan, X. et al. Transcriptional repression by the methyl-CpG-binding protein MeCP2 involves a histone deacetylase complex. Nature 393, 386–389 (1998).

    Article  CAS  PubMed  Google Scholar 

  77. Jones, P. L. et al. Methylated DNA and MeCP2 recruit histone deacetylase to repress transcription. Nat. Genet. 19, 187–191 (1998).

    Article  CAS  PubMed  Google Scholar 

  78. Kokura, K. et al. The Ski protein family is required for MeCP2-mediated transcriptional repression. J. Biol. Chem. 276, 34115–34121 (2001).

    Article  CAS  PubMed  Google Scholar 

  79. Stancheva, I., Collins, A. L., Van den Veyver, I. B., Zoghbi, H. & Meehan, R. R. A mutant form of MeCP2 protein associated with human Rett syndrome cannot be displaced from methylated DNA by Notch in Xenopus embryos. Mol. Cell 12, 425–435 (2003).

    Article  CAS  PubMed  Google Scholar 

  80. Sugino, K. et al. Cell-type-specific repression by methyl-CpG-binding protein 2 is biased toward long genes. J. Neurosci. 34, 12877–12883 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Gabel, H. W. et al. Disruption of DNA-methylation-dependent long gene repression in Rett syndrome. Nature 522, 89–93 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  82. Brero, A. et al. Methyl CpG-binding proteins induce large-scale chromatin reorganization during terminal differentiation. J. Cell Biol. 169, 733–743 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. Young, J. I. et al. Regulation of RNA splicing by the methylation-dependent transcriptional repressor methyl-CpG binding protein 2. Proc. Natl Acad. Sci. USA 102, 17551–17558 (2005).

    Article  CAS  PubMed  Google Scholar 

  84. Maunakea, A. K., Chepelev, I., Cui, K. & Zhao, K. Intragenic DNA methylation modulates alternative splicing by recruiting MeCP2 to promote exon recognition. Cell Res. 23, 1256–1269 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  85. Cheng, T. L. et al. MeCP2 suppresses nuclear microRNA processing and dendritic growth by regulating the DGCR8/Drosha complex. Dev. Cell 28, 547–560 (2014).

    Article  CAS  PubMed  Google Scholar 

  86. Klein, M. E. et al. Homeostatic regulation of MeCP2 expression by a CREB-induced microRNA. Nat. Neurosci. 10, 1513–1514 (2007).

    Article  CAS  PubMed  Google Scholar 

  87. Han, K. et al. Human-specific regulation of MeCP2 levels in fetal brains by microRNA miR-483-5p. Genes Dev. 27, 485–490 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  88. Tao, J. et al. Phosphorylation of MeCP2 at serine 80 regulates its chromatin association and neurological function. Proc. Natl Acad. Sci. USA 106, 4882–4887 (2009).

    Article  CAS  PubMed  Google Scholar 

  89. Ebert, D. H. et al. Activity-dependent phosphorylation of MeCP2 threonine 308 regulates interaction with NCoR. Nature 499, 341–345 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  90. Meins, M. et al. Submicroscopic duplication in Xq28 causes increased expression of the MECP2 gene in a boy with severe mental retardation and features of Rett syndrome. J. Med. Genet. 42, e12 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  91. Van Esch, H. et al. 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 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  92. Collins, A. L. et al. Mild overexpression of MeCP2 causes a progressive neurological disorder in mice. Hum. Mol. Genet. 13, 2679–2689 (2004).

    Article  CAS  PubMed  Google Scholar 

  93. Lim, Z., Downs, J., Wong, K., Ellaway, C. & Leonard, H. Expanding the clinical picture of the MECP2 duplication syndrome. Clin. Genet. http://dx.doi.org/10.1111/cge.12814 (2016).

  94. Sztainberg, Y. et al. Reversal of phenotypes in MECP2 duplication mice using genetic rescue or antisense oligonucleotides. Nature 528, 123–126 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  95. Tudor, M., Akbarian, S., Chen, R. Z. & Jaenisch, R. Transcriptional profiling of a mouse model for Rett syndrome reveals subtle transcriptional changes in the brain. Proc. Natl Acad. Sci. USA 99, 15536–15541 (2002).

    Article  CAS  PubMed  Google Scholar 

  96. Dani, V. S. et al. Reduced cortical activity due to a shift in the balance between excitation and inhibition in a mouse model of Rett syndrome. Proc. Natl Acad. Sci. USA 102, 12560–12565 (2005).

    Article  CAS  PubMed  Google Scholar 

  97. Nelson, E. D., Kavalali, E. T. & Monteggia, L. M. MeCP2-dependent transcriptional repression regulates excitatory neurotransmission. Curr. Biol. 16, 710–716 (2006).

    Article  CAS  PubMed  Google Scholar 

  98. Chao, H. T., Zoghbi, H. Y. & Rosenmund, C. MeCP2 controls excitatory synaptic strength by regulating glutamatergic synapse number. Neuron 56, 58–65 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  99. Li, H., Zhong, X., Chau, K. F., Williams, E. C. & Chang, Q. Loss of activity-induced phosphorylation of MeCP2 enhances synaptogenesis, LTP and spatial memory. Nat. Neurosci. 14, 1001–1008 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  100. Weng, S. M., McLeod, F., Bailey, M. E. & Cobb, S. R. Synaptic plasticity deficits in an experimental model of Rett syndrome: long-term potentiation saturation and its pharmacological reversal. Neuroscience 180, 314–321 (2011).

    Article  CAS  PubMed  Google Scholar 

  101. Ricciardi, S. et al. Reduced AKT/mTOR signaling and protein synthesis dysregulation in a Rett syndrome animal model. Hum. Mol. Genet. 20, 1182–1196 (2011).

    Article  CAS  PubMed  Google Scholar 

  102. Kriaucionis, S. et al. Gene expression analysis exposes mitochondrial abnormalities in a mouse model of Rett syndrome. Mol. Cell. Biol. 26, 5033–5042 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  103. De Felice, C. et al. Oxidative brain damage in Mecp2-mutant murine models of Rett syndrome. Neurobiol. Dis. 68, 66–77 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  104. Trevathan, E. & Adams, M. J. The epidemiology and public health significance of Rett syndrome. J. Child Neurol. 3, S17–S20 (1988).

    Article  PubMed  Google Scholar 

  105. Trevathan, E. Rett syndrome. Pediatrics 83, 636–637 (1989).

    CAS  PubMed  Google Scholar 

  106. Goutières, F. & Aicardi, J. Atypical forms of Rett syndrome. Am. J. Med. Genet. Suppl. 1, 183–194 (1986).

    Article  PubMed  Google Scholar 

  107. Zappella, M. The Rett girls with preserved speech. Brain Dev. 14, 98–101 (1992).

    Article  CAS  PubMed  Google Scholar 

  108. Hagberg, B. A. & Skjeldal, O. H. Rett variants: a suggested model for inclusion criteria. Pediatr. Neurol. 11, 5–11 (1994). This paper developed a model for the clinical delineation of atypical cases of Rett syndrome, based on the presence of combined clusters of at least three of six primary criteria and at least five of 11 supportive manifestations in children aged ≥10 years. Importantly, the paper acknowledged that many of the supportive criteria, such as epilepsy and scoliosis, are not present in children aged <5 years, but appear with age.

    Article  CAS  PubMed  Google Scholar 

  109. Leonard, H. & Bower, C. Is the girl with Rett syndrome normal at birth? Dev. Med. Child Neurol. 40, 115–121 (1998).

    CAS  PubMed  Google Scholar 

  110. Hagberg, G., Stenbom, Y. & Engerstrom, I. W. Head growth in Rett syndrome. Brain Dev. 23, S227–S229 (2001).

    Article  PubMed  Google Scholar 

  111. Naidu, S. & Johnston, M. V. Neurodevelopmental disorders: clinical criteria for Rett syndrome. Nat. Rev. Neurol. 7, 312–314 (2011).

    Article  PubMed  Google Scholar 

  112. Beale, S., Sanderson, D., Sanniti, A., Dundar, Y. & Boland, A. A scoping study to explore the cost-effectiveness of next-generation sequencing compared with traditional genetic testing for the diagnosis of learning disabilities in children. Health Technol. Assess. 19, 1–90 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  113. Fehr, S. et al. The CDKL5 disorder is an independent clinical entity associated with early-onset encephalopathy. Eur. J. Hum. Genet. 21, 266–273 (2013).

    Article  CAS  PubMed  Google Scholar 

  114. Ariani, F. et al. FOXG1 is responsible for the congenital variant of Rett syndrome. Am. J. Hum. Genet. 83, 89–93 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  115. Urbanowicz, A., Downs, J., Girdler, S., Ciccone, N. & Leonard, H. Aspects of speech–language abilities are influenced by MECP2 mutation type in girls with Rett syndrome. Am. J. Med. Genet. A 167A, 354–362 (2015).

    Article  CAS  PubMed  Google Scholar 

  116. De Bona, C. et al. Preserved speech variant is allelic of classic Rett syndrome. Eur. J. Hum. Genet. 8, 325–330 (2000).

    Article  CAS  PubMed  Google Scholar 

  117. Hagberg, B. Clinical delineation of Rett syndrome variants. Neuropediatrics 26, 62 (1995).

    Article  CAS  PubMed  Google Scholar 

  118. Opitz, J. M. & Lewin, S. O. Rett syndrome — a review and discussion of syndrome delineation and syndrome definition. Brain Dev. 9, 445–450 (1987).

    Article  CAS  PubMed  Google Scholar 

  119. Erlandson, A. & Hagberg, B. MECP2 abnormality phenotypes: clinicopathologic area with broad variability. J. Child Neurol. 20, 727–732 (2005).

    Article  PubMed  Google Scholar 

  120. Bebbington, A. et al. Updating the profile of C-terminal MECP2 deletions in Rett syndrome. J. Med. Genet. 47, 242–248 (2010).

    Article  CAS  PubMed  Google Scholar 

  121. Suter, B., Treadwell-Deering, D., Zoghbi, H. Y., Glaze, D. G. & Neul, J. L. Brief report: MECP2 mutations in people without Rett syndrome. J. Autism Dev. Disord. 44, 703–711 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  122. Kerr, A. M. et al. Guidelines for reporting clinical features in cases with MECP2 mutations. Brain Dev. 23, 208–211 (2001). An international group developed a simple scoring system to assess clinical severity and capture the variability in Rett syndrome, especially for the purpose of genotype–phenotype comparisons. 20 items were included, and 2 points were allocated for a severe abnormality, 1 point for a perceptible but not extreme abnormality, and 0 points for no abnormality.

    Article  CAS  PubMed  Google Scholar 

  123. Percy, A. K. Rett syndrome: clinical correlates of the newly discovered gene. Brain Dev. 23, S202–S205 (2001).

    Article  PubMed  Google Scholar 

  124. Monros, E. et al. Rett syndrome in Spain: mutation analysis and clinical correlations. Brain Dev. 23, S251–S253 (2001).

    Article  PubMed  Google Scholar 

  125. Colvin, L. et al. Describing the phenotype in Rett syndrome using a population database. Arch. Dis. Child. 88, 38–43 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  126. Bebbington, A. et al. Investigating genotype–phenotype relationships in Rett syndrome using an international data set. Neurology 70, 868–875 (2008).

    Article  CAS  PubMed  Google Scholar 

  127. Neul, J. L. et al. Specific mutations in methyl-CpG-binding protein 2 confer different severity in Rett syndrome. Neurology 70, 1313–1321 (2008). References 126 and 127 described the first adequately sized samples to provide definitive information about genotype–phenotype relationships. The findings were broadly similar, with the most severe MECP2 mutations being Arg270X, Arg255X and Arg168X, whereas Arg133Cys and Arg294X had less-severe effects. Overall, individuals with severe mutations were less likely to walk, retain hand use, or use words.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  128. Fehr, S. et al. Altered attainment of developmental milestones influences the age of diagnosis of Rett syndrome. J. Child Neurol. 26, 980–987 (2011).

    Article  PubMed  Google Scholar 

  129. Bebbington, A. et al. The phenotype associated with a large deletion on MECP2. Eur. J. Hum. Genet. 20, 921–927 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  130. Archer, H. et al. Correlation between clinical severity in patients with Rett syndrome with a p. R168X or p. T158M MECP2 mutation, and the direction and degree of skewing of X-chromosome inactivation. J. Med. Genet. 44, 148–152 (2007). One of the few papers to undertake a thorough examination of the effects of X-chromosome inactivation in individuals with the same mutation, namely, the two common MECP2 mutations Arg168X and Thr158Met. A statistically significant increase in clinical severity with increase in the proportion of active mutated allele was shown for both these mutations.

    Article  CAS  PubMed  Google Scholar 

  131. Zeev, B. B. et al. The common BDNF polymorphism may be a modifier of disease severity in Rett syndrome. Neurology 72, 1242–1247 (2009). This study investigated the effect of a potential genetic modifier, the BDNF Val66Met polymorphism, on clinical severity. In individuals with the Arg168X mutation in MECP2 , heterozygosity for the Val66Met polymorphism was associated with an increase in Rett syndrome severity and earlier age of seizure onset in comparison with individuals homozygous for the wild-type BDNF allele.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  132. Kondo, M. et al. Environmental enrichment ameliorates a motor coordination deficit in a mouse model of Rett syndrome — Mecp2 gene dosage effects and BDNF expression. Eur. J. Neurosci. 27, 3342–3350 (2008).

    Article  PubMed  Google Scholar 

  133. Lee, J. Y., Leonard, H., Piek, J. P. & Downs, J. Early development and regression in Rett syndrome. Clin. Genet. 84, 572–576 (2013).

    Article  CAS  PubMed  Google Scholar 

  134. Downs, J. et al. Level of purposeful hand function as a marker of clinical severity in Rett syndrome. Dev. Med. Child Neurol. 52, 817–823 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  135. Downs, J. et al. Validating the Rett Syndrome Gross Motor Scale. PLoS ONE 11, e0147555 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  136. Foley, K. R. et al. Change in gross motor abilities of girls and women with Rett syndrome over a 3- to 4-year period. J. Child Neurol. 26, 1237–1245 (2011).

    Article  PubMed  Google Scholar 

  137. Anderson, A., Wong, K., Jacoby, P., Downs, J. & Leonard, H. Twenty years of surveillance in Rett syndrome: what does this tell us? Orphanet J. Rare Dis. 9, 87 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  138. Sigafoos, J. Communication intervention in Rett syndrome: a systematic review. Res. Autism Spectr. Disord. 3, 304 (2009).

    Article  Google Scholar 

  139. Lotan, M., Schenker, R., Wine, J. & Downs, J. The conductive environment enhances gross motor function of girls with Rett syndrome. A pilot study. Dev. Neurorehabil. 15, 19–25 (2012).

    Article  PubMed  Google Scholar 

  140. Glaze, D. G., Frost, J. D. Jr, Zoghbi, H. Y. & Percy, A. K. Rett's syndrome. Correlation of electroencephalographic characteristics with clinical staging. Arch. Neurol. 44, 1053–1056 (1987).

    Article  CAS  PubMed  Google Scholar 

  141. Glaze, D. G., Schultz, R. J. & Frost, J. D. Rett syndrome: characterization of seizures versus non-seizures. Electroencephalogr. Clin. Neurophysiol. 106, 79–83 (1998).

    Article  CAS  PubMed  Google Scholar 

  142. Steffenburg, U., Hagberg, G. & Hagberg, B. Epilepsy in a representative series of Rett syndrome. Acta Paediatr. 90, 34–39 (2001).

    Article  CAS  PubMed  Google Scholar 

  143. Jian, L. et al. Predictors of seizure onset in Rett syndrome. J. Pediatr. 149, 542–547 (2006).

    Article  PubMed  Google Scholar 

  144. Jian, L. et al. Seizures in Rett syndrome: an overview from a one-year calendar study. Eur. J. Paediatr. Neurol. 11, 310–317 (2007).

    Article  PubMed  PubMed Central  Google Scholar 

  145. Glaze, D. G. et al. Epilepsy and the natural history of Rett syndrome. Neurology 74, 909–912 (2010). This study used the Rare Disease Consortium Research Network for Rett syndrome to identify 602 cases who met the criteria for classic or atypical Rett syndrome. Just under half of the cohort had seizures according to physician assessment. Individuals with the MECP2 Thr158Met or Arg106Trp mutation were most likely to have epilepsy (74% and 78%, respectively), and those with the Arg255X or Arg306Cys mutation were least likely to have epilepsy (both 49%).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  146. Bao, X., Downs, J., Wong, K., Williams, S. & Leonard, H. Using a large international sample to investigate epilepsy in Rett syndrome. Dev. Med. Child Neurol. 55, 553–558 (2013).

    Article  PubMed  Google Scholar 

  147. Nissenkorn, A. et al. Epilepsy in Rett syndrome — lessons from the Rett networked database. Epilepsia 56, 569–576 (2015).

