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.

  • Letter
  • Published:

Histone H2A.Z subunit exchange controls consolidation of recent and remote memory

Abstract

Memory formation is a multi-stage process that initially requires cellular consolidation in the hippocampus, after which memories are downloaded to the cortex for maintenance, in a process termed systems consolidation1. Epigenetic mechanisms regulate both types of consolidation2,3,4,5,6,7, but histone variant exchange, in which canonical histones are replaced with their variant counterparts, is an entire branch of epigenetics that has received limited attention in the brain8,9,10,11,12 and has never, to our knowledge, been studied in relation to cognitive function. Here we show that histone H2A.Z, a variant of histone H2A, is actively exchanged in response to fear conditioning in the hippocampus and the cortex, where it mediates gene expression and restrains the formation of recent and remote memory. Our data provide evidence for H2A.Z involvement in cognitive function and specifically implicate H2A.Z as a negative regulator of hippocampal consolidation and systems consolidation, probably through downstream effects on gene expression. Moreover, alterations in H2A.Z binding at later stages of systems consolidation suggest that this histone has the capacity to mediate stable molecular modifications required for memory retention. Overall, our data introduce histone variant exchange as a novel mechanism contributing to the molecular basis of cognitive function and implicate H2A.Z as a potential therapeutic target for memory disorders.

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

Figure 1: H2A.Z exchange in CA1.
Figure 2: H2A.Z depletion in CA1.
Figure 3: RNA sequencing data depicting genome-wide transcriptional impact of AAV-mediated H2A.Z depletion.
Figure 4: H2A.Z depletion in mPFC.

Similar content being viewed by others

Accession codes

Primary accessions

Gene Expression Omnibus

References

  1. Wang, S. H. & Morris, R. G. Hippocampal-neocortical interactions in memory formation, consolidation, and reconsolidation. Annu. Rev. Psychol. 61, 49–79 (2010)

    Article  Google Scholar 

  2. Lesburgueres, E. et al. Early tagging of cortical networks is required for the formation of enduring associative memory. Science 331, 924–928 (2011)

    Article  ADS  CAS  Google Scholar 

  3. Levenson, J. M. et al. Regulation of histone acetylation during memory formation in the hippocampus. J. Biol. Chem. 279, 40545–40559 (2004)

    Article  CAS  Google Scholar 

  4. Lubin, F. D., Roth, T. L. & Sweatt, J. D. Epigenetic regulation of bdnf gene transcription in the consolidation of fear memory. J. Neurosci. 28, 10576–10586 (2008)

    Article  CAS  Google Scholar 

  5. Miller, C. A., Campbell, S. L. & Sweatt, J. D. DNA methylation and histone acetylation work in concert to regulate memory formation and synaptic plasticity. Neurobiol. Learn. Mem. 89, 599–603 (2008)

    Article  CAS  Google Scholar 

  6. Miller, C. A. et al. Cortical DNA methylation maintains remote memory. Nature Neurosci. 13, 664–666 (2010)

    Article  ADS  CAS  Google Scholar 

  7. Miller, C. A. & Sweatt, J. D. Covalent modification of DNA regulates memory formation. Neuron 53, 857–869 (2007)

    Article  CAS  Google Scholar 

  8. Pina, B. & Suau, P. Changes in histones H2A and H3 variant composition in differentiating and mature rat brain cortical neurons. Dev. Biol. 123, 51–58 (1987)

    Article  CAS  Google Scholar 

  9. Santoro, S. W. & Dulac, C. The activity-dependent histone variant H2BE modulates the life span of olfactory neurons. Elife 1, e00070 (2012)

    Article  Google Scholar 

  10. Michod, D. et al. Calcium-dependent dephosