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.

Purkinje cells derived from TSC patients display hypoexcitability and synaptic deficits associated with reduced FMRP levels and reversed by rapamycin

Abstract

Accumulating evidence suggests that cerebellar dysfunction early in life is associated with autism spectrum disorder (ASD), but the molecular mechanisms underlying the cerebellar deficits at the cellular level are unclear. Tuberous sclerosis complex (TSC) is a neurocutaneous disorder that often presents with ASD. Here, we developed a cerebellar Purkinje cell (PC) model of TSC with patient-derived human induced pluripotent stem cells (hiPSCs) to characterize the molecular mechanisms underlying cerebellar abnormalities in ASD and TSC. Our results show that hiPSC-derived PCs from patients with pathogenic TSC2 mutations displayed mTORC1 pathway hyperactivation, defects in neuronal differentiation and RNA regulation, hypoexcitability and reduced synaptic activity when compared with those derived from controls. Our gene expression analyses revealed downregulation of several components of fragile X mental retardation protein (FMRP) targets in TSC2-deficient hiPSC-PCs. We detected decreased expression of FMRP, glutamate receptor δ2 (GRID2), and pre- and post-synaptic markers such as synaptophysin and PSD95 in the TSC2-deficient hiPSC-PCs. The mTOR inhibitor rapamycin rescued the deficits in differentiation, synaptic dysfunction, and hypoexcitability of TSC2 mutant hiPSC-PCs in vitro. Our findings suggest that these gene expression changes and cellular abnormalities contribute to aberrant PC function during development in TSC affected individuals.

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

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

References

  1. Casanova MF. The neuropathology of autism. Brain Pathol. 2007;17:422–33.

    Article  Google Scholar 

  2. Whitney ER, Kemper TL, Bauman ML, Rosene DL, Blatt GJ. Cerebellar Purkinje cells are reduced in a subpopulation of autistic brains: a stereological experiment using calbindin-D28k. Cerebellum. 2008;7:406–16.

    Article  CAS  Google Scholar 

  3. Limperopoulos C, Bassan H, Gauvreau K, Robertson RL Jr., Sullivan NR, Benson CB, et al. Does cerebellar injury in premature infants contribute to the high prevalence of long-term cognitive, learning, and behavioral disability in survivors? Pediatrics. 2007;120:584–93.

    Article  Google Scholar 

  4. Lipton JO, Sahin M. The neurology of mTOR. Neuron. 2014;84:275–91.

    Article  CAS  Google Scholar 

  5. Crino PB, Nathanson KL, Henske EP. The tuberous sclerosis complex. N Engl J Med. 2006;355:1345–56.

    Article  CAS  Google Scholar 

  6. Dibble CC, Elis W, Menon S, Qin W, Klekota J, Asara JM, et al. TBC1D7 is a third subunit of the TSC1-TSC2 complex upstream of mTORC1. Mol Cell. 2012;47:535–46.

    Article  CAS  Google Scholar 

  7. Inoki K, Li Y, Xu T, Guan KL. Rheb GTPase is a direct target of TSC2 GAP activity and regulates mTOR signaling. Genes Dev. 2003;17:1829–34.

    Article  CAS  Google Scholar 

  8. Eluvathingal TJ, Behen ME, Chugani HT, Janisse J, Bernardi B, Chakraborty P, et al. Cerebellar lesions in tuberous sclerosis complex: neurobehavioral and neuroimaging correlates. J Child Neurol. 2006;21:846–51.

    Article  Google Scholar 

  9. Weber AM, Egelhoff JC, McKellop JM, Franz DN. Autism and the cerebellum: evidence from tuberous sclerosis. J Autism Dev Disord. 2000;30:511–7.

    Article  CAS  Google Scholar 

  10. Asano E, Chugani DC, Muzik O, Behen M, Janisse J, Rothermel R, et al. Autism in tuberous sclerosis complex is related to both cortical and subcortical dysfunction. Neurology. 2001;57:1269–77.

    Article  CAS  Google Scholar 

  11. Ertan G, Arulrajah S, Tekes A, Jordan L, Huisman TA. Cerebellar abnormality in children and young adults with tuberous sclerosis complex: MR and diffusion weighted imaging findings. J Neuroradiol. 2010;37:231–8.

    Article  CAS  Google Scholar 

  12. Tsai PT, Hull C, Chu Y, Greene-Colozzi E, Sadowski AR, Leech JM, et al. Autistic-like behaviour and cerebellar dysfunction in Purkinje cell Tsc1 mutant mice. Nature. 2012;488:647–51.

    Article  CAS  Google Scholar 

  13. Reith RM, McKenna J, Wu H, Hashmi SS, Cho SH, Dash PK, et al. Loss of Tsc2 in Purkinje cells is associated with autistic-like behavior in a mouse model of tuberous sclerosis complex. Neurobiol Dis. 2013;51:93–103.

    Article  CAS  Google Scholar 

  14. Schlaeger TM, Daheron L, Brickler TR, Entwisle S, Chan K, Cianci A, et al. A comparison of non-integrating reprogramming methods. Nat Biotechnol. 2015;33:58–63.

    Article  CAS  Google Scholar 

  15. Takahashi K, Tanabe K, Ohnuki M, Narita M, Ichisaka T, Tomoda K, et al. Induction of pluripotent stem cells from adult human fibroblasts by defined factors. Cell. 2007;131:861–72.

    Article  CAS  Google Scholar 

  16. Mussolino C, Morbitzer R, Lutge F, Dannemann N, Lahaye T, Cathomen T. A novel TALE nuclease scaffold enables high genome editing activity in combination with low toxicity. Nucleic Acids Res. 2011;39:9283–93.

    Article  CAS  Google Scholar 

  17. Horii T, Tamura D, Morita S, Kimura M, Hatada I. Generation of an ICF syndrome model by efficient genome editing of human induced pluripotent stem cells using the CRISPR system. Int J Mol Sci. 2013;14:19774–81.

    Article  Google Scholar 

  18. Hou Z, Zhang Y, Propson NE, Howden SE, Chu LF, Sontheimer EJ, et al. Efficient genome engineering in human pluripotent stem cells using Cas9 from Neisseria meningitidis. Proc Natl Acad Sci USA. 2013;110:15644–9.

    Article  CAS  Google Scholar 

  19. Guo Q, Li K, Sunmonu NA, Li JY. Fgf8b-containing spliceforms, but not Fgf8a, are essential for Fgf8 function during development of the midbrain and cerebellum. Dev Biol. 2010;338:183–92.

    Article  CAS  Google Scholar 

  20. Selvadurai HJ, Mason JO. Wnt/beta-catenin signalling is active in a highly dynamic pattern during development of the mouse cerebellum. PLoS ONE. 2011;6:e23012.

    Article  CAS  Google Scholar 

  21. Sundberg M, Bogetofte H, Lawson T, Jansson J, Smith G, Astradsson A, et al. Improved cell therapy protocols for Parkinson’s disease based on differentiation efficiency and safety of hESC-, hiPSC-, and non-human primate iPSC-derived dopaminergic neurons. Stem Cells. 2013;31:1548–62.

    Article  CAS  Google Scholar 

  22. Buchholz DE, Pennington BO, Croze RH, Hinman CR, Coffey PJ, Clegg DO. Rapid and efficient directed differentiation of human pluripotent stem cells into retinal pigmented epithelium. Stem Cells Transl Med. 2013;2:384–93.

    Article  CAS  Google Scholar 

  23. Chambers SM, Fasano CA, Papapetrou EP, Tomishima M, Sadelain M, Studer L. Highly efficient neural conversion of human ES and iPS cells by dual inhibition of SMAD signaling. Nat Biotechnol. 2009;27:275–80.

    Article  CAS  Google Scholar 

  24. Muguruma K, Nishiyama A, Kawakami H, Hashimoto K, Sasai Y. Self-organization of polarized cerebellar tissue in 3D culture of human pluripotent stem cells. Cell Rep. 2015;10:537–50.

    Article  CAS  Google Scholar 

  25. Heuer H, Mason CA. Thyroid hormone induces cerebellar Purkinje cell dendritic development via the thyroid hormone receptor alpha1. J Neurosci. 2003;23:10604–12.

    Article  CAS  Google Scholar 

  26. Wang S, Wang B, Pan N, Fu L, Wang C, Song G, et al. Differentiation of human induced pluripotent stem cells to mature functional Purkinje neurons. Sci Rep. 2015;5:9232.

    Article  CAS  Google Scholar 

  27. Crino PB, Aronica E, Baltuch G, Nathanson KL. Biallelic TSC gene inactivation in tuberous sclerosis complex. Neurology. 2010;74:1716–23.

    Article  Google Scholar 

  28. Costa V, Aigner S, Vukcevic M, Sauter E, Behr K, Ebeling M, et al. mTORC1 inhibition corrects neurodevelopmental and synaptic alterations in a human stem cell model of tuberous sclerosis. Cell Rep. 2016;15:86–95.

    Article  CAS  Google Scholar 

  29. Huang J, Manning BD. The TSC1-TSC2 complex: a molecular switchboard controlling cell growth. Biochem J. 2008;412:179–90.

    Article  CAS  Google Scholar 

  30. Brennand KJ, Simone A, Jou J, Gelboin-Burkhart C, Tran N, Sangar S, et al. Modelling schizophrenia using human induced pluripotent stem cells. Nature. 2011;473:221–5.

    Article  CAS  Google Scholar 

  31. Grabole N, Zhang JD, Aigner S, Ruderisch N, Costa V, Weber FC, et al. Genomic analysis of the molecular neuropathology of tuberous sclerosis using a human stem cell model. Genome Med. 2016;8:94.

    Article  Google Scholar 

  32. Ascano M Jr, Mukherjee N, Bandaru P, Miller JB, Nusbaum JD, Corcoran DL, et al. FMRP targets distinct mRNA sequence elements to regulate protein expression. Nature. 2012;492:382–6.

    Article  CAS  Google Scholar 

  33. Darnell JC, Van Driesche SJ, Zhang C, Hung KY, Mele A, Fraser CE, et al. FMRP stalls ribosomal translocation on mRNAs linked to synaptic function and autism. Cell. 2011;146:247–61.

    Article  CAS  Google Scholar 

  34. Cloetta D, Thomanetz V, Baranek C, Lustenberger RM, Lin S, Oliveri F, et al. Inactivation of mTORC1 in the developing brain causes microcephaly and affects gliogenesis. J Neurosci. 2013;33:7799–810.

    Article  CAS  Google Scholar 

  35. Hagan N, Guarente J, Ellisor D, Zervas M. The temporal contribution of the Gbx2 lineage to cerebellar neurons. Front Neuroanat. 2017;11:50.

    Article  Google Scholar 

  36. Kassai H, Sugaya Y, Noda S, Nakao K, Maeda T, Kano M, et al. Selective activation of mTORC1 signaling recapitulates microcephaly, tuberous sclerosis, and neurodegenerative diseases. Cell Rep. 2014;7:1626–39.

    Article  CAS  Google Scholar 

  37. Magri L, Cambiaghi M, Cominelli M, Alfaro-Cervello C, Cursi M, Pala M, et al. Sustained activation of mTOR pathway in embryonic neural stem cells leads to development of tuberous sclerosis complex-associated lesions. Cell Stem Cell. 2011;9:447–62.

    Article  CAS  Google Scholar 

  38. Normand EA, Crandall SR, Thorn CA, Murphy EM, Voelcker B, Browning C, et al. Temporal and mosaic Tsc1 deletion in the developing thalamus disrupts thalamocortical circuitry, neural function, and behavior. Neuron. 2013;78:895–909.

    Article  CAS  Google Scholar 

  39. Grajkowska W, Kotulska K, Jurkiewicz E, Matyja E. Brain lesions in tuberous sclerosis complex. Review. Folia Neuropathol. 2010;48:139–49.

    PubMed  Google Scholar 

  40. Jay V, Edwards V, Musharbash A, Rutka JT. Cerebellar pathology in tuberous sclerosis. Ultrastruct Pathol. 1998;22:331–9.