    Article  CAS  PubMed  Google Scholar 

  148. Schultz, R., Glaze, D., Motil, K., Hebert, D. & Percy, A. Hand and foot growth failure in Rett syndrome. J. Child Neurol. 13, 71–74 (1998).

    Article  CAS  PubMed  Google Scholar 

  149. Motil, K. J., Schultz, R., Brown, B., Glaze, D. G. & Percy, A. K. Altered energy balance may account for growth failure in Rett syndrome. J. Child Neurol. 9, 315–319 (1994).

    Article  CAS  PubMed  Google Scholar 

  150. Oddy, W. H. et al. Feeding experiences and growth status in a Rett syndrome population. J. Pediatr. Gastroenterol. Nutr. 45, 582–590 (2007).

    Article  PubMed  Google Scholar 

  151. Platte, P., Jaschke, H., Herbert, C. & Korenke, G. C. Increased resting metabolic rate in girls with Rett syndrome compared to girls with developmental disabilities. Neuropediatrics 42, 179–182 (2011).

    Article  CAS  PubMed  Google Scholar 

  152. Tarquinio, D. C. et al. Growth failure and outcome in Rett syndrome: specific growth references. Neurology 79, 1653–1661 (2012). This study created growth charts for head circumference, weight, height and BMI on the basis of 9,749 observations of 816 females with Rett syndrome. Growth was decreased compared with a normative US population, and pubertal increases in height and weight were not observed.

    Article  PubMed  PubMed Central  Google Scholar 

  153. Downs, J. et al. Experience of gastrostomy using a quality care framework: the example of Rett syndrome. Medicine (Baltimore) 93, e328 (2014).

    Article  Google Scholar 

  154. Leonard, H. et al. Assessment and management of nutrition and growth in Rett syndrome. J. Pediatr. Gastroenterol. Nutr. 57, 451–460 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  155. Julu, P. O. et al. Characterisation of breathing and associated central autonomic dysfunction in the Rett disorder. Arch. Dis. Child. 85, 29–37 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  156. Weese-Mayer, D. E. et al. Autonomic nervous system dysregulation: breathing and heart rate perturbation during wakefulness in young girls with Rett syndrome. Pediatr. Res. 60, 443–449 (2006).

    Article  PubMed  Google Scholar 

  157. Baikie, G. et al. Gastrointestinal dysmotility in Rett syndrome. J. Pediatr. Gastroenterol. Nutr. 58, 237–244 (2014).

    Article  PubMed  Google Scholar 

  158. Lioy, D. T., Wu, W. W. & Bissonnette, J. M. Autonomic dysfunction with mutations in the gene that encodes methyl-CpG-binding protein 2: insights into Rett syndrome. Auton. Neurosci. 161, 55–62 (2011).

    Article  CAS  PubMed  Google Scholar 

  159. Hagberg, B. & Romell, M. Rett females: patterns of characteristic side-asymmetric neuroimpairments at long-term follow-up. Neuropediatrics 33, 324–326 (2002).

    Article  CAS  PubMed  Google Scholar 

  160. Downs, J. et al. The natural history of scoliosis in females with Rett syndrome. Spine (Phila. Pa 1976) 41, 856–863 (2016).

    Article  Google Scholar 

  161. Percy, A. K. et al. Profiling scoliosis in Rett syndrome. Pediatr. Res. 67, 435–439 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  162. Sponseller, P. D., Yazici, M., Demetracopoulos, C. & Emans, J. B. Evidence basis for management of spine and chest wall deformities in children. Spine (Phila. Pa 1976) 32, S81–S90 (2007).

    Article  Google Scholar 

  163. Downs, J. et al. Guidelines for management of scoliosis in Rett syndrome patients based on expert consensus and clinical evidence. Spine (Phila. Pa 1976) 34, E607–E617 (2009).

    Article  Google Scholar 

  164. Downs, J. et al. Surgical fusion of early onset severe scoliosis increases survival in Rett syndrome: a cohort study. Dev. Med. Child Neurol. 58, 632–638 (2016). Using the Australian population-based database, this study demonstrated the effects of spinal fusion to treat severe scoliosis in Rett syndrome. The findings indicated that survival was better in individuals who underwent surgery than in those who received conservative management, especially if scoliosis developed before 8 years of age.

    Article  PubMed  Google Scholar 

  165. Downs, J., Forbes, D., Johnson, M. & Leonard, H. How can clinical ethics guide the management of comorbidities in the child with Rett syndrome? J. Paediatr. Child Health 52, 809–813 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  166. Ellaway, C., Peat, J., Leonard, H. & Christodoulou, J. Sleep dysfunction in Rett syndrome: lack of age related decrease in sleep duration. Brain Dev. 23, S101–S103 (2001).

    Article  PubMed  Google Scholar 

  167. Young, D. et al. Sleep problems in Rett syndrome. Brain Dev. 29, 609–616 (2007).

    Article  PubMed  PubMed Central  Google Scholar 

  168. Wong, K., Leonard, H., Jacoby, P., Ellaway, C. & Downs, J. The trajectories of sleep disturbances in Rett syndrome. J. Sleep Res. 24, 223–233 (2015).

    Article  PubMed  Google Scholar 

  169. Boban, S. et al. Determinants of sleep disturbances in Rett syndrome: novel findings in relation to genotype. Am. J. Med. Genet. A 170, 2292–2300 (2016).

    Article  CAS  PubMed  Google Scholar 

  170. McArthur, A. J. & Budden, S. S. Sleep dysfunction in Rett syndrome: a trial of exogenous melatonin treatment. Dev. Med. Child Neurol. 40, 186–192 (1998).

    Article  CAS  PubMed  Google Scholar 

  171. Haas, R. H., Dixon, S. D., Sartoris, D. J. & Hennessy, M. J. Osteopenia in Rett syndrome. J. Pediatr. 131, 771–774 (1997).

    Article  CAS  PubMed  Google Scholar 

  172. Leonard, H. et al. A population-based approach to the investigation of osteopenia in Rett syndrome. Dev. Med. Child Neurol. 41, 323–328 (1999).

    Article  CAS  PubMed  Google Scholar 

  173. Downs, J. et al. Early determinants of fractures in Rett syndrome. Pediatrics 121, 540–546 (2008).

    Article  PubMed  Google Scholar 

  174. Roende, G. et al. DXA measurements in Rett syndrome reveal small bones with low bone mass. J. Bone Miner. Res. 26, 2280–2286 (2011).

    Article  CAS  PubMed  Google Scholar 

  175. Roende, G. et al. Patients with Rett syndrome sustain low-energy fractures. Pediatr. Res. 69, 359–364 (2011).

    Article  PubMed  Google Scholar 

  176. Motil, K. J., Ellis, K. J., Barrish, J. O., Caeg, E. & Glaze, D. G. Bone mineral content and bone mineral density are lower in older than in younger females with Rett syndrome. Pediatr. Res. 64, 435–439 (2008).

    Article  PubMed  PubMed Central  Google Scholar 

  177. Shapiro, J. R. et al. Bone mass in Rett syndrome: association with clinical parameters and MECP2 mutations. Pediatr. Res. 68, 446–451 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  178. Jefferson, A. L. et al. Bone mineral content and density in Rett syndrome and their contributing factors. Pediatr. Res. 69, 293–298 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  179. Jefferson, A. et al. Longitudinal bone mineral content and density in Rett syndrome and their contributing factors. Bone 74, 191–198 (2015).

    Article  PubMed  Google Scholar 

  180. Motil, K. J. et al. Vitamin D deficiency is prevalent in girls and women with Rett syndrome. J. Pediatr. Gastroenterol. Nutr. 53, 569–574 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  181. Leonard, H. et al. Valproate and risk of fracture in Rett syndrome. Arch. Dis. Child. 95, 444–448 (2010).

    Article  CAS  PubMed  Google Scholar 

  182. Roende, G. et al. Low bone turnover phenotype in Rett syndrome: results of biochemical bone marker analysis. Pediatr. Res. 75, 551–558 (2014).

    Article  CAS  PubMed  Google Scholar 

  183. Jefferson, A. et al. Clinical guidelines for management of bone health in Rett syndrome based on expert consensus and available evidence. PLoS ONE 11, e0146824 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  184. Lotan, M., Reves-Siesel, R., Eliav-Shalev, R. S. & Merrick, J. Osteoporosis in Rett syndrome: a case study presenting a novel management intervention for severe osteoporosis. Osteoporos. Int. 24, 3059–3063 (2013).

    Article  CAS  PubMed  Google Scholar 

  185. Katz, D. M. et al. Rett syndrome: crossing the threshold to clinical translation. Trends Neurosci. 39, 100–113 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  186. Gadalla, K. K., Bailey, M. E. & Cobb, S. R. MeCP2 and Rett syndrome: reversibility and potential avenues for therapy. Biochem. J. 439, 1–14 (2011).

    Article  CAS  PubMed  Google Scholar 

  187. Pozzo-Miller, L., Pati, S. & Percy, A. K. Rett syndrome: reaching for clinical trials. Neurotherapeutics 12, 631–640 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  188. Ricceri, L., De Filippis, B. & Laviola, G. Rett syndrome treatment in mouse models: searching for effective targets and strategies. Neuropharmacology 68, 106–115 (2013).

    Article  CAS  PubMed  Google Scholar 

  189. Lang, M. et al. Rescue of behavioral and EEG deficits in male and female Mecp2-deficient mice by delayed Mecp2 gene reactivation. Hum. Mol. Genet. 23, 303–318 (2014).

    Article  CAS  PubMed  Google Scholar 

  190. Gadalla, K. K. et al. Improved survival and reduced phenotypic severity following AAV9/MECP2 gene transfer to neonatal and juvenile male Mecp2 knockout mice. Mol. Ther. 21, 18–30 (2013).

    Article  CAS  PubMed  Google Scholar 

  191. Garg, S. K. et al. Systemic delivery of MeCP2 rescues behavioral and cellular deficits in female mouse models of Rett syndrome. J. Neurosci. 33, 13612–13620 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  192. Gadalla, K. E. et al. Gene therapy for Rett syndrome: prospects and challenges. Future Neurol. 10, 467–484 (2015).

    Article  CAS  Google Scholar 

  193. Chao, H. T. & Zoghbi, H. Y. MeCP2: only 100% will do. Nat. Neurosci. 15, 176–177 (2012).

    Article  CAS  PubMed  Google Scholar 

  194. Bhatnagar, S. et al. Genetic and pharmacological reactivation of the mammalian inactive X chromosome. Proc. Natl Acad. Sci. USA 111, 12591–12598 (2014).

    Article  CAS  PubMed  Google Scholar 

  195. Vecsler, M. et al. Ex vivo treatment with a novel synthetic aminoglycoside NB54 in primary fibroblasts from Rett syndrome patients suppresses MECP2 nonsense mutations. PLoS ONE 6, e20733 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  196. Brendel, C. et al. Readthrough of nonsense mutations in Rett syndrome: evaluation of novel aminoglycosides and generation of a new mouse model. J. Mol. Med. (Berl.) 89, 389–398 (2011).

    Article  CAS  Google Scholar 

  197. Schanen, C. et al. Phenotypic manifestations of MECP2 mutations in classical and atypical Rett syndrome. Am. J. Med. Genet. A 126A, 129–140 (2004).

    Article  PubMed  Google Scholar 

  198. Khwaja, O. S. et al. Safety, pharmacokinetics, and preliminary assessment of efficacy of mecasermin (recombinant human IGF-1) for the treatment of Rett syndrome. Proc. Natl Acad. Sci. USA 111, 4596–4601 (2014).

    Article  CAS  PubMed  Google Scholar 

  199. FitzGerald, P. M., Jankovic, J. & Percy, A. K. Rett syndrome and associated movement disorders. Mov. Disord. 5, 195–202 (1990).

    Article  CAS  PubMed  Google Scholar 

  200. Mount, R. H., Charman, T., Hastings, R. P., Reilly, S. & Cass, H. The Rett Syndrome Behaviour Questionnaire (RSBQ): refining the behavioural phenotype of Rett syndrome. J. Child Psychol. Psychiatry 43, 1099–1110 (2002).

    Article  PubMed  Google Scholar 

  201. Robertson, L. et al. The association between behavior and genotype in Rett syndrome using the Australian Rett Syndrome Database. Am. J. Med. Genet. B Neuropsychiatr. Genet. 141B, 177–183 (2006).

    Article  PubMed  PubMed Central  Google Scholar 

  202. Barnes, K. V. et al. Anxiety-like behavior in Rett syndrome: characteristics and assessment by anxiety scales. J. Neurodev. Disord. 7, 30 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  203. Neul, J. L. et al. Improving treatment trial outcomes for Rett syndrome: the development of Rett-specific anchors for the Clinical Global Impression Scale. J. Child Neurol. 30, 1743–1748 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  204. Weese-Mayer, D. E. et al. Autonomic dysregulation in young girls with Rett syndrome during nighttime in-home recordings. Pediatr. Pulmonol. 43, 1045–1060 (2008).

    Article  PubMed  Google Scholar 

  205. Kozinetz, C. A. et al. Epidemiology of Rett syndrome: a population-based registry. Pediatrics 91, 445–450 (1993).

    CAS  PubMed  Google Scholar 

  206. Leonard, H., Bower, C. & English, D. The prevalence and incidence of Rett syndrome in Australia. Eur. Child Adolesc. Psychiatry 6 (Suppl. 1), 8–10 (1997).

    PubMed  Google Scholar 

  207. Fehr, S. et al. Trends in the diagnosis of Rett syndrome in Australia. Pediatr. Res. 70, 313–319 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  208. Corbett, J. & Kerr, A. Rett syndrome: from gene to gesture. J. R. Soc. Med. 87, 562–566 (1994).

    PubMed Central  Google Scholar 

  209. Young, D. et al. The relationship between MECP2 mutation type and health status and service use trajectories over time in a Rett syndrome population. Res. Autism Spectr. Disord. 5, 442–449 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  210. Colvin, L. et al. Refining the phenotype of common mutations in Rett syndrome. J. Med. Genet. 41, 25–30 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  211. Leonard, H. et al. Resourceful and creative methods are necessary to research rare disorders. Dev. Med. Child Neurol. 55, 870–871 (2013).

    Article  PubMed  Google Scholar 

  212. Percy, A. The American history of Rett syndrome. Pediatr. Neurol. 50, 1–3 (2014).

    Article  PubMed  Google Scholar 

  213. Louise, S. et al. InterRett, a model for international data collection in a rare genetic disorder. Res. Autism Spectr. Disord. 3, 639–659 (2009).

    Article  Google Scholar 

  214. Christodoulou, J., Grimm, A., Maher, T. & Bennetts, B. RettBASE: the IRSA MECP2 variation database — a new mutation database in evolution. Hum. Mutat. 21, 466–472 (2003).

    Article  CAS  PubMed  Google Scholar 

  215. Hunter, K. Role of the International Rett Syndrome Association. J. Child Neurol. 3, S87–S88 (1988).

    Article  PubMed  Google Scholar 

  216. Hunter, K. Looking from the inside out: a parent's perspective. Ment. Retard. Dev. Disabil. Res. Rev. 8, 77–81 (2002).

    Article  PubMed  Google Scholar 

  217. Kerr, A. M., Webb, P., Prescott, R. J. & Milne, Y. Results of surgery for scoliosis in Rett syndrome. J. Child Neurol. 18, 703–708 (2003).

    Article  PubMed  Google Scholar 

  218. Downs, J. et al. Family satisfaction following spinal fusion in Rett syndrome. Dev. Neurorehabil. 19, 31–37 (2016).

    Article  PubMed  Google Scholar 

  219. Freilinger, M. et al. Survival with Rett syndrome: comparing Rett's original sample with data from the Australian Rett Syndrome Database. Dev. Med. Child Neurol. 52, 962–965 (2010). This study compared survival in Rett's original cohort with an Australian population-based cohort in 2009, and demonstrated that survival at 25 years had increased from 21% to 71%. These findings have major implications for the clinical care of individuals with Rett syndrome into adulthood.

    Article  PubMed  Google Scholar 

  220. Kirby, R. S. et al. Longevity in Rett syndrome: analysis of the North American Database. J. Pediatr. 156, 135–138.e1 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  221. Tarquinio, D. C. et al. The changing face of survival in Rett syndrome and MECP2-related disorders. Pediatr. Neurol. 53, 402–411 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  222. Leonard, H. et al. How can the Internet help parents of children with rare neurologic disorders? J. Child Neurol. 19, 902–907 (2004).

    Article  PubMed  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Contributions

The authors contributed equally to all aspects of the manuscript.

Corresponding author

Correspondence to Helen Leonard.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Supplementary information

Supplementary Video 1

Severe Rett syndrome phenotype. (MP4 27289 kb)

Supplementary Video 2

Mild Rett syndrome phenotype. (MP4 28097 kb)

PowerPoint slides

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Leonard, H., Cobb, S. & Downs, J. Clinical and biological progress over 50 years in Rett syndrome. Nat Rev Neurol 13, 37–51 (2017). https://doi.org/10.1038/nrneurol.2016.186

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nrneurol.2016.186

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing