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.

  • Original Article
  • Published:

MeCP2-regulated miRNAs control early human neurogenesis through differential effects on ERK and AKT signaling

Abstract

Rett syndrome (RTT) is an X-linked, neurodevelopmental disorder caused primarily by mutations in the methyl-CpG-binding protein 2 (MECP2) gene, which encodes a multifunctional epigenetic regulator with known links to a wide spectrum of neuropsychiatric disorders. Although postnatal functions of MeCP2 have been thoroughly investigated, its role in prenatal brain development remains poorly understood. Given the well-established importance of microRNAs (miRNAs) in neurogenesis, we employed isogenic human RTT patient-derived induced pluripotent stem cell (iPSC) and MeCP2 short hairpin RNA knockdown approaches to identify novel MeCP2-regulated miRNAs enriched during early human neuronal development. Focusing on the most dysregulated miRNAs, we found miR-199 and miR-214 to be increased during early brain development and to differentially regulate extracellular signal-regulated kinase (ERK)/mitogen-activated protein kinase and protein kinase B (PKB/AKT) signaling. In parallel, we characterized the effects on human neurogenesis and neuronal differentiation brought about by MeCP2 deficiency using both monolayer and three-dimensional (cerebral organoid) patient-derived and MeCP2-deficient neuronal culture models. Inhibiting miR-199 or miR-214 expression in iPSC-derived neural progenitors deficient in MeCP2 restored AKT and ERK activation, respectively, and ameliorated the observed alterations in neuronal differentiation. Moreover, overexpression of miR-199 or miR-214 in the wild-type mouse embryonic brains was sufficient to disturb neurogenesis and neuronal migration in a similar manner to Mecp2 knockdown. Taken together, our data support a novel miRNA-mediated pathway downstream of MeCP2 that influences neurogenesis via interactions with central molecular hubs linked to autism spectrum disorders.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Purchase on Springer Link

Instant access to full article PDF

Prices may be subject to local taxes which are calculated during checkout

Figure 1
Figure 2
Figure 3
Figure 4
Figure 5

Similar content being viewed by others

References

  1. Krol J, Loedige I, Filipowicz W . The widespread regulation of microRNA biogenesis, function and decay. Nat Rev Genet 2010; 11: 597–610.

    Article  CAS  PubMed  Google Scholar 

  2. Coolen M, Bally-Cuif L . MicroRNAs in brain development and physiology. Curr Opin Neurobiol 2009; 19: 461–470.

    Article  CAS  PubMed  Google Scholar 

  3. Mellios N, Sur M . The emerging role of microRNAs in schizophrenia and autism spectrum disorders. Front Psychiatry 2012; 3: 39.

    Article  PubMed  PubMed Central  Google Scholar 

  4. Mellios N, Sugihara H, Castro J, Banerjee A, Le C, Kumar A et al. miR-132, an experience-dependent microRNA, is essential for visual cortex plasticity. Nat Neurosci 2011; 14: 1240–1242.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Amir RE, Van den Veyver IB, Wan M, Tran CQ, Francke U, Zoghbi HY . Rett syndrome is caused by mutations in X-linked MECP2, encoding methyl-CpG-binding protein 2. Nat Genet 1999; 23: 185–188.

    Article  CAS  PubMed  Google Scholar 

  6. Castro J, Mellios N, Sur M . Mechanisms and therapeutic challenges in autism spectrum disorders: insights from Rett syndrome. Curr Opin Neurol 2013; 26: 154–159.

    Article  CAS  PubMed  Google Scholar 

  7. Cheng TL, Wang Z, Liao Q, Zhu Y, Zhou WH, Xu W et al. MeCP2 suppresses nuclear microRNA processing and dendritic growth by regulating the DGCR8/Drosha complex. Dev Cell 2014; 28: 547–560.

    Article  CAS  PubMed  Google Scholar 

  8. Szulwach KE, Li X, Smrt RD, Li Y, Luo Y, Lin L et al. Cross talk between microRNA and epigenetic regulation in adult neurogenesis. J Cell Biol 2010; 189: 127–141.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Urdinguio RG, Fernandez AF, Lopez-Nieva P, Rossi S, Huertas D, Kulis M et al. Disrupted microRNA expression caused by Mecp2 loss in a mouse model of Rett syndrome. Epigenetics 2010; 5: 656–663.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Wu H, Tao J, Chen PJ, Shahab A, Ge W, Hart RP et al. Genome-wide analysis reveals methyl-CpG-binding protein 2-dependent regulation of microRNAs in a mouse model of Rett syndrome. Proc Natl Acad Sci USA 2010; 107: 18161–18166.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Neul JL, Zoghbi HY . Rett syndrome: a prototypical neurodevelopmental disorder. Neuroscientist 2004; 10: 118–128.

    Article  CAS  PubMed  Google Scholar 

  12. Einspieler C, Kerr AM, Prechtl HF . Is the early development of girls with Rett disorder really normal? Pediatr Res 2005; 57: 696–700.

    Article  PubMed  Google Scholar 

  13. Kerr AM . Early clinical signs in the Rett disorder. Neuropediatrics 1995; 26: 67–71.

    Article  CAS  PubMed  Google Scholar 

  14. Schanen NC, Kurczynski TW, Brunelle D, Woodcock MM, Dure LSt, Percy AK . Neonatal encephalopathy in two boys in families with recurrent Rett syndrome. J Child Neurol 1998; 13: 229–231.

    Article  CAS  PubMed  Google Scholar 

  15. Trappe R, Laccone F, Cobilanschi J, Meins M, Huppke P, Hanefeld F et al. MECP2 mutations in sporadic cases of Rett syndrome are almost exclusively of paternal origin. Am J Hum Genet 2001; 68: 1093–1101.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Castro J, Garcia RI, Kwok S, Banerjee A, Petravicz J, Woodson J et al. Functional recovery with recombinant human IGF1 treatment in a mouse model of Rett Syndrome. Proc Natl Acad Sci USA 2014; 111: 9941–9946.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Giacometti E, Luikenhuis S, Beard C, Jaenisch R . Partial rescue of MeCP2 deficiency by postnatal activation of MeCP2. Proc Natl Acad Sci USA 2007; 104: 1931–1936.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Khwaja OS, Ho E, Barnes KV, O'Leary HM, Pereira LM, Finkelstein Y 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 2014; 111: 4596–4601.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Mellios N, Woodson J, Garcia RI, Crawford B, Sharma J, Sheridan SD et al. beta2-Adrenergic receptor agonist ameliorates phenotypes and corrects microRNA-mediated IGF1 deficits in a mouse model of Rett syndrome. Proc Natl Acad Sci USA 2014; 111: 9947–9952.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Fukuda T, Itoh M, Ichikawa T, Washiyama K, Goto Y . Delayed maturation of neuronal architecture and synaptogenesis in cerebral cortex of Mecp2-deficient mice. J Neuropathol Exp Neurol 2005; 64: 537–544.

    Article  CAS  PubMed  Google Scholar 

  22. Kishi N, Macklis JD . MECP2 is progressively expressed in post-migratory neurons and is involved in neuronal maturation rather than cell fate decisions. Mol Cell Neurosci 2004; 27: 306–321.

    Article  CAS  PubMed  Google Scholar 

  23. Shahbazian MD, Antalffy B, Armstrong DL, Zoghbi HY . Insight into Rett syndrome: MeCP2 levels display tissue- and cell-specific differences and correlate with neuronal maturation. Hum Mol Genet 2002; 11: 115–124.

    Article  CAS  PubMed  Google Scholar 

  24. Armstrong DD, Deguchi K, Antallfy B . Survey of MeCP2 in the Rett syndrome and the non-Rett syndrome brain. J Child Neurol 2003; 18: 683–687.

    Article  PubMed  Google Scholar 

  25. Petazzi P, Akizu N, Garcia A, Estaras C, Martinez de Paz A, Rodriguez-Paredes M et al. An increase in MECP2 dosage impairs neural tube formation. Neurobiol Dis 2014; 67: 49–56.

    Article  CAS  PubMed  Google Scholar 

  26. Tsujimura K, Abematsu M, Kohyama J, Namihira M, Nakashima K . Neuronal differentiation of neural precursor cells is promoted by the methyl-CpG-binding protein MeCP2. Exp Neurol 2009; 219: 104–111.

    Article  CAS  PubMed  Google Scholar 

  27. Bedogni F, Cobolli Gigli C, Pozzi D, Rossi RL, Scaramuzza L, Rossetti G et al. Defects during Mecp2 null embryonic cortex development precede the onset of overt neurological symptoms. Cereb Cortex 2016; 26: 2517–2529.

