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.

  • Immediate Communication
  • Published:

Pathogenic TRIO variants associated with neurodevelopmental disorders perturb the molecular regulation of TRIO and axon pathfinding in vivo

Abstract

The RhoGEF TRIO is known to play a major role in neuronal development by controlling actin cytoskeleton remodeling, primarily through the activation of the RAC1 GTPase. Numerous de novo mutations in the TRIO gene have been identified in individuals with neurodevelopmental disorders (NDDs). We have previously established the first phenotype/genotype correlation in TRIO-associated diseases, with striking correlation between the clinical features of the individuals and the opposite modulation of RAC1 activity by TRIO variants targeting different domains. The mutations hyperactivating RAC1 are of particular interest, as they are recurrently found in patients and are associated with a severe form of NDD and macrocephaly, indicating their importance in the etiology of the disease. Yet, it remains unknown how these pathogenic TRIO variants disrupt TRIO activity at a molecular level and how they affect neurodevelopmental processes such as axon outgrowth or guidance. Here we report an additional cohort of individuals carrying a pathogenic TRIO variant that reinforces our initial phenotype/genotype correlation. More importantly, by performing conformation predictions coupled to biochemical validation, we propose a model whereby TRIO is inhibited by an intramolecular fold and NDD-associated variants relieve this inhibition, leading to RAC1 hyperactivation. Moreover, we show that in cultured primary neurons and in the zebrafish developmental model, these gain-of-function variants differentially affect axon outgrowth and branching in vitro and in vivo, as compared to loss-of-function TRIO variants. In summary, by combining clinical, molecular, cellular and in vivo data, we provide compelling new evidence for the pathogenicity of novel genetic variants targeting the TRIO gene in NDDs. We report a novel mechanism whereby the fine-tuned regulation of TRIO activity is critical for proper neuronal development and is disrupted by pathogenic mutations.

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: Impact on RAC1 signaling of newly identified TRIO variants associated to neurodevelopmental disorders.
Fig. 2: Structural modeling reveals intramolecular interactions within the TRIO spectrin domain that are altered in pathogenic variants from cluster 1.
Fig. 3: Molecular mechanisms by which pathogenic variants targeting either 7th spectrin domain or GEFD1 residues increase TRIO activity.
Fig. 4: TRIO variants of clusters 1 and 2 differentially affect neuronal morphology.
Fig. 5: TRIO variants affect growth cone morphology and alter F-actin dynamics.
Fig. 6: TRIO variants of cluster 1 and 2 differentially affect axonal development in the zebrafish.

Similar content being viewed by others

References

  1. Vissers LELM, Gilissen C, Veltman JA. Genetic studies in intellectual disability and related disorders. Nat Rev Genet. 2016;17:9–18.

    Article  CAS  PubMed  Google Scholar 

  2. Gomez TM, Letourneau PC. Actin dynamics in growth cone motility and navigation. J Neurochem. 2014;129:221–34.

    Article  CAS  PubMed  Google Scholar 

  3. Dent EW, Gupton SL, Gertler FB. The growth cone cytoskeleton in axon outgrowth and guidance. Cold Spring Harb Perspect Biol. 2011;3:a001800.

    Article  PubMed  PubMed Central  Google Scholar 

  4. Govek EE, Newey SE, Van Aelst L. The role of the Rho GTPases in neuronal development. Genes Dev. 2005;19:1–49.

    Article  CAS  PubMed  Google Scholar 

  5. Schmidt S, Debant A. Function and regulation of the Rho guanine nucleotide exchange factor Trio. Small GTPases. 2014;5:e29769.

    Article  PubMed  PubMed Central  Google Scholar 

  6. Bircher JE, Koleske AJ. Trio family proteins as regulators of cell migration and morphogenesis in development and disease - mechanisms and cellular contexts. J Cell Sci. 2021;134:jcs248393.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Paskus JD, Herring BE, Roche KW. Kalirin and trio: RhoGEFs in synaptic transmission, plasticity, and complex brain disorders. Trends Neurosci. 2020;43:505–18.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. O’Brien SP, Seipel K, Medley QG, Bronson R, Segal R, Streuli M. Skeletal muscle deformity and neuronal disorder in trio exchange factor-deficient mouse embryos [In Process Citation]. Proc Natl Acad Sci USA. 2000;97:12074–8.

    Article  PubMed  PubMed Central  Google Scholar 

  9. Zong W, Liu S, Wang X, Zhang J, Zhang T, Liu Z, et al. Trio gene is required for mouse learning ability. Brain Res. 2015;1608:82–90.

    Article  CAS  PubMed  Google Scholar 

  10. Katrancha SM, Shaw JE, Zhao AY, Myers SA, Cocco AR, Jeng AT, et al. Trio haploinsufficiency causes neurodevelopmental disease-associated deficits. Cell Rep. 2019;26:2805–17.e9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Debant A, Serra-Pages C, Seipel K, O’Brien S, Tang M, Park SH, et al. The multidomain protein Trio binds the LAR transmembrane tyrosine phosphatase, contains a protein kinase domain, and has separate rac- specific and rho-specific guanine nucleotide exchange factor domains. Proc Natl Acad Sci USA 1996;93:5466–71.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Chhatriwala MK, Betts L, Worthylake DK, Sondek J. The DH and PH domains of Trio coordinately engage Rho GTPases for their efficient activation. J Mol Biol. 2007;368:1307–20.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Bellanger JM, Estrach S, Schmidt S, Briancon-Marjollet A, Zugasti O, Fromont S, et al. Different regulation of the Trio Dbl-Homology domains by their associated PH domains. Biol Cell. 2003;95:625–34.

    Article  CAS  PubMed  Google Scholar 

  14. Chen SY, Huang PH, Cheng HJ. Disrupted-in-Schizophrenia 1-mediated axon guidance involves TRIO-RAC-PAK small GTPase pathway signaling. Proc Natl Acad Sci USA 2011;108:5861–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Neubrand VE, Thomas C, Schmidt S, Debant A, Schiavo G. Kidins220/ARMS regulates Rac1-dependent neurite outgrowth by direct interaction with the RhoGEF Trio. J Cell Sci. 2010;123:2111–23.

    Article  CAS  PubMed  Google Scholar 

  16. van Haren J, Boudeau J, Schmidt S, Basu S, Liu Z, Lammers D, et al. Dynamic microtubules catalyze formation of navigator-TRIO complexes to regulate neurite extension. Curr Biol. 2014;24:1778–85.

    Article  PubMed  Google Scholar 

  17. Sanders SJ, Murtha MT, Gupta AR, Murdoch JD, Raubeson MJ, Willsey AJ, et al. De novo mutations revealed by whole-exome sequencing are strongly associated with autism. Nature. 2012;485:237–41.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. O’Roak BJ, Vives L, Girirajan S, Karakoc E, Krumm N, Coe BP, et al. Sporadic autism exomes reveal a highly interconnected protein network of de novo mutations. Nature. 2012;485:246–50.

    Article  PubMed  PubMed Central  Google Scholar 

  19. Iossifov I, O’Roak BJ, Sanders SJ, Ronemus M, Krumm N, Levy D, et al. The contribution of de novo coding mutations to autism spectrum disorder. Nature. 2014;515:216–21.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. de Ligt J, Willemsen MH, van Bon BWM, Kleefstra T, Yntema HG, Kroes T, et al. Diagnostic exome sequencing in persons with severe intellectual disability. N Engl J Med. 2012;367:1921–9.

    Article  PubMed  Google Scholar 

  21. Ba W, Yan Y, Reijnders MRF, Schuurs-Hoeijmakers JHM, Feenstra I, Bongers EMHF, et al. TRIO loss of function is associated with mild intellectual disability and affects dendritic branching and synapse function. Hum Mol Genet. 2016;25:892–902.

    Article  CAS  PubMed  Google Scholar 

  22. Pengelly R, Greville-Heygate S, Schmidt S, Seaby E, Fagotto-Kaufmann C, Jabalameli R, et al. Mutations specific to the Rac-GEF domain of TRIO cause intellectual disability and microcephaly. J Med Genet. 2016;53:735–42.

    Article  CAS  PubMed  Google Scholar 

  23. Sadybekov A, Tian C, Arnesano C, Katritch V, Herring BE. An autism spectrum disorder-related de novo mutation hotspot discovered in the GEF1 domain of Trio. - PubMed - NCBI. Nat Comm. 2017;8:601.

    Article  Google Scholar 

  24. Kloth K, Graul-Neumann L, Hermann K, Johannsen J, Bierhals T, Kortüm F. More evidence on TRIO missense mutations in the spectrin repeat domain causing severe developmental delay and recognizable facial dysmorphism with macrocephaly. Neurogenetics. 2021;22:221–4.

    Article  CAS  PubMed  Google Scholar 

  25. Barbosa S, Greville-Heygate S, Bonnet M, Godwin A, Fagotto-Kaufmann C, Kajava AV, et al. Opposite modulation of RAC1 by mutations in TRIO is associated with distinct, domain specific neurodevelopmental disorders. Am J Hum Genet. 2020;106:338–55.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Jumper J, Evans R, Pritzel A, Green T, Figurnov M, Ronneberger O, et al. Highly accurate protein structure prediction with AlphaFold. Nature. 2021;596:583–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Cannet A, Schmidt S, Delaval B, Debant A. Identification of a mitotic Rac-GEF, Trio, that counteracts MgcRacGAP function during cytokinesis. Mol Biol Cell. 2014;25:4063–71.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Akerboom J, Chen T-W, Wardill TJ, Tian L, Marvin JS, Mutlu S, et al. Optimization of a GCaMP Calcium Indicator for Neural Activity Imaging. J Neurosci. 2012;32:13819–40.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Kimmel CB, Ballard WW, Kimmel SR, Ullmann B, Schilling TF. Stages of embryonic development of the zebrafish. Dev Dynt. 1995;203:253–310.

    Article  CAS  Google Scholar 

  30. Horstick EJ, Jordan DC, Bergeron SA, Tabor KM, Serpe M, Feldman B, et al. Increased functional protein expression using nucleotide sequence features enriched in highly expressed genes in zebrafish. Nucleic Acids Res. 2015;43:e48.

    Article  PubMed  PubMed Central  Google Scholar 

  31. Raynaud F, Janossy A, Dahl J, Bertaso F, Perroy J, Varrault A, et al. Shank3-Rich2 interaction regulates AMPA receptor recycling and synaptic long-term potentiation. J Neurosci. 2013;33:9699–715.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Aspromonte MC, Bellini M, Gasparini A, Carraro M, Bettella E, Polli R, et al. Characterization of intellectual disability and autism comorbidity through gene panel sequencing. Hum Mutat. 2019;40:1346–63.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Katrancha SM, Wu Y, Zhu M, Eipper BA, Koleske AJ, Mains RE. Neurodevelopmental disease-associated de novo mutations and rare sequence variants affect TRIO GDP/GTP exchange factor activity. Hum Mol Genet. 2017;26:4728–40.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Hall A, Lalli G. Rho and Ras GTPases in axon growth, guidance, and branching. Cold Spring Harb Perspect Biol. 2010;2:a001818.

    Article  PubMed  PubMed Central  Google Scholar 

  35. Briancon-Marjollet A, Ghogha A, Nawabi H, Triki I, Auziol C, Fromont S, et al. Trio mediates netrin-1-induced Rac1 activation in axon outgrowth and guidance. Mol Cell Biol. 2008;28:2314–23.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Song JK, Giniger E. Noncanonical Notch function in motor axon guidance is mediated by Rac GTPase and the GEF1 domain of Trio. Dev Dyn. 2011;240:324–32.

    Article  PubMed  PubMed Central  Google Scholar 

  37. Pilorge M, Fassier C, Le Corronc H, Potey A, Bai J, De Gois S, et al. Genetic and functional analyses demonstrate a role for abnormal glycinergic signaling in autism. Mol Psychiatry. 2016;21:936–45.

    Article  CAS  PubMed  Google Scholar 

  38. Scala M, Nishikawa M, Nagata K-I, Striano P. Pathophysiological mechanisms in neurodevelopmental disorders caused by Rac GTPases dysregulation: what’s behind neuro-RACopathies. Cells. 2021;10:3395.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Harms FL, Kloth K, Bley A, Denecke J, Santer R, Lessel D, et al. Activating mutations in PAK1, encoding p21-activated kinase 1, cause a neurodevelopmental disorder. Am J Hum Genet. 2018;103:579–91.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Horn S, Au M, Basel-Salmon L, Bayrak-Toydemir P, Chapin A, Cohen L, et al. De novo variants in PAK1 lead to intellectual disability with macrocephaly and seizures. Brain J Neurol. 2019. https://doi.org/10.1093/brain/awz264.

  41. Bircher J, Corcoran EE, Lam TT, Trnka MJ, Koleske AJ. Autoinhibition of the GEF activity of cytoskeletal regulatory protein Trio is disrupted in neurodevelopmental disorder-related genetic variants. J Biol Chem. 2022;298:102361.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Tian C, Jd P, E F, Kw R, Be H. Autism spectrum disorder/intellectual disability-associated mutations in trio disrupt neuroligin 1-mediated synaptogenesis. J Neurosci. 2021;41:7768–78.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Zamboni V, Armentano M, Berto G, Ciraolo E, Ghigo A, Garzotto D, et al. Hyperactivity of Rac1-GTPase pathway impairs neuritogenesis of cortical neurons by altering actin dynamics. Sci Rep. 2018;8:7254.

    Article  PubMed  PubMed Central  Google Scholar 

  44. Dogterom M, Koenderink GH. Actin-microtubule crosstalk in cell biology. Nat Rev Mol Cell Biol. 2019;20:38–54.

    Article  CAS  PubMed  Google Scholar 

  45. Korobova F, Svitkina T. Arp2/3 complex is important for filopodia formation, growth cone motility, and neuritogenesis in neuronal cells. Mol Biol Cell. 2008;19:1561–74.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Minegishi T, Inagaki N. Forces to drive neuronal migration steps. Front Cell Dev Biol. 2020;0:863.

    Article  Google Scholar 

  47. Lundquist EA. Rac proteins and the control of axon development. Curr Opin Neurobiol. 2003;13:384–90.

    Article  CAS  PubMed  Google Scholar 

  48. Struckhoff EC, Lundquist EA. The actin-binding protein UNC-115 is an effector of Rac signaling during axon pathfinding in C. elegans. Development. 2003;130:693–704.

    Article  CAS  PubMed  Google Scholar 

  49. Lundquist EA, Reddien PW, Hartwieg E, Horvitz HR, Bargmann CI. Three C. elegans Rac proteins and several alternative Rac regulators control axon guidance, cell migration and apoptotic cell phagocytosis. Development. 2001;128:4475–88.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We are grateful to all the patients and their families for their participation in this study, and in particular to Candice Williams. We also acknowledge Eva-Lena Stattin (Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden) and Philip Wyatt (Department of Obstetrics and Gynecology, York Central Hospital, Toronto, Ontario, Canada) for their patient case contribution.

We thank all members of the Debant team as well as Xavier Nicol (Institut de la Vision, Paris) for helpful discussions, and Damien Laouteouet and Jean-Christophe Perez for generating and performing initial experiments with the artificial spectrin mutants. This work was supported by grants from the Agence Nationale de la Recherche to AD (ANR-2019 TRIOTISM) and to CF (ANR-20-CE16-0019) and from The Fondation pour la Recherche Médicale (program Equipes FRM2016, DEQ20160334942) to AD. MaxB and MarB  are recipients of a PhD fellowship from the Ministère de l’Enseignement Supérieur et de la Recherche (MESR). DB is generously supported by a National Institute for Health and Care Research research professorship RP-2016-07-011. We acknowledge the imaging facility MRI, and in particular Volker Bäcker, part of the national France-BioImaging infrastructure supported by the French National Research Agency (ANR-10-INBS-04, “Investments for the future”).

Author information

Authors and Affiliations

Authors

Contributions

MaxB, AD, CF and SS conceived research and designed research experiments; DB, GG, DH, MPR, AC, DS, MM, FV and AC conducted subject recruitment and consented assessment; DB and GG interpreted subjects phenotypes; MaxB, CFK, IT, SS, FC and SB performed site-directed mutagenesis, cloning of plasmids, Western Blot and pulldown experiments; MaxB and SB prepared neuronal cultures; MaxB performed immunofluorescence and microscopy studies, with the help of MarB; MaxB and NN performed image analyses and statistical analyses. FR and CF performed the zebrafish experiments; MaxB, FR and CF analyzed and quantified images from zebrafish motoneurons and made statistics; AVK performed molecular modeling and suggested artificial TRIO mutants; SS and AD supervised the project and coordinated the study, in collaboration with DB; SS, AD, MaxB and CF wrote the manuscript. All authors reviewed and approved the manuscript.

Corresponding authors

Correspondence to Anne Debant or Susanne Schmidt.

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

Bonnet, M., Roche, F., Fagotto-Kaufmann, C. et al. Pathogenic TRIO variants associated with neurodevelopmental disorders perturb the molecular regulation of TRIO and axon pathfinding in vivo. Mol Psychiatry 28, 1527–1544 (2023). https://doi.org/10.1038/s41380-023-01963-x

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41380-023-01963-x

This article is cited by

Search

Quick links