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.

  • Article
  • Published:

Mispatterning and interneuron deficit in Tourette Syndrome basal ganglia organoids

Abstract

Tourette Syndrome (TS) is a neuropsychiatric disorder thought to involve a reduction of basal ganglia (BG) interneurons and malfunctioning of the BG circuitry. However, whether interneurons fail to develop or are lost postnatally remains unknown. To investigate the pathophysiology of early development in TS, induced pluripotent stem cell (iPSC)-derived BG organoids from TS patients and healthy controls were compared on multiple levels of measurement and analysis. BG organoids from TS individuals manifested an impaired medial ganglionic eminence fate and a decreased differentiation of cholinergic and GABAergic interneurons. Transcriptome analyses revealed organoid mispatterning in TS, with a preference for dorsolateral at the expense of ventromedial fates. Our results point to altered expression of GLI transcription factors downstream of the Sonic Hedgehog signaling pathway with cilia disruption at the earliest stages of BG organoid differentiation as a potential mechanism for the BG mispatterning in TS. This study uncovers early neurodevelopmental underpinnings of TS neuropathological deficits using organoids as a model system.

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

Access options

Buy this article

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

Fig. 1: Characterization of ventral telencephalic or basal ganglia (BG) organoids in control (CT) iPSC lines.
Fig. 2: Decreased ventral and increased dorsal patterning in TS BG organoids at TD14.
Fig. 3: Bulk RNA sequencing at TD30 show consistent disruption in ventral telencephalic development in TS.
Fig. 4: Upregulation of GLI genes and repressor proteins at TD0 in TS-derived BG organoids.
Fig. 5: Development of cilia in TS.

Similar content being viewed by others

References

  1. Eblen F, Graybiel AM. Highly restricted origin of prefrontal cortical inputs to striosomes in the macaque monkey. J Neurosci. 1995;15:5999–6013.

    CAS  PubMed  PubMed Central  Google Scholar 

  2. Wang Z, Maia TV, Marsh R, Colibazzi T, Gerber A, Peterson BS. The neural circuits that generate tics in Tourette’s syndrome. Am J Psychiatry. 2011;168:1326–37.

    PubMed  PubMed Central  Google Scholar 

  3. Albin RL, Young AB, Penney JB. The functional anatomy of basal ganglia disorders. TINS. 1995;18:63–4.

    CAS  PubMed  Google Scholar 

  4. Bloch MH, Leckman JF, Zhu H, Peterson BS. Caudate volumes in childhood predict symptom severity in adults with Tourette syndrome. Neurology. 2005;65:1253–8.

    PubMed  Google Scholar 

  5. Peterson BS, Thomas P, Kane MJ, Scahill L, Zhang H, Bronen R, et al. Basal Ganglia volumes in patients with Gilles de la Tourette syndrome. Arch Gen Psychiatry. 2003;60:415–24.

    PubMed  Google Scholar 

  6. Kalanithi PS, Zheng W, Kataoka Y, DiFiglia M, Grantz H, Saper CB, et al. Altered parvalbumin-positive neuron distribution in basal ganglia of individuals with Tourette syndrome. Proc Natl Acad Sci USA. 2005;102:13307–12.

    CAS  PubMed  PubMed Central  Google Scholar 

  7. Kataoka Y, Kalanithi PS, Grantz H, Schwartz ML, Saper C, Leckman JF, et al. Decreased number of parvalbumin and cholinergic interneurons in the striatum of individuals with Tourette Syndrome. J Comp Neurol. 2010;518:277–91.

    PubMed  PubMed Central  Google Scholar 

  8. Lennington JB, Coppola G, Kataoka-Sasaki Y, Fernandez TV, Palejev D, Li Y, et al. Transcriptome analysis of the human striatum in Tourette Syndrome. Biol Psychiatry. 2016;79:372–82.

    CAS  PubMed  Google Scholar 

  9. Kawaguchi Y. Physiological, morphological and histochemical characterization of three classess of interneurons in rat neostriatum. JNeurosci. 1993;13:4908–23.

    CAS  Google Scholar 

  10. Mariani J, Simonini MV, Palejev D, Tomasini L, Coppola G, Szekely AM, et al. Modeling human cortical development in vitro using induced pluripotent stem cells. Proc Natl Acad Sci USA. 2012;109:12770–5.

    CAS  PubMed  PubMed Central  Google Scholar 

  11. Camp JG, Badsha F, Florio M, Kanton S, Gerber T, Wilsch-Brauninger M, et al. Human cerebral organoids recapitulate gene expression programs of fetal neocortex development. Proc Natl Acad Sci USA. 2015;112:15672–7.

    CAS  PubMed  PubMed Central  Google Scholar 

  12. Lancaster MA, Renner M, Martin CA, Wenzel D, Bicknell LS, Hurles ME, et al. Cerebral organoids model human brain development and microcephaly. Nature. 2013;501:373–9.

    CAS  PubMed  Google Scholar 

  13. Quadrato G, Nguyen T, Macosko EZ, Sherwood JL, Min Yang S, Berger DR, et al. Cell diversity and network dynamics in photosensitive human brain organoids. Nature. 2017;545:48–53.

    CAS  PubMed  PubMed Central  Google Scholar 

  14. Marton RM, Pasca SP. Organoid and assembloid technologies for investigating cellular crosstalk in human brain development and disease. Trends Cell Biol. 2020;30:133–43.

    CAS  PubMed  Google Scholar 

  15. Lim L, Mi D, Llorca A, Marin O. Development and functional diversification of cortical interneurons. Neuron. 2018;100:294–313.

    CAS  PubMed  PubMed Central  Google Scholar 

  16. Hoch RV, Clarke JA, Rubenstein JL. Fgf signaling controls the telencephalic distribution of Fgf-expressing progenitors generated in the rostral patterning center. Neural Dev. 2015;10:8.

    PubMed  PubMed Central  Google Scholar 

  17. Bloch MH, Peterson BS, Scahill L, Otka J, Katsovich L, Zhang H, et al. Adulthood outcome of tic and obsessive-compulsive symptom severity in children with Tourette syndrome. Arch Pediatr Adolesc Med. 2006;160:65–9.

    PubMed  PubMed Central  Google Scholar 

  18. Park IH, Lerou PH, Zhao R, Huo H, Daley GQ. Generation of human-induced pluripotent stem cells. Nat Protoc. 2008;3:1180–6.

    CAS  PubMed  Google Scholar 

  19. Okita K, Matsumura Y, Sato Y, Okada A, Morizane A, Okamoto S, et al. A more efficient method to generate integration-free human iPS cells. Nat Methods. 2011;8:409–12.

    CAS  PubMed  Google Scholar 

  20. 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;162:375–90.

    CAS  PubMed  PubMed Central  Google Scholar 

  21. Lopez-Coviella I, Berse B, Krauss R, Thies RS, Blusztajn JK. Induction and maintenance of the neuronal cholinergic phenotype in the central nervous system by BMP-9. Science. 2000;289:313–6.

    CAS  PubMed  Google Scholar 

  22. Lopez-Coviella I, Follettie MT, Mellott TJ, Kovacheva VP, Slack BE, Diesl V, et al. Bone morphogenetic protein 9 induces the transcriptome of basal forebrain cholinergic neurons. Proc Natl Acad Sci USA. 2005;102:6984–9.

    CAS  PubMed  PubMed Central  Google Scholar 

  23. Dobin A, Davis CA, Schlesinger F, Drenkow J, Zaleski C, Jha S, et al. STAR: ultrafast universal RNA-seq aligner. Bioinformatics. 2013;29:15–21.

    CAS  PubMed  Google Scholar 

  24. Li H, Handsaker B, Wysoker A, Fennell T, Ruan J, Homer N, et al. The sequence alignment/map format and SAMtools. Bioinformatics. 2009;25:2078–9.

    PubMed  PubMed Central  Google Scholar 

  25. Liao Y, Smyth GK, Shi W. featureCounts: an efficient general purpose program for assigning sequence reads to genomic features. Bioinformatics. 2014;30:923–30.

    CAS  PubMed  Google Scholar 

  26. Robinson MD, McCarthy DJ, Smyth GK. edgeR: a Bioconductor package for differential expression analysis of digital gene expression data. Bioinformatics. 2010;26:139–40.

    CAS  PubMed  Google Scholar 

  27. Kamburov A, Pentchev K, Galicka H, Wierling C, Lehrach H, Herwig R. ConsensusPathDB: toward a more complete picture of cell biology. Nucleic Acids Res. 2011;39:D712–7.

    CAS  PubMed  Google Scholar 

  28. Li M, Santpere G, Imamura Kawasawa Y, Evgrafov OV, Gulden FO, Pochareddy S, et al. Integrative functional genomic analysis of human brain development and neuropsychiatric risks. Science. 2018;362.