    Article  CAS  Google Scholar 

  41. Way SW, McKenna J 3rd, Mietzsch U, Reith RM, Wu HC, Gambello MJ. Loss of Tsc2 in radial glia models the brain pathology of tuberous sclerosis complex in the mouse. Hum Mol Genet. 2009;18:1252–65.

    Article  CAS  Google Scholar 

  42. Mietzsch U, McKenna J 3rd, Reith RM, Way SW, Gambello MJ. Comparative analysis of Tsc1 and Tsc2 single and double radial glial cell mutants. J Comp Neurol. 2013;521:3817–31.

    Article  CAS  Google Scholar 

  43. Carson RP, Kelm ND, West KL, Does MD, Fu C, Weaver G, et al. Hypomyelination following deletion of Tsc2 in oligodendrocyte precursors. Ann Clin Transl Neurol. 2015;2:1041–54.

    Article  CAS  Google Scholar 

  44. Lebrun-Julien F, Bachmann L, Norrmen C, Trotzmuller M, Kofeler H, Ruegg MA, et al. Balanced mTORC1 activity in oligodendrocytes is required for accurate CNS myelination. J Neurosci. 2014;34:8432–48.

    Article  Google Scholar 

  45. Ercan E, Han JM, Di Nardo A, Winden K, Han MJ, Hoyo L, et al. Neuronal CTGF/CCN2 negatively regulates myelination in a mouse model of tuberous sclerosis complex. J Exp Med. 2017;214:681–97.

    CAS  PubMed  PubMed Central  Google Scholar 

  46. Meikle L, Talos DM, Onda H, Pollizzi K, Rotenberg A, Sahin M, et al. A mouse model of tuberous sclerosis: neuronal loss of Tsc1 causes dysplastic and ectopic neurons, reduced myelination, seizure activity, and limited survival. J Neurosci. 2007;27:5546–58.

    Article  CAS  Google Scholar 

  47. Makki MI, Chugani DC, Janisse J, Chugani HT. Characteristics of abnormal diffusivity in normal-appearing white matter investigated with diffusion tensor MR imaging in tuberous sclerosis complex. Am J Neuroradiol. 2007;28:1662–7.

    Article  CAS  Google Scholar 

  48. Arulrajah S, Ertan G, Jordan L, Tekes A, Khaykin E, Izbudak I, et al. Magnetic resonance imaging and diffusion-weighted imaging of normal-appearing white matter in children and young adults with tuberous sclerosis complex. Neuroradiology. 2009;51:781–6.

    Article  Google Scholar 

  49. Peters JM, Sahin M, Vogel-Farley VK, Jeste SS, Nelson CA 3rd, Gregas MC, et al. Loss of white matter microstructural integrity is associated with adverse neurological outcome in tuberous sclerosis complex. Acad Radiol. 2012;19:17–25.

    Article  Google Scholar 

  50. Douvaras P, Wang J, Zimmer M, Hanchuk S, O’Bara MA, Sadiq S, et al. Efficient generation of myelinating oligodendrocytes from primary progressive multiple sclerosis patients by induced pluripotent stem cells. Stem Cell Rep. 2014;3:250–9.

    Article  CAS  Google Scholar 

  51. Wang S, Bates J, Li X, Schanz S, Chandler-Militello D, Levine C, et al. Human iPSC-derived oligodendrocyte progenitor cells can myelinate and rescue a mouse model of congenital hypomyelination. Cell Stem Cell. 2013;12:252–64.

    Article  CAS  Google Scholar 

  52. Stacpoole SR, Spitzer S, Bilican B, Compston A, Karadottir R, Chandran S, et al. High yields of oligodendrocyte lineage cells from human embryonic stem cells at physiological oxygen tensions for evaluation of translational biology. Stem Cell Rep. 2013;1:437–50.

    Article  CAS  Google Scholar 

  53. Sundberg M, Hyysalo A, Skottman H, Shin S, Vemuri M, Suuronen R, et al. A xeno-free culturing protocol for pluripotent stem cell-derived oligodendrocyte precursor cell production. Regen Med. 2011;6:449–60.

    Article  CAS  Google Scholar 

  54. Nistor GI, Totoiu MO, Haque N, Carpenter MK, Keirstead HS. Human embryonic stem cells differentiate into oligodendrocytes in high purity and myelinate after spinal cord transplantation. Glia. 2005;49:385–96.

    Article  Google Scholar 

  55. Bauman ML, Kemper TL. Neuroanatomic observations of the brain in autism: a review and future directions. Int J Dev Neurosci. 2005;23:183–7.

    Article  Google Scholar 

  56. Bailey A, Luthert P, Dean A, Harding B, Janota I, Montgomery M, et al. A clinicopathological study of autism. Brain. 1998;121:889–905.

    Article  Google Scholar 

  57. Vargas DL, Nascimbene C, Krishnan C, Zimmerman AW, Pardo CA. Neuroglial activation and neuroinflammation in the brain of patients with autism. Ann Neurol. 2005;57:67–81.

    Article  CAS  Google Scholar 

  58. Weisenfeld NI, Peters JM, Tsai PT, Prabhu SP, Dies KA, Sahin M, et al. A magnetic resonance imaging study of cerebellar volume in tuberous sclerosis complex. Pediatr Neurol. 2013;48:105–10.

    Article  Google Scholar 

  59. Boer K, Troost D, Jansen F, Nellist M, van den Ouweland AM, Geurts JJ, et al. Clinicopathological and immunohistochemical findings in an autopsy case of tuberous sclerosis complex. Neuropathology. 2008;28:577–90.

    PubMed  Google Scholar 

  60. Crino PB. Molecular pathogenesis of tuber formation in tuberous sclerosis complex. J Child Neurol. 2004;19:716–25.

    Article  Google Scholar 

  61. Miyata H, Chiang AC, Vinters HV. Insulin signaling pathways in cortical dysplasia and TSC-tubers: tissue microarray analysis. Ann Neurol. 2004;56:510–9.

    Article  CAS  Google Scholar 

  62. Tavazoie SF, Alvarez VA, Ridenour DA, Kwiatkowski DJ, Sabatini BL. Regulation of neuronal morphology and function by the tumor suppressors Tsc1 and Tsc2. Nat Neurosci. 2005;8:1727–34.

    Article  CAS  Google Scholar 

  63. Uhlmann EJ, Wong M, Baldwin RL, Bajenaru ML, Onda H, Kwiatkowski DJ, et al. Astrocyte-specific TSC1 conditional knockout mice exhibit abnormal neuronal organization and seizures. Ann Neurol. 2002;52:285–96.

    Article  CAS  Google Scholar 

  64. Jaworski J, Spangler S, Seeburg DP, Hoogenraad CC, Sheng M. Control of dendritic arborization by the phosphoinositide-3′-kinase-Akt-mammalian target of rapamycin pathway. J Neurosci. 2005;25:11300–12.

    Article  CAS  Google Scholar 

  65. Kumar V, Zhang MX, Swank MW, Kunz J, Wu GY. Regulation of dendritic morphogenesis by Ras-PI3K-Akt-mTOR and Ras-MAPK signaling pathways. J Neurosci. 2005;25:11288–99.

    Article  CAS  Google Scholar 

  66. Mirzaa GM, Parry DA, Fry AE, Giamanco KA, Schwartzentruber J, Vanstone M, et al. De novo CCND2 mutations leading to stabilization of cyclin D2 cause megalencephaly-polymicrogyria-polydactyly-hydrocephalus syndrome. Nat Genet. 2014;46:510–5.

    Article  CAS  Google Scholar 

  67. Mirzaa G, Dodge NN, Glass I, Day C, Gripp K, Nicholson L, et al. Megalencephaly and perisylvian polymicrogyria with postaxial polydactyly and hydrocephalus: a rare brain malformation syndrome associated with mental retardation and seizures. Neuropediatrics. 2004;35:353–9.

    Article  CAS  Google Scholar 

  68. Di Nardo A, Wertz MH, Kwiatkowski E, Tsai PT, Leech JD, Greene-Colozzi E, et al. Neuronal Tsc1/2 complex controls autophagy through AMPK-dependent regulation of ULK1. Hum Mol Genet. 2014;23:3865–74.

    Article  Google Scholar 

  69. Ebrahimi-Fakhari D, Saffari A, Wahlster L, Di Nardo A, Turner D, Lewis TL Jr, et al. Impaired mitochondrial dynamics and mitophagy in neuronal models of tuberous sclerosis complex. Cell Rep. 2016;17:2162.

    Article  CAS  Google Scholar 

  70. Adusei DC, Pacey LK, Chen D, Hampson DR. Early developmental alterations in GABAergic protein expression in fragile X knockout mice. Neuropharmacology. 2010;59:167–71.

    Article  CAS  Google Scholar 

  71. Curia G, Papouin T, Seguela P, Avoli M. Downregulation of tonic GABAergic inhibition in a mouse model of fragile X syndrome. Cereb Cortex. 2009;19:1515–20.

    Article  Google Scholar 

  72. Sabanov V, Braat S, D’Andrea L, Willemsen R, Zeidler S, Rooms L, et al. Impaired GABAergic inhibition in the hippocampus of Fmr1 knockout mice. Neuropharmacology. 2017;116:71–81.

    Article  CAS  Google Scholar 

  73. Narayanan U, Nalavadi V, Nakamoto M, Thomas G, Ceman S, Bassell GJ, et al. S6K1 phosphorylates and regulates fragile X mental retardation protein (FMRP) with the neuronal protein synthesis-dependent mammalian target of rapamycin (mTOR) signaling cascade. J Biol Chem. 2008;283:18478–82.

    Article  CAS  Google Scholar 

  74. Sarbassov DD, Ali SM, Sengupta S, Sheen JH, Hsu PP, Bagley AF, et al. Prolonged rapamycin treatment inhibits mTORC2 assembly and Akt/PKB. Mol Cell. 2006;22:159–68.

    Article  CAS  Google Scholar 

  75. Sundberg M, Sahin M. Cerebellar development and autism spectrum disorder in tuberous sclerosis complex. J Child Neurol. 2015;30:1954–62.

    Article  Google Scholar 

Download references

Acknowledgements

We thank the individuals affected by TSC who took part in this study. We are grateful to Dr. Kleiman, Dr. Chen, and Dr. Buttermore for helpful discussion, and Kerem Muslu for assistance in qRT-PCR analyses. We would like to thank research associate Christopher Stoddard (UConn Health, Human Genome Editing Core, US) for assistance with CRISPR-gene modification of hiPSCs. The hiPSC line F628 was provided by Dr. George Daley (Boston Children’s Hospital). We thank BCH IDDRC Cellular Imaging Core for help with confocal imaging (U54 HD090255). This study was funded by U.S. Army Medical Research Tuberous Sclerosis Complex Research Program (W81XWH-15-1-0189), Nancy Lurie Marks Family Foundation, Harvard Stem Cell Institute, and the Children’s Hospital Boston Translational Research Program (to MS). KW is supported by R25 NS07068207S1. The project was co-sponsored by the Iris and Jumming Le Foundation and the Rockefeller University Center for Clinical and Translational Science #UL1 TR000043 (NCATS, NIH), CTSA and NIH R21 NS093540-01 (to MEH).

Author contributions

MS: PC differentiation protocol development for hiPSC, experimental design, PC differentiation, phenotyping in vitro, FC, transcriptional profiling, data analyses, writing of manuscript. IT: electrophysiology, data analyses, writing of manuscript. DEB: PC differentiation protocol development for hESC. KW: transcriptional expression analyses, writing of manuscript. VK: confocal microscopy. KK: statistical analyses. DC: cell analyses. DT: technical assistance. M-JH: hiPSC line derivation, CJW and MEH: discussion of data. MS and MEH: experimental design, writing of manuscript.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Mustafa Sahin.

Ethics declarations

Conflict of interest

The authors declare that they have no conflict of interest.

Electronic supplementary material

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Sundberg, M., Tochitsky, I., Buchholz, D.E. et al. Purkinje cells derived from TSC patients display hypoexcitability and synaptic deficits associated with reduced FMRP levels and reversed by rapamycin. Mol Psychiatry 23, 2167–2183 (2018). https://doi.org/10.1038/s41380-018-0018-4

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41380-018-0018-4

This article is cited by

Search

Quick links