    Article  PubMed  Google Scholar 

  28. Farra N, Zhang WB, Pasceri P, Eubanks JH, Salter MW, Ellis J . Rett syndrome induced pluripotent stem cell-derived neurons reveal novel neurophysiological alterations. Mol Psychiatry 2012; 17: 1261–1271.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Kim KY, Hysolli E, Park IH . Neuronal maturation defect in induced pluripotent stem cells from patients with Rett syndrome. Proc Natl Acad Sci USA 2011; 108: 14169–14174.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Li Y, Wang H, Muffat J, Cheng AW, Orlando DA, Loven J et al. Global transcriptional and translational repression in human-embryonic-stem-cell-derived Rett syndrome neurons. Cell Stem Cell 2013; 13: 446–458.

    Article  PubMed  PubMed Central  Google Scholar 

  31. Marchetto MC, Carromeu C, Acab A, Yu D, Yeo GW, Mu Y et al. A model for neural development and treatment of Rett syndrome using human induced pluripotent stem cells. Cell 2010; 143: 527–539.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Williams EC, Zhong X, Mohamed A, Li R, Liu Y, Dong Q et al. Mutant astrocytes differentiated from Rett syndrome patients-specific iPSCs have adverse effects on wild-type neurons. Hum Mol Genet 2014; 23: 2968–2980.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Fernandes TG, Duarte ST, Ghazvini M, Gaspar C, Santos DC, Porteira AR et al. Neural commitment of human pluripotent stem cells under defined conditions recapitulates neural development and generates patient-specific neural cells. Biotechnol J 2015; 10: 1578–1588.

    Article  CAS  PubMed  Google Scholar 

  34. Park IH, Zhao R, West JA, Yabuuchi A, Huo H, Ince TA et al. Reprogramming of human somatic cells to pluripotency with defined factors. Nature 2008; 451: 141–146.

    Article  CAS  PubMed  Google Scholar 

  35. Sheridan SD, Theriault KM, Reis SA, Zhou F, Madison JM, Daheron L et al. Epigenetic characterization of the FMR1 gene and aberrant neurodevelopment in human induced pluripotent stem cell models of fragile X syndrome. PLoS ONE 2011; 6: e26203.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Abraham S, Sheridan SD, Laurent LC, Albert K, Stubban C, Ulitsky I et al. Propagation of human embryonic and induced pluripotent stem cells in an indirect co-culture system. Biochem Biophys Res Commun 2010; 393: 211–216.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Sheridan SD, Surampudi V, Rao RR . Analysis of embryoid bodies derived from human induced pluripotent stem cells as a means to assess pluripotency. Stem Cells Int 2012; 2012: 738910.

    Article  PubMed  PubMed Central  Google Scholar 

  38. Shi Y, Kirwan P, Smith J, Robinson HP, Livesey FJ . Human cerebral cortex development from pluripotent stem cells to functional excitatory synapses. Nat Neurosci 2012; 15: 477–486, S471.

    Article  CAS  PubMed  Google Scholar 

  39. Lancaster MA, Renner M, Martin CA, Wenzel D, Bicknell LS, Hurles ME et al. Cerebral organoids model human brain development and microcephaly. Nature 2013; 501: 373–379.

    Article  CAS  PubMed  Google Scholar 

  40. Lee YB, Bantounas I, Lee DY, Phylactou L, Caldwell MA, Uney JB . Twist-1 regulates the miR-199a/214 cluster during development. Nucleic Acids Res 2009; 37: 123–128.

    Article  CAS  PubMed  Google Scholar 

  41. Florio M, Huttner WB . Neural progenitors, neurogenesis and the evolution of the neocortex. Development 2014; 141: 2182–2194.

    Article  CAS  PubMed  Google Scholar 

  42. Englund C, Fink A, Lau C, Pham D, Daza RA, Bulfone A et al. Pax6, Tbr2, and Tbr1 are expressed sequentially by radial glia, intermediate progenitor cells, and postmitotic neurons in developing neocortex. J Neurosci 2005; 25: 247–251.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Cobos I, Calcagnotto ME, Vilaythong AJ, Thwin MT, Noebels JL, Baraban SC et al. Mice lacking Dlx1 show subtype-specific loss of interneurons, reduced inhibition and epilepsy. Nat Neurosci 2005; 8: 1059–1068.

    Article  CAS  PubMed  Google Scholar 

  44. Penna E, Orso F, Taverna D . miR-214 as a key hub that controls cancer networks: small player, multiple functions. J Invest Dermatol 2015; 135: 960–969.

    Article  CAS  PubMed  Google Scholar 

  45. Hou J, Lin L, Zhou W, Wang Z, Ding G, Dong Q et al. Identification of miRNomes in human liver and hepatocellular carcinoma reveals miR-199a/b-3p as therapeutic target for hepatocellular carcinoma. Cancer Cell 2011; 19: 232–243.

    Article  CAS  PubMed  Google Scholar 

  46. Pucilowska J, Puzerey PA, Karlo JC, Galan RF, Landreth GE . Disrupted ERK signaling during cortical development leads to abnormal progenitor proliferation, neuronal and network excitability and behavior, modeling human neuro-cardio-facial-cutaneous and related syndromes. J Neurosci 2012; 32: 8663–8677.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Samuels IS, Karlo JC, Faruzzi AN, Pickering K, Herrup K, Sweatt JD et al. Deletion of ERK2 mitogen-activated protein kinase identifies its key roles in cortical neurogenesis and cognitive function. J Neurosci 2008; 28: 6983–6995.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Zhang J, Shemezis JR, McQuinn ER, Wang J, Sverdlov M, Chenn A . AKT activation by N-cadherin regulates beta-catenin signaling and neuronal differentiation during cortical development. Neural Dev 2013; 8: 7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Gonzales ML, LaSalle JM . The role of MeCP2 in brain development and neurodevelopmental disorders. Curr Psychiatry Rep 2010; 12: 127–134.

    Article  PubMed  PubMed Central  Google Scholar 

  50. Hammer S, Dorrani N, Dragich J, Kudo S, Schanen C . The phenotypic consequences of MECP2 mutations extend beyond Rett syndrome. Ment Retard Dev Disabil Res Rev 2002; 8: 94–98.

    Article  PubMed  Google Scholar 

  51. Akbarian S, Chen RZ, Gribnau J, Rasmussen TP, Fong H, Jaenisch R et al. Expression pattern of the Rett syndrome gene MeCP2 in primate prefrontal cortex. Neurobiol Dis 2001; 8: 784–791.

    Article  CAS  PubMed  Google Scholar 

  52. Chen RZ, 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 2001; 27: 327–331.

    Article  CAS  PubMed  Google Scholar 

  53. Guy J, Hendrich B, Holmes M, Martin JE, Bird A . A mouse Mecp2-null mutation causes neurological symptoms that mimic Rett syndrome. Nat Genet 2001; 27: 322–326.

    Article  CAS  PubMed  Google Scholar 

  54. Luikenhuis S, Giacometti E, Beard CF, Jaenisch R . Expression of MeCP2 in postmitotic neurons rescues Rett syndrome in mice. Proc Natl Acad Sci USA 2004; 101: 6033–6038.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Chang Q, Khare G, Dani V, Nelson S, Jaenisch R . The disease progression of Mecp2 mutant mice is affected by the level of BDNF expression. Neuron 2006; 49: 341–348.

    Article  CAS  PubMed  Google Scholar 

  56. Ogier M, Wang H, Hong E, Wang Q, Greenberg ME, Katz DM . Brain-derived neurotrophic factor expression and respiratory function improve after ampakine treatment in a mouse model of Rett syndrome. J Neurosci 2007; 27: 10912–10917.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Schmid DA, Yang T, Ogier M, Adams I, Mirakhur Y, Wang Q et al. A TrkB small molecule partial agonist rescues TrkB phosphorylation deficits and improves respiratory function in a mouse model of Rett syndrome. J Neurosci 2012; 32: 1803–1810.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Tropea D, Giacometti E, Wilson NR, Beard C, McCurry C, Fu DD et al. Partial reversal of Rett Syndrome-like symptoms in MeCP2 mutant mice. Proc Natl Acad Sci USA 2009; 106: 2029–2034.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Davis BN, Hilyard AC, Lagna G, Hata A . SMAD proteins control DROSHA-mediated microRNA maturation. Nature 2008; 454: 56–61.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Bauman ML, Kemper TL, Arin DM . Pervasive neuroanatomic abnormalities of the brain in three cases of Rett's syndrome. Neurology 1995; 45: 1581–1586.

    Article  CAS  PubMed  Google Scholar 

  61. Armstrong DD . Rett syndrome neuropathology review 2000. Brain Dev 2001; 23 (Suppl 1): S72–S76.

    Article  PubMed  Google Scholar 

  62. Armstrong D, Dunn JK, Antalffy B, Trivedi R . Selective dendritic alterations in the cortex of Rett syndrome. J Neuropathol Exp Neurol 1995; 54: 195–201.

    Article  CAS  PubMed  Google Scholar 

  63. Mariani J, Coppola G, Zhang P, Abyzov A, Provini L, Tomasini L et al. FOXG1-dependent dysregulation of GABA/glutamate neuron differentiation in autism spectrum disorders. Cell 2015; 162: 375–390.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Ariani F, Hayek G, Rondinella D, Artuso R, Mencarelli MA, Spanhol-Rosseto A et al. FOXG1 is responsible for the congenital variant of Rett syndrome. Am J Hum Genet 2008; 83: 89–93.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Ito-Ishida A, Ure K, Chen H, Swann JW, Zoghbi HY . Loss of MeCP2 in parvalbumin-and somatostatin-expressing neurons in mice leads to distinct Rett syndrome-like phenotypes. Neuron 2015; 88: 651–658.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Selvamani A, Sathyan P, Miranda RC, Sohrabji F . An antagomir to microRNA Let7f promotes neuroprotection in an ischemic stroke model. PLoS ONE 2012; 7: e32662.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Cimadamore F, Amador-Arjona A, Chen C, Huang CT, Terskikh AV . SOX2-LIN28/let-7 pathway regulates proliferation and neurogenesis in neural precursors. Proc Natl Acad Sci USA 2013; 110: E3017–E3026.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Tsujimura K, Irie K, Nakashima H, Egashira Y, Fukao Y, Fujiwara M et al. miR-199a links MeCP2 with mTOR signaling and its dysregulation leads to Rett syndrome phenotypes. Cell Rep 2015; 12: 1887–1901.

    Article  CAS  PubMed  Google Scholar 

  69. del Gaudio D, Fang P, Scaglia F, Ward PA, Craigen WJ, Glaze DG et al. Increased MECP2 gene copy number as the result of genomic duplication in neurodevelopmentally delayed males. Genet Med 2006; 8: 784–792.

    Article  CAS  PubMed  Google Scholar 

  70. Van Esch H, Bauters M, Ignatius J, Jansen M, Raynaud M, Hollanders K 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 2005; 77: 442–453.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Klein ME, Lioy DT, Ma L, Impey S, Mandel G, Goodman RH . Homeostatic regulation of MeCP2 expression by a CREB-induced microRNA. Nat Neurosci 2007; 10: 1513–1514.

    Article  CAS  PubMed  Google Scholar 

  72. Ricciardi S, Boggio EM, Grosso S, Lonetti G, Forlani G, Stefanelli G et al. Reduced AKT/mTOR signaling and protein synthesis dysregulation in a Rett syndrome animal model. Hum Mol Genet 2011; 20: 1182–1196.

    Article  CAS  PubMed  Google Scholar 

  73. Casanova EL, Casanova MF . Genetics studies indicate that neural induction and early neuronal maturation are disturbed in autism. Front Cell Neurosci 2014; 8: 397.

    PubMed  PubMed Central  Google Scholar 

  74. Gulsuner S, Walsh T, Watts AC, Lee MK, Thornton AM, Casadei S et al. Spatial and temporal mapping of de novo mutations in schizophrenia to a fetal prefrontal cortical network. Cell 2013; 154: 518–529.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank Ting Fu, Tatiana Karadimitriou, Chuong Le, Anita Liu, Jonathan Woodson and Arooshi Kumar for excellent technical support and Chloe Delepine for advice on the manuscript, as well as additional members of the Jaenisch, Haggarty and Sur laboratories, for helpful discussion. We also thank Dr Nora Perrone-Bizzozero, Dr Andrea Allan and Dr Meilian Liu, as well as Robert Oliver, Elizabeth Solomon and Xing Zhang, for technical advice and assistance. This manuscript was supported by an NIH grant MH085802 (to MS), 2R01MH104610-15 (to RJ) and the Simons Foundation Autism Research Initiative (to MS, RJ), NIH/NIMH R01MH095088 (to SJH), Pitt-Hopkins Research Foundation (to SJH), FRAXA Research Foundation (to SJH) and Human Frontier Science Program Long-Term Fellowship (to JPKI).

Author information

Authors and Affiliations

Authors

Corresponding authors

Correspondence to N Mellios or M Sur.

Ethics declarations

Competing interests

The authors declare no conflict of interest.

Additional information

Supplementary Information accompanies the paper on the Molecular Psychiatry website

Supplementary information

PowerPoint slides

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Mellios, N., Feldman, D., Sheridan, S. et al. MeCP2-regulated miRNAs control early human neurogenesis through differential effects on ERK and AKT signaling. Mol Psychiatry 23, 1051–1065 (2018). https://doi.org/10.1038/mp.2017.86

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/mp.2017.86

This article is cited by

Search

Quick links