  29. Shi Y, Wang M, Mi D, Lu T, Wang B, Dong H, et al. Mouse and human share conserved transcriptional programs for interneuron development. Science. 2021;374:eabj6641.

    CAS  PubMed  Google Scholar 

  30. Nobrega-Pereira S, Kessaris N, Du T, Kimura S, Anderson SA, Marin O. Postmitotic Nkx2-1 controls the migration of telencephalic interneurons by direct repression of guidance receptors. Neuron. 2008;59:733–45.

    CAS  PubMed  PubMed Central  Google Scholar 

  31. Sussel L, Marin O, Kimura S, Rubenstein JL. Loss of Nkx2.1 homeobox gene function results in a ventral to dorsal molecular respecification within the basal telencephalon: evidence for a transformation of the pallidum into the striatum. Development. 1999;126:3359–70.

    CAS  PubMed  Google Scholar 

  32. Corbin JG, Rutlin M, Gaiano N, Fishell G. Combinatorial function of the homeodomain proteins Nkx2.1 and Gsh2 in ventral telencephalic patterning. Development. 2003;130:4895–906.

    CAS  PubMed  Google Scholar 

  33. Magno L, Barry C, Schmidt-Hieber C, Theodotou P, Hausser M, Kessaris N. NKX2-1 is required in the embryonic septum for cholinergic system development, learning, and memory. Cell Rep. 2017;20:1572–84.

    CAS  PubMed  PubMed Central  Google Scholar 

  34. Silberberg SN, Taher L, Lindtner S, Sandberg M, Nord AS, Vogt D, et al. Subpallial enhancer transgenic lines: a data and tool resource to study transcriptional regulation of GABAergic cell fate. Neuron. 2016;92:59–74.

    CAS  PubMed  PubMed Central  Google Scholar 

  35. Luccardini C, Hennekinne L, Viou L, Yanagida M, Murakami F, Kessaris N, et al. N-cadherin sustains motility and polarity of future cortical interneurons during tangential migration. J Neurosci. 2013;33:18149–60.

    CAS  PubMed  PubMed Central  Google Scholar 

  36. Rataj-Baniowska M, Niewiadomska-Cimicka A, Paschaki M, Szyszka-Niagolov M, Carramolino L, Torres M, et al. Retinoic acid receptor beta controls development of striatonigral projection neurons through FGF-dependent and Meis1-dependent mechanisms. J Neurosci. 2015;35:14467–75.

    CAS  PubMed  PubMed Central  Google Scholar 

  37. Long JE, Cobos I, Potter GB, Rubenstein JL. Dlx1&2 and Mash1 transcription factors control MGE and CGE patterning and differentiation through parallel and overlapping pathways. Cereb Cortex. 2009;19:i96–106.

    PubMed  PubMed Central  Google Scholar 

  38. Sandberg M, Flandin P, Silberberg S, Su-Feher L, Price JD, Hu JS, et al. Transcriptional networks controlled by NKX2-1 in the development of Forebrain GABAergic neurons. Neuron 2016;91:1260–75.

    CAS  PubMed  PubMed Central  Google Scholar 

  39. Scott BB, Lois C. Generation of tissue-specific transgenic birds with lentiviral vectors. Proc Natl Acad Sci USA. 2005;102:16443–7.

    CAS  PubMed  PubMed Central  Google Scholar 

  40. Bassett AS, Scherer SW. Copy number variation in Tourette Syndrome. Neuron 2017;94:1041–3.

    CAS  PubMed  Google Scholar 

  41. Huang AY, Yu D, Davis LK, Sul JH, Tsetsos F, Ramensky V, et al. Rare copy number variants in NRXN1 and CNTN6 increase risk for Tourette Syndrome. Neuron. 2017;94:1101–11 e7.

    CAS  PubMed  PubMed Central  Google Scholar 

  42. Qin S, Madhavan M, Waclaw RR, Nakafuku M, Campbell K. Characterization of a new Gsx2-cre line in the developing mouse telencephalon. Genesis. 2016;54:542–9.

    CAS  PubMed  PubMed Central  Google Scholar 

  43. Yun K, Potter S, Rubenstein JL. Gsh2 and Pax6 play complementary roles in dorsoventral patterning of the mammalian telencephalon. Development. 2001;128:193–205.

    CAS  PubMed  Google Scholar 

  44. Xu Q, Tam M, Anderson SA. Fate mapping Nkx2.1-lineage cells in the mouse telencephalon. J Comp Neurol. 2008;506:16–29.

    CAS  PubMed  Google Scholar 

  45. Manabe T, Tatsumi K, Inoue M, Makinodan M, Yamauchi T, Makinodan E, et al. L3/Lhx8 is a pivotal factor for cholinergic differentiation of murine embryonic stem cells. Cell Death Differ. 2007;14:1080–5.

    CAS  PubMed  Google Scholar 

  46. Zhao Y, Marin O, Hermesz E, Powell A, Flames N, Palkovits M, et al. The LIM-homeobox gene Lhx8 is required for the development of many cholinergic neurons in the mouse forebrain. Proc Natl Acad Sci USA. 2003;100:9005–10.

    CAS  PubMed  PubMed Central  Google Scholar 

  47. Murdoch JN, Copp AJ. The relationship between sonic Hedgehog signaling, cilia, and neural tube defects. Birth Defects Res A Clin Mol Teratol. 2010;88:633–52.

    CAS  PubMed  PubMed Central  Google Scholar 

  48. Andreu-Cervera A, Anselme I, Karam A, Laclef C, Catala M, Schneider-Maunoury S. The ciliopathy gene Ftm/Rpgrip1l controls mouse forebrain patterning via region-specific modulation of Hedgehog/Gli signaling. J Neurosci. 2019;39:2398–415.

    PubMed  PubMed Central  Google Scholar 

  49. Andreu-Cervera A, Catala M, Schneider-Maunoury S. Cilia, ciliopathies and hedgehog-related forebrain developmental disorders. Neurobiol Dis. 2021;150:105236.

    CAS  PubMed  Google Scholar 

  50. Park SM, Jang HJ, Lee JH. Roles of primary cilia in the developing brain. Front Cell Neurosci. 2019;13:218.

    CAS  PubMed  PubMed Central  Google Scholar 

  51. Backman M, Machon O, Mygland L, van den Bout CJ, Zhong W, Taketo MM, et al. Effects of canonical Wnt signaling on dorso-ventral specification of the mouse telencephalon. Dev Biol. 2005;279:155–68.

    CAS  PubMed  Google Scholar 

  52. Arai Y, Cwetsch AW, Coppola E, Cipriani S, Nishihara H, Kanki H, et al. Evolutionary gain of Dbx1 expression drives subplate identity in the cerebral cortex. Cell Rep. 2019;29:645–58 e5.

    CAS  PubMed  Google Scholar 

  53. Thomsen MS, Routhe LJ, Moos T. The vascular basement membrane in the healthy and pathological brain. J Cereb Blood Flow Metab. 2017;37:3300–17.

    PubMed  PubMed Central  Google Scholar 

  54. Hartwig C, Veske A, Krejcova S, Rosenberger G, Finckh U. Plexin B3 promotes neurite outgrowth, interacts homophilically, and interacts with Rin. BMC Neurosci. 2005;6:53.

    PubMed  PubMed Central  Google Scholar 

  55. Zhu B, Chen C, Xue G, Moyzis RK, Dong Q, Chen C. et al. The SEMA5A gene is associated with hippocampal volume, and their interaction is associated with performance on Raven’s Progressive Matrices. NeuroImage. 2014;88:181–7.

    CAS  PubMed  Google Scholar 

  56. Bloch MH, Leckman JF. Clinical course of Tourette Syndrome. J Psychosom Res. 2009;67:497–501.

    PubMed  PubMed Central  Google Scholar 

  57. Leckman JF, Peterson B. The pathogenesis of Tourette’s syndrome: role od epigenetic factors active in early CNS development. Biol Psychiatry. 1993;34:425–7.

    CAS  PubMed  Google Scholar 

  58. Peterson B, Riddle MA, Cohen DJ, Katz LD, Smith JC, Hardin MT, et al. Reduced basal ganglia volumes in Tourette’s syndrome using three-dimensional reconstruction techniques from magnetic resonance images. Neurology. 1993;43:941–9.

    CAS  PubMed  Google Scholar 

  59. Heinz A, Knable MB, Wolf SS, Jones DW, Gorey JG, Hyde TM. et al. Tourette’s syndrome: [I-123]beta-CIT SPECT correlates of vocal tic severity. Neurology. 1998;51:1069–74.

    CAS  PubMed  Google Scholar 

  60. Muller-Vahl KR, Meyer GJ, Knapp WH, Emrich HM, Gielow P, Brucke T, et al. Serotonin transporter binding in Tourette Syndrome. Neurosci Lett. 2005;385:120–5.

    PubMed  Google Scholar 

  61. Wong DF, Brasic JR, Singer HS, Schretlen DJ, Kuwabara H, Zhou Y, et al. Mechanisms of dopaminergic and serotonergic neurotransmission in Tourette Syndrome: clues from an in vivo neurochemistry study with PET. Neuropsychopharmacology. 2008;33:1239–51.

    CAS  PubMed  Google Scholar 

  62. Muller-Vahl KR, Szejko N, Wilke F, Jakubovski E, Geworski L, Bengel F, et al. Serotonin transporter binding is increased in Tourette Syndrome with obsessive compulsive disorder. Sci Rep. 2019;9:972.

    CAS  PubMed  PubMed Central  Google Scholar 

  63. Rallu M, Corbin JG, Fishell G. Parsing the prosencephalon. Nat Rev Neurosci. 2002;3:943–51.

    CAS  PubMed  Google Scholar 

  64. Wen X, Lai CK, Evangelista M, Hongo JA, de Sauvage FJ, Scales SJ. Kinetics of hedgehog-dependent full-length Gli3 accumulation in primary cilia and subsequent degradation. Mol Cell Biol. 2010;30:1910–22.

    CAS  PubMed  PubMed Central  Google Scholar 

  65. Haycraft CJ, Banizs B, Aydin-Son Y, Zhang Q, Michaud EJ, Yoder BK. Gli2 and Gli3 localize to cilia and require the intraflagellar transport protein polaris for processing and function. PLoS Genet. 2005;1:e53.

    PubMed  PubMed Central  Google Scholar 

  66. Kim J, Kato M, Beachy PA. Gli2 trafficking links Hedgehog-dependent activation of Smoothened in the primary cilium to transcriptional activation in the nucleus. Proc Natl Acad Sci USA. 2009;106:21666–71.

    CAS  PubMed  PubMed Central  Google Scholar 

  67. Li J, Wang C, Wu C, Cao T, Xu G, Meng Q, et al. PKA-mediated Gli2 and Gli3 phosphorylation is inhibited by Hedgehog signaling in cilia and reduced in Talpid3 mutant. Dev Biol. 2017;429:147–57.

    CAS  PubMed  PubMed Central  Google Scholar 

  68. Lee B, Panda S, Lee HY. Primary ciliary deficits in the dentate gyrus of fragile X syndrome. Stem Cell Rep. 2020;15:454–66.

    CAS  Google Scholar 

Download references

Acknowledgements

We wish to thank the participants who donated samples and time to our study. We want to thank Jeremy Schreiner and Livia Tomasini for technical assistance and Scott Norton for guidance in performing the transcriptome analyses. We thank Michael Higley and Riccardo Parra for use of the dual photon microscope. We are grateful to Drs. Christopher Pittenger and Nenad Sestan for comments and suggestions on an earlier version of this work. We acknowledge the Yale Center for Clinical Investigation for clinical support in obtaining the biopsy specimens, the Yale Stem Cell Center for the generation of the iPSC lines, and the Yale Center for Genome Analysis for library preparation and sequencing. We thank Dr. Pamela Ventola, Dr. Katarzyna Chawarska and Dr. Kevin Pelphrey for help with recruitment of control subjects.

Funding

The recruitment and production of iPSC lines for control subjects were supported by the following grants: MH087879, MH089176, and MH109648 from the National Institutes of Health, and by the Simons Foundation. The recruitment and production of iPSC lines for TS subjects were supported by MH118453 from the National Institutes of Health, by the NARSAD- Brain and Behavior Research Fund and by the Tourette Association of America.

Author information

Authors and Affiliations

Authors

Contributions

FMV conceived the study, designed and supervise experiments; JFL, RAK, AL-W, MHB, helped recruit patients and obtained clinical data; AS evaluated donor subjects and obtained skin biopsies; MVB contributed to the experimental design, cultured primary cells, performed reprogramming, developed the BG organoid protocol, generated organoid preps, processed them for all assays and performed and analyzed all experiments; JM oversaw organoid protocol development and optimization; YK performed the RNA-seq bioinformatic analyses; MVB, JM, YK and FMV generated display items and wrote the manuscript; all authors provided edits and comments on the manuscript.

Corresponding author

Correspondence to Flora M. Vaccarino.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary information

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Brady, M.V., Mariani, J., Koca, Y. et al. Mispatterning and interneuron deficit in Tourette Syndrome basal ganglia organoids. Mol Psychiatry 27, 5007–5019 (2022). https://doi.org/10.1038/s41380-022-01880-5

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41380-022-01880-5

Search

Quick links