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:

Ankyrin-G regulates forebrain connectivity and network synchronization via interaction with GABARAP

A Correction to this article was published on 31 January 2019

This article has been updated

Abstract

GABAergic circuits are critical for the synchronization and higher order function of brain networks. Defects in this circuitry are linked to neuropsychiatric diseases, including bipolar disorder, schizophrenia, and autism. Work in cultured neurons has shown that ankyrin-G plays a key role in the regulation of GABAergic synapses on the axon initial segment and somatodendritic domain of pyramidal neurons, where it interacts directly with the GABAA receptor-associated protein (GABARAP) to stabilize cell surface GABAA receptors. Here, we generated a knock-in mouse model expressing a mutation that abolishes the ankyrin-G/GABARAP interaction (Ank3 W1989R) to understand how ankyrin-G and GABARAP regulate GABAergic circuitry in vivo. We found that Ank3 W1989R mice exhibit a striking reduction in forebrain GABAergic synapses resulting in pyramidal cell hyperexcitability and disruptions in network synchronization. In addition, we identified changes in pyramidal cell dendritic spines and axon initial segments consistent with compensation for hyperexcitability. Finally, we identified the ANK3 W1989R variant in a family with bipolar disorder, suggesting a potential role of this variant in disease. Our results highlight the importance of ankyrin-G in regulating forebrain circuitry and provide novel insights into how ANK3 loss-of-function variants may contribute to human disease.

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

Similar content being viewed by others

Data availability

Atomic coordinates and structure factors are available from the Protein Data Bank under accession code PDB 6A9X (AnkG:GABARAP).

Change history

References

  1. Benes FM. Amygdalocortical circuitry in schizophrenia: from circuits to molecules. Neuropsychopharmacology. 2010;35:239–57.

    PubMed  Google Scholar 

  2. Benes FM, Berretta S. GABAergic interneurons: implications for understanding schizophrenia and bipolar disorder. Neuropsychopharmacology. 2001;25:1–27.

    CAS  PubMed  Google Scholar 

  3. Konradi C, Zimmerman EI, Yang CK, Lohmann KM, Gresch P, Pantazopoulos H, et al. Hippocampal interneurons in bipolar disorder. Arch Gen Psychiatry. 2011;68:340–50.

    PubMed  Google Scholar 

  4. Lazarus MS, Krishnan K, Huang ZJ. GAD67 deficiency in parvalbumin interneurons produces deficits in inhibitory transmission and network disinhibition in mouse prefrontal cortex. Cereb Cortex. 2015;25:1290–6.

    PubMed  Google Scholar 

  5. Ozerdema A, Guntekind B, Atagune MI, Basar E. Brain oscillations in bipolar disorder in search of new biomarkers. Suppl Clin Neurophysiol. 2013;62:207–21.

    PubMed  Google Scholar 

  6. Schubert KO, Focking M, Cotter DR. Proteomic pathway analysis of the hippocampus in schizophrenia and bipolar affective disorder implicates 14-3-3 signaling, aryl hydrocarbon receptor signaling, and glucose metabolism: potential roles in GABAergic interneuron pathology. Schizophr Res. 2015;167:64–72.

    PubMed  Google Scholar 

  7. Sohal VS. Insights into cortical oscillations arising from optogenetic studies. Biol Psychiatry. 2012;71:1039–45.

    PubMed  PubMed Central  Google Scholar 

  8. Torrey EF, Barci BM, Webster MJ, Bartko JJ, Meador-Woodruff JH, Knable MB. Neurochemical markers for schizophrenia, bipolar disorder, and major depression in postmortem brains. Biol Psychiatry. 2005;57:252–60.

    CAS  PubMed  Google Scholar 

  9. Bennett V, Lorenzo DN. Spectrin- and ankyrin-based membrane domains and the evolution of vertebrates. Curr Top Membr. 2013;72:1–37.

    CAS  PubMed  Google Scholar 

  10. Nelson AD, Jenkins PM. Axonal membranes and their domains: assembly and function of the axon initial segment and node of Ranvier. Front Cell Neurosci. 2017;11:136.

    PubMed  PubMed Central  Google Scholar 

  11. Kordeli E, Lambert S, Bennett V, Ankyrin G. A new ankyrin gene with neural-specific isoforms localized at the axonal initial segment and node of Ranvier. J Biol Chem. 1995;270:2352–9.

    CAS  PubMed  Google Scholar 

  12. Nelson AD, Jenkins PM. Axonal membranes and their domains: assembly and function of the axon initial segment and node of Ranvier. Front Cell Neurosci. 2017;11:136.

    PubMed  PubMed Central  Google Scholar 

  13. Tseng WC, Jenkins PM, Tanaka M, Mooney R, Bennett V. Giant ankyrin-G stabilizes somatodendritic GABAergic synapses through opposing endocytosis of GABAA receptors. Proc Natl Acad Sci USA. 2015;112:1214–9.

    CAS  PubMed  Google Scholar 

  14. Chen ZW, Olsen RW. GABAA receptor associated proteins: a key factor regulating GABAA receptor function. J Neurochem. 2007;100:279–94.

    CAS  PubMed  Google Scholar 

  15. Alemu EA, Lamark T, Torgersen KM, Birgisdottir AB, Larsen KB, Jain A, et al. ATG8 family proteins act as scaffolds for assembly of the ULK complex: sequence requirements for LC3-interacting region (LIR) motifs. J Biol Chem. 2012;287:39275–90.

    CAS  PubMed  PubMed Central  Google Scholar 

  16. Jenkins PM, Kim N, Jones SL, Tseng WC, Svitkina TM, Yin HH, et al. Giant ankyrin-G: a critical innovation in vertebrate evolution of fast and integrated neuronal signaling. Proc Natl Acad Sci USA. 2015;112:957–64.

    CAS  PubMed  Google Scholar 

  17. Li J, Zhu R, Chen K, Zheng H, Zhao H, Yuan C, et al. Potent and specific Atg8-targeting autophagy inhibitory peptides from giant ankyrins. Nat Chem Biol. 2018;14:778–87.

    CAS  PubMed  Google Scholar 

  18. Rogov VV, Stolz A, Ravichandran AC, Rios-Szwed DO, Suzuki H, Kniss A, et al. Structural and functional analysis of the GABARAP interaction motif (GIM). EMBO Rep. 2017;18:1382–96.

    CAS  PubMed  PubMed Central  Google Scholar 

  19. Jenkins PM, Vasavda C, Hostettler J, Davis JQ, Abdi K, Bennett V. E-cadherin polarity is determined by a multifunction motif mediating lateral membrane retention through ankyrin-G and apical-lateral transcytosis through clathrin. J Biol Chem. 2013;288:14018–31.

    CAS  PubMed  PubMed Central  Google Scholar 

  20. Rudy B, Fishell G, Lee S, Hjerling-Leffler J. Three groups of interneurons account for nearly 100% of neocortical GABAergic neurons. Dev Neurobiol. 2011;71:45–61.

    PubMed  PubMed Central  Google Scholar 

  21. Somogyi P, Katona L, Klausberger T, Lasztoczi B, Viney TJ. Temporal redistribution of inhibition over neuronal subcellular domains underlies state-dependent rhythmic change of excitability in the hippocampus. Philos Trans R Soc Lond B Biol Sci. 2014;369:20120518.

    PubMed  PubMed Central  Google Scholar 

  22. Sohal VS, Zhang F, Yizhar O, Deisseroth K. Parvalbumin neurons and gamma rhythms enhance cortical circuit performance. Nature. 2009;459:698–702.

    CAS  PubMed  PubMed Central  Google Scholar 

  23. Tamas G, Buhl EH, Lorincz A, Somogyi P. Proximally targeted GABAergic synapses and gap junctions synchronize cortical interneurons. Nat Neurosci. 2000;3:366–71.

    CAS  PubMed  Google Scholar 

  24. Fogarty M, Grist M, Gelman D, Marin O, Pachnis V, Kessaris N. Spatial genetic patterning of the embryonic neuroepithelium generates GABAergic interneuron diversity in the adult cortex. J Neurosci. 2007;27:10935–46.

    CAS  PubMed  PubMed Central  Google Scholar 

  25. Buzsaki G, Wang XJ. Mechanisms of gamma oscillations. Annu Rev Neurosci. 2012;35:203–25.

    CAS  PubMed  PubMed Central  Google Scholar 

  26. Cardin JA, Carlen M, Meletis K, Knoblich U, Zhang F, Deisseroth K, et al. Driving fast-spiking cells induces gamma rhythm and controls sensory responses. Nature. 2009;459:663–7.

    CAS  PubMed  PubMed Central  Google Scholar 

  27. Fisahn A, Contractor A, Traub RD, Buhl EH, Heinemann SF, McBain CJ. Distinct roles for the kainate receptor subunits GluR5 and GluR6 in kainate-induced hippocampal gamma oscillations. J Neurosci. 2004;24:9658–68.

    CAS  PubMed  PubMed Central  Google Scholar 

  28. Marin O. Interneuron dysfunction in psychiatric disorders. Nat Rev Neurosci. 2012;13:107–20.

    CAS  PubMed  Google Scholar 

  29. Lin YC, Koleske AJ. Mechanisms of synapse and dendrite maintenance and their disruption in psychiatric and neurodegenerative disorders. Annu Rev Neurosci. 2010;33:349–78.

    CAS  PubMed  PubMed Central  Google Scholar 

  30. Smith KR, Kopeikina KJ, Fawcett-Patel JM, Leaderbrand K, Gao R, Schurmann B, et al. Psychiatric risk factor ANK3/ankyrin-G nanodomains regulate the structure and function of glutamatergic synapses. Neuron. 2014;84:399–415.

    CAS  PubMed  PubMed Central  Google Scholar 

  31. Lek M, Karczewski KJ, Minikel EV, Samocha KE, Banks E, Fennell T, et al. Analysis of protein-coding genetic variation in 60,706 humans. Nature. 2016;536:285–91.

    CAS  PubMed  PubMed Central  Google Scholar 

  32. Goes FS, Pirooznia M, Parla JS, Kramer M, Ghiban E, Mavruk S, et al. Exome Sequencing of Familial Bipolar Disorder. JAMA Psychiatry. 2016;73:590–7.

    PubMed  PubMed Central  Google Scholar 

  33. Harrison PJ, Geddes JR, Tunbridge EM. The emerging neurobiology of bipolar disorder. Trends Neurosci. 2017;41:18–30.

  34. Hatzimanolis A, Smyrnis N, Avramopoulos D, Stefanis CN, Evdokimidis I, Stefanis NC. Bipolar disorder ANK3 risk variant effect on sustained attention is replicated in a large healthy population. Psychiatr Genet. 2012;22:210–3.

    PubMed  PubMed Central  Google Scholar 

  35. Leussis MP, Madison JM, Petryshen TL. Ankyrin 3: genetic association with bipolar disorder and relevance to disease pathophysiology. Biol Mood Anxiety Disord. 2012;2:18.

    CAS  PubMed  PubMed Central  Google Scholar 

  36. Schulze TG, Detera-Wadleigh SD, Akula N, Gupta A, Kassem L, Steele J, et al. Two variants in Ankyrin 3 (ANK3) are independent genetic risk factors for bipolar disorder. Mol Psychiatry. 2009;14:487–91.

    CAS  PubMed  Google Scholar 

  37. Yamada R, Kuba H. Structural and functional plasticity at the axon initial segment. Front Cell Neurosci. 2016;10:250.

    PubMed  PubMed Central  Google Scholar 

  38. Ango F, di Cristo G, Higashiyama H, Bennett V, Wu P, Huang ZJ. Ankyrin-based subcellular gradient of neurofascin, an immunoglobulin family protein, directs GABAergic innervation at purkinje axon initial segment. Cell. 2004;119:257–72.

    CAS  PubMed  Google Scholar 

  39. Zhu S, Cordner ZA, Xiong J, Chiu CT, Artola A, Zuo Y, et al. Genetic disruption of ankyrin-G in adult mouse forebrain causes cortical synapse alteration and behavior reminiscent of bipolar disorder. Proc Natl Acad Sci USA. 2017;114:10479–84.

    CAS  PubMed  PubMed Central  Google Scholar 

  40. Lopez AY, Wang X, Xu M, Maheshwari A, Curry D, Lam S, et al. Ankyrin-G isoform imbalance and interneuronopathy link epilepsy and bipolar disorder. Mol Psychiatry. 2017;22:1464–72.

    CAS  PubMed  Google Scholar 

  41. Leussis MP, Berry-Scott EM, Saito M, Jhuang H, de Haan G, Alkan O, et al. The ANK3 bipolar disorder gene regulates psychiatric-related behaviors that are modulated by lithium and stress. Biol Psychiatry. 2013;73:683–90.

    CAS  PubMed  Google Scholar 

  42. Benes FM, Lim B, Matzilevich D, Walsh JP, Subburaju S, Minns M. Regulation of the GABA cell phenotype in hippocampus of schizophrenics and bipolars. Proc Natl Acad Sci USA. 2007;104:10164–9.

    CAS  PubMed  PubMed Central  Google Scholar 

  43. Glantz LA, Lewis DA. Decreased dendritic spine density on prefrontal cortical pyramidal neurons in schizophrenia. Arch Gen Psychiatry. 2000;57:65–73.

    CAS  PubMed  Google Scholar 

  44. Konopaske GT, Lange N, Coyle JT, Benes FM. Prefrontal cortical dendritic spine pathology in schizophrenia and bipolar disorder. JAMA Psychiatry. 2014;71:1323–31.

    PubMed  PubMed Central  Google Scholar 

  45. Lee Y, Zhang Y, Kim S, Han K. Excitatory and inhibitory synaptic dysfunction in mania: an emerging hypothesis from animal model studies. Exp Mol Med. 2018;50:12.

    PubMed Central  Google Scholar 

  46. Ohgi Y, Futamura T, Hashimoto K. Glutamate signaling in synaptogenesis and NMDA receptors as potential therapeutic targets for psychiatric disorders. Curr Mol Med. 2015;15:206–21.

    CAS  PubMed  Google Scholar 

  47. Penzes P, Buonanno A, Passafaro M, Sala C, Sweet RA. Developmental vulnerability of synapses and circuits associated with neuropsychiatric disorders. J Neurochem. 2013;126:165–82.

    CAS  PubMed  PubMed Central  Google Scholar 

  48. Penzes P, Cahill ME, Jones KA, VanLeeuwen JE, Woolfrey KM. Dendritic spine pathology in neuropsychiatric disorders. Nat Neurosci. 2011;14:285–93.

    CAS  PubMed  PubMed Central  Google Scholar 

  49. Chattopadhyaya B, Cristo GD. GABAergic circuit dysfunctions in neurodevelopmental disorders. Front Psychiatry. 2012;3:51.

    CAS  PubMed  PubMed Central  Google Scholar 

  50. Ozerdem A, Guntekin B, Saatci E, Tunca Z, Basar E. Disturbance in long distance gamma coherence in bipolar disorder. Prog Neuropsychopharmacol Biol Psychiatry. 2010;34:861–5.

    PubMed  Google Scholar 

  51. Wirgenes KV, Tesli M, Inderhaug E, Athanasiu L, Agartz I, Melle I, et al. ANK3 gene expression in bipolar disorder and schizophrenia. Br J Psychiatry. 2014;205:244–5.

    PubMed  Google Scholar 

  52. Kloth K, Denecke J, Hempel M, Johannsen J, Strom TM, Kubisch C, et al. First de novo ANK3 nonsense mutation in a boy with intellectual disability, speech impairment and autistic features. Eur J Med Genet. 2017;60:494–8.

    PubMed  Google Scholar 

  53. Iqbal Z, Vandeweyer G, van der Voet M, Waryah AM, Zahoor MY, Besseling JA, et al. Homozygous and heterozygous disruptions of ANK3: at the crossroads of neurodevelopmental and psychiatric disorders. Hum Mol Genet. 2013;22:1960–70.

    CAS  PubMed  Google Scholar 

  54. Craddock N, Sklar P. Genetics of bipolar disorder. Lancet. 2013;381:1654–62.

    CAS  PubMed  Google Scholar 

  55. Ferreira MA, O’Donovan MC, Meng YA, Jones IR, Ruderfer DM, Jones L, et al. Collaborative genome-wide association analysis supports a role for ANK3 and CACNA1C in bipolar disorder. Nat Genet. 2008;40:1056–8.

    CAS  PubMed  PubMed Central  Google Scholar 

  56. Muhleisen TW, Leber M, Schulze TG, Strohmaier J, Degenhardt F, Treutlein J, et al. Genome-wide association study reveals two new risk loci for bipolar disorder. Nat Commun. 2014;5:3339.

    PubMed  Google Scholar 

  57. Roussos P, Katsel P, Davis KL, Bitsios P, Giakoumaki SG, Jogia J, et al. Molecular and genetic evidence for abnormalities in the nodes of Ranvier in schizophrenia. Arch Gen Psychiatry. 2012;69:7–15.

    CAS  PubMed  Google Scholar 

  58. Rueckert EH, Barker D, Ruderfer D, Bergen SE, O’Dushlaine C, Luce CJ, et al. Cis-acting regulation of brain-specific ANK3 gene expression by a genetic variant associated with bipolar disorder. Mol Psychiatry. 2013;18:922–9.

    CAS  PubMed  Google Scholar 

  59. Hughes T, Hansson L, Sonderby IE, Athanasiu L, Zuber V, Tesli M, et al. A loss-of-function variant in a minor isoform of ANK3 protects against bipolar disorder and schizophrenia. Biol Psychiatry. 2016;80:323–30.

    CAS  PubMed  Google Scholar 

  60. Doyle GA, Lai AT, Chou AD, Wang MJ, Gai X, Rappaport EF, et al. Re-sequencing of ankyrin 3 exon 48 and case-control association analysis of rare variants in bipolar disorder type I. Bipolar Disord. 2012;14:809–21.

    CAS  PubMed  Google Scholar 

  61. Ament SA, Szelinger S, Glusman G, Ashworth J, Hou L, Akula N, et al. Rare variants in neuronal excitability genes influence risk for bipolar disorder. Proc Natl Acad Sci USA. 2015;112:3576–81.

    CAS  PubMed  PubMed Central  Google Scholar 

  62. Cauli B, Porter JT, Tsuzuki K, Lambolez B, Rossier J, Quenet B, et al. Classification of fusiform neocortical interneurons based on unsupervised clustering. Proc Natl Acad Sci USA. 2000;97:6144–9.

    CAS  PubMed  PubMed Central  Google Scholar 

  63. Kizhatil K, Yoon W, Mohler PJ, Davis LH, Hoffman JA, Bennett V. Ankyrin-G and beta2-spectrin collaborate in biogenesis of lateral membrane of human bronchial epithelial cells. J Biol Chem. 2007;282:2029–37.

    CAS  PubMed  Google Scholar 

  64. Cooper EC, Harrington E, Jan YN, Jan LY. M channel KCNQ2 subunits are localized to key sites for control of neuronal network oscillations and synchronization in mouse brain. J Neurosci. 2001;21:9529–40.

    CAS  PubMed  PubMed Central  Google Scholar 

  65. Davis JQ, Lambert S, Bennett V. Molecular composition of the node of Ranvier: identification of ankyrin-binding cell adhesion molecules neurofascin (mucin+/third FNIII domain-) and NrCAM at nodal axon segments. J Cell Biol. 1996;135:1355–67.

    CAS  PubMed  Google Scholar 

  66. Nurnberger JI Jr., Blehar MC, Kaufmann CA, York-Cooler C, Simpson SG, Harkavy-Friedman J, et al. Diagnostic interview for genetic studies. Rationale, unique features, and training. NIMH Genetics Initiative. Arch Gen Psychiatry. 1994;51:849–59. discussion 863-844

    PubMed  Google Scholar 

Download references

Acknowledgements

We thank the Shanghai Radiation Facility BL19U1 and BL17U1 for X-ray beam time. Funding for this work was provided by the Michigan Predoctoral Training in Genetics (T32GM007544) (ADN), NIH Early Stage Training in the Neurosciences Training Grant (T32NS076401) (JCRD and JMH), Heinz C. Prechter Bipolar Research program and Richard Tam Foundation, University of Michigan Depression Center (PMJ), R37NS076752 (LLI), National Natural Science Foundation of China (NSFC 31670734) (CW), Research Grants Council of Hong Kong (16100517 and AoE-M09-12) (MZ), R01DA020140 (KSJ), and the Howard Hughes Medical Institute (VB). Finally, we thank Dr. Ed Cooper (Baylor College of Medicine) for providing the KCNQ2N1 antibody.

Author contributions

ADN, RNFC, CW, MZ, KSJ, LLS, LLI, and PMJ designed research. PMJ, KKW, KC, and VB generated and initially characterized the Ank3 W1989R mutant mice. ADN, RNCF, JCRD, JMH, YY, JL, KC, and CW performed experiments. ADN, RNCF, JCRD, CW, MZ, KSJ, and PMJ analyzed data. MMG provided human samples and clinical diagnosis. ADN and PMJ wrote the manuscript.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to P. M. Jenkins.

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

Nelson, A.D., Caballero-Florán, R.N., Rodríguez Díaz, J.C. et al. Ankyrin-G regulates forebrain connectivity and network synchronization via interaction with GABARAP. Mol Psychiatry 25, 2800–2817 (2020). https://doi.org/10.1038/s41380-018-0308-x

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41380-018-0308-x

This article is cited by

Search

Quick links