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:

The neuron-specific chromatin regulatory subunit BAF53b is necessary for synaptic plasticity and memory

Abstract

Recent exome sequencing studies have implicated polymorphic Brg1-Associated Factor (BAF) complexes (mammalian SWI/SNF chromatin remodeling complexes) in several human intellectual disabilities and cognitive disorders. However, it is currently unknown how mutations in BAF complexes result in impaired cognitive function. Postmitotic neurons express a neuron-specific assembly, nBAF, characterized by the neuron-specific subunit BAF53b. Mice harboring selective genetic manipulations of BAF53b have severe defects in long-term memory and long-lasting forms of hippocampal synaptic plasticity. We rescued memory impairments in BAF53b mutant mice by reintroducing BAF53b in the adult hippocampus, which suggests a role for BAF53b beyond neuronal development. The defects in BAF53b mutant mice appeared to derive from alterations in gene expression that produce abnormal postsynaptic components, such as spine structure and function, and ultimately lead to deficits in synaptic plasticity. Our results provide new insight into the role of dominant mutations in subunits of BAF complexes in human intellectual and cognitive 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: Characterization of BAF53bΔHD and Baf53b+/− mice.
Figure 2: BAF53bΔHD and Baf53b+/− mice have impaired long-term memory.
Figure 3: BAF53bΔHDlow and Baf53b+/− mice have impairments in long-term memory for contextual fear, but normal cued fear memory.
Figure 4: Hippocampal AAV-Baf53b rescues OLM, but not ORM, deficits in Baf53b+/− mice.
Figure 5: BAF53bΔHD and Baf53b+/− mice have disrupted LTP in hippocampal slices.
Figure 6: TBS-induced phosphorylation of Cofilin is altered in Baf53b+/− mice.
Figure 7: Differential gene expression in Baf53b+/− mice by RNA sequencing.

Similar content being viewed by others

Accession codes

Primary accessions

Gene Expression Omnibus

References

  1. Day, J.J. & Sweatt, J.D. Cognitive neuroepigenetics: a role for epigenetic mechanisms in learning and memory. Neurobiol. Learn. Mem. 96, 2–12 (2011).

    Article  CAS  PubMed  Google Scholar 

  2. Zahir, F.R. & Brown, C.J. Epigenetic impacts on neurodevelopment: pathophysiological mechanisms and genetic modes of action. Pediatr. Res. 69, 92R–100R (2011).

    Article  PubMed  Google Scholar 

  3. Hargreaves, D.C. & Crabtree, G.R. ATP-dependent chromatin remodeling: genetics, genomics and mechanisms. Cell Res. 21, 396–420 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  4. Wu, J.I., Lessard, J. & Crabtree, G.R. Understanding the words of chromatin regulation. Cell 136, 200–206 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Wang, W. et al. Purification and biochemical heterogeneity of the mammalian SWI-SNF complex. EMBO J. 15, 5370–5382 (1996).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Olave, I., Wang, W., Xue, Y., Kuo, A. & Crabtree, G.R. Identification of a polymorphic, neuron-specific chromatin remodeling complex. Genes Dev. 16, 2509–2517 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Zhao, K. et al. Rapid and phosphoinositol-dependent binding of the SWI/SNF-like BAF complex to chromatin after T lymphocyte receptor signaling. Cell 95, 625–636 (1998).

    Article  CAS  PubMed  Google Scholar 

  8. Wu, J.I. et al. Regulation of dendritic development by neuron-specific chromatin remodeling complexes. Neuron 56, 94–108 (2007).

    Article  CAS  PubMed  Google Scholar 

  9. Lessard, J. et al. An essential switch in subunit composition of a chromatin remodeling complex during neural development. Neuron 55, 201–215 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Yoo, A.S., Staahl, B.T., Chen, L. & Crabtree, G.R. MicroRNA-mediated switching of chromatin-remodeling complexes in neural development. Nature 460, 642–646 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Yoo, A.S. et al. MicroRNA-mediated conversion of human fibroblasts to neurons. Nature 476, 228–231 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Tsurusaki, Y. et al. Mutations affecting components of the SWI/SNF complex cause Coffin-Siris syndrome. Nat. Genet. 44, 376–378 (2012).

    Article  CAS  PubMed  Google Scholar 

  13. Santen, G.W.E. et al. Mutations in SWI/SNF chromatin remodeling complex gene ARID1B cause Coffin-Siris syndrome. Nat. Genet. 44, 379–380 (2012).

    Article  CAS  PubMed  Google Scholar 

  14. Van Houdt, J.K. et al. Heterozygous missense mutations in SMARCA2 cause Nicolaides-Baraitser syndrome. Nat. Genet. 44, 445–449 (2012).

    Article  CAS  PubMed  Google Scholar 

  15. Halgren, C. et al. Corpus callosum abnormalities, intellectual disability, speech impairment, and autism in patients with haploinsufficiency of ARID1B. Clin. Genet. 82, 248–255 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Hoyer, J., Ekici, A.B., Endele, S., Popp, B. & Zweier, C. Haploinsufficiency of ARID1B, a member of the SWI/SNF-A chromatin-remodeling complex, is a frequent cause of intellectual disability. Am. J. Hum. Genet. 90, 565–572 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Neale, B.M. et al. Patterns and rates of exonic de novo mutations in autism spectrum disorders. Nature 485, 242–245 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Park, J., Wood, M.A. & Cole, M.D. BAF53 forms distinct nuclear complexes and functions as a critical c-Myc–interacting nuclear cofactor for oncogenic transformation. Mol. Cell Biol. 22, 1307–1316 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Mayford, M. et al. Control of memory formation through regulated expression of a CaMKII transgene. Science 274, 1678–1683 (1996).

    Article  CAS  PubMed  Google Scholar 

  20. Kojima, N. et al. Rescuing impairment of long-term potentiation in fyn-deficient mice by introducing Fyn transgene. Proc. Natl. Acad. Sci. USA 94, 4761–4765 (1997).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Larson, J., Wong, D. & Lynch, G. Patterned stimulation at the theta frequency is optimal for the induction of hippocampal long-term potentiation. Brain Res. 368, 347–350 (1986).

    Article  CAS  PubMed  Google Scholar 

  22. Lauterborn, J.C. et al. Brain-derived neurotrophic factor rescues synaptic plasticity in a mouse model of fragile X syndrome. J. Neurosci. 27, 10685–10694 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Rex, C.S. et al. Different Rho GTPase–dependent signaling pathways initiate sequential steps in the consolidation of long-term potentiation. J. Cell Biol. 186, 85–97 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Chen, L.Y., Rex, C.S., Casale, M.S., Gall, C.M. & Lynch, G. Changes in synaptic morphology accompany actin signaling during LTP. J. Neurosci. 27, 5363–5372 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Bourne, J.N. & Harris, K.M. Balancing structure and function at hippocampal dendritic spines. Annu. Rev. Neurosci. 31, 47–67 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Grutzendler, J., Kasthuri, N. & Gan, W.B. Long-term dendritic spine stability in the adult cortex. Nature 420, 812–816 (2002).

    Article  CAS  PubMed  Google Scholar 

  27. Harris, K.M., Jensen, F.E. & Tsao, B. Three-dimensional structure of dendritic spines and synapses in rat hippocampus (CA1) at postnatal day 15 and adult ages: implications for the maturation of synaptic physiology and long-term potentiation. J. Neurosci. 12, 2685–2705 (1992).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Barrett, R.M. et al. Hippocampal focal knockout of CBP affects specific histone modifications, long-term potentiation and long-term memory. Neuropsychopharmacology 36, 1545–1556 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Fujita, P.A. et al. The UCSC Genome Browser database: update 2011. Nucleic Acids Res. 39, D876–D882 (2011).

    Article  CAS  PubMed  Google Scholar 

  30. Langmead, B., Trapnell, C., Pop, M. & Salzberg, S.L. Ultrafast and memory-efficient alignment of short DNA sequences to the human genome. Genome Biol. 10, R25 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Mortazavi, A., Williams, B.A., McCue, K., Schaeffer, L. & Wold, B. Mapping and quantifying mammalian transcriptomes by RNA-Seq. Nat. Methods 5, 621–628 (2008).

    Article  CAS  PubMed  Google Scholar 

  32. Kayala, M.A. & Baldi, P. Cyber-T web server: differential analysis of high-throughput data. Nucleic Acids Res. 40, W553–559 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Baldi, P. & Long, A.D. A Bayesian framework for the analysis of microarray expression data: regularized t -test and statistical inferences of gene changes. Bioinformatics 17, 509–519 (2001).

    Article  CAS  PubMed  Google Scholar 

  34. Alberini, C.M. Transcription factors in long-term memory and synaptic plasticity. Physiol. Rev. 89, 121–145 (2009).

    Article  CAS  PubMed  Google Scholar 

  35. Ashburner, M. et al. Gene ontology: tool for the unification of biology. The Gene Ontology Consortium. Nat. Genet. 25, 25–29 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Ogata, H. et al. KEGG: Kyoto Encyclopedia of Genes and Genomes. Nucleic Acids Res. 27, 29–34 (1999).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Kanehisa, M., Goto, S., Sato, Y., Furumichi, M. & Tanabe, M. KEGG for integration and interpretation of large-scale molecular data sets. Nucleic Acids Res. 40, D109–D114 (2012).

    Article  CAS  PubMed  Google Scholar 

  38. Dennis, G. et al. DAVID: Database for Annotation, Visualization and Integrated Discovery. Genome Biol. 4, 3 (2003).

    Article  Google Scholar 

  39. Wayman, G.A. et al. An activity-regulated microRNA controls dendritic plasticity by down-regulating p250GAP. Proc. Natl. Acad. Sci. USA 105, 9093–9098 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Impey, S. et al. An activity-induced microRNA controls dendritic spine formation by regulating Rac1-PAK signaling. Mol. Cell Neurosci. 43, 146–156 (2010).

    Article  CAS  PubMed  Google Scholar 

  41. Hansen, K.F., Sakamoto, K., Wayman, G.A., Impey, S. & Obrietan, K. Transgenic miR132 alters neuronal spine density and impairs novel object recognition memory. PLoS ONE 5, e15497 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Fedulov, V. et al. Evidence that long-term potentiation occurs within individual hippocampal synapses during learning. J. Neurosci. 27, 8031–8039 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Fukazawa, Y. et al. Hippocampal LTP is accompanied by enhanced F-actin content within the dendritic spine that is essential for late LTP maintenance in vivo. Neuron 38, 447–460 (2003).

    Article  CAS  PubMed  Google Scholar 

  44. Klein, M.E. et al. Homeostatic regulation of MeCP2 expression by a CREB-induced microRNA. Nat. Neurosci. 10, 1513–1514 (2007).

    Article  CAS  PubMed  Google Scholar 

  45. Nelson, E.D. & Monteggia, L.M. Epigenetics in the mature mammalian brain: effects on behavior and synaptic transmission. Neurobiol. Learn. Mem. 96, 53–60 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Amir, R.E. et al. Rett syndrome is caused by mutations in X-linked MECP2, encoding methylCpG-binding protein 2. Nat. Genet. 23, 185–188 (1999).

    Article  CAS  PubMed  Google Scholar 

  47. Levenga, J. & Willemsen, R. Perturbation of dendritic protrusions in intellectual disability. Prog. Brain Res. 197, 153–168 (2012).

    Article  CAS  PubMed  Google Scholar 

  48. Aizawa, H. Dendrite development regulated by CREST, a calcium-regulated transcriptional activator. Science 303, 197–202 (2004).

    Article  CAS  PubMed  Google Scholar 

  49. Qiu, Z. & Ghosh, A. A calcium-dependent switch in a CREST-BRG1 complex regulates activity-dependent gene expression. Neuron 60, 775–787 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Edgar, R., Domrachev, M. & Lash, A.E. Gene Expression Omnibus: NCBI gene expression and hybridization array data repository. Nucleic Acids Res. 30, 207–210 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. McQuown, S.C. et al. HDAC3 is a critical negative regulator of long-term memory formation. J. Neurosci. 31, 764–774 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Vecsey, C.G. et al. Histone deacetylase inhibitors enhance memory and synaptic plasticity via CREB:CBP-dependent transcriptional activation. J. Neurosci. 27, 6128–6140 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Babayan, A.H. et al. Integrin dynamics produce a delayed stage of long-term potentiation and memory consolidation. J. Neurosci. 32, 12854–12861 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Chen, Y. et al. Impairment of synaptic plasticity by the stress mediator CRH involves selective destruction of thin dendritic spines via RhoA signaling. Mol. Psychiatry published, online doi:10.1038/mp.2012.17 (13 March 2012)..

  55. Geisler, S., Heilmann, H. & Veh, R.W. An optimized method for simultaneous demonstration of neurons and myelinated fiber tracts for delineation of individual trunco-and palliothalamic nuclei in the mammalian brain. Histochem. Cell Biol. 117, 69–79 (2002).

    Article  CAS  PubMed  Google Scholar 

  56. Colgin, L.L., Jia, Y., Sabatier, J.-M. & Lynch, G. Blockade of NMDA receptors enhances spontaneous sharp waves in rat hippocampal slices. Neurosci. Lett. 385, 46–51 (2005).

    Article  CAS  PubMed  Google Scholar 

  57. Lawlor, P.A. et al. Novel rat Alzheimer's disease models based on AAV-mediated gene transfer to selectively increase hippocampal Abeta levels. Mol. Neurodegener. 2, 11 (2007).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  58. Pfaffl, M.W. A new mathematical model for relative quantification in real-time RT-PCR. Nucleic Acids Res. 29, e45 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Pfaffl, M.W. et al. Real-time RT-PCR quantification of insulin-like growth factor (IGF)-1, IGF-1 receptor, IGF-2, IGF-2 receptor, insulin receptor, growth hormone receptor, IGF-binding proteins 1, 2 and 3 in the bovine species. Domest. Anim. Endocrinol. 22, 91–102 (2002).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We wish to acknowledge the University of California at Irvine Institute for Genomics and Bioinformatics and Genomics High-Throughput Facility, and J. Hayes for additional computing support. We would like to thank J. Guzowski and T. Miyashita for their technical expertise and the use of the BX61 microscope and XC10 camera. This work was supported by grants from the US National Institutes of Health (MH081004 and DA025922 to M.A.W., and training grant T32-AG00096-29 to A.V.C.). G.L., E.K. and Y.J. were supported by grants from the US National Institutes of Health (P01 NS045260) and ONR (#N00014-10-1-0072). T.Z.B. was supported by US National Institutes of Health grants NS 28912 and MH73136. The work of M.Z., C.M. and P.B. was supported by grants from the National Science Foundation (IIS-0513376), the US National Institutes of Health (LM010235) and the National Library of Medicine (T15 LM07443) to P.B.

Author information

Authors and Affiliations

Authors

Contributions

A.V.-C., D.P.M., R.M.B. and M.A.W. designed the experiments. A.V.-C., D.P.M. and R.M.B. conducted the experiments. A.V.-C. and M.A.W. wrote the manuscript. E.K. and Y.J. conducted the electrophysiological experiments and analyzed the results. A.B. conducted and analyzed the pCofilin experiments. Y.C. and T.Z.B. designed and conducted the spine analysis. C.N.M., M.Z. and P.B. performed the RNA sequencing analysis. S.A., A.S., J.H., A.T., R.D. and R.J.P. performed the behavioral experiments. M.C. made the AAV-hrGFP virus. J.I.W. and G.R.C. provided technical assistance and assisted in manuscript preparation. P.B., T.Z.B. and G.L. assisted in experimental design, data analysis and manuscript preparation.

Corresponding author

Correspondence to Marcelo A Wood.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–6 and Supplementary Tables 1–13 (PDF 19404 kb)

Rights and permissions

Reprints and permissions

About this article

Cite this article

Vogel-Ciernia, A., Matheos, D., Barrett, R. et al. The neuron-specific chromatin regulatory subunit BAF53b is necessary for synaptic plasticity and memory. Nat Neurosci 16, 552–561 (2013). https://doi.org/10.1038/nn.3359

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nn.3359

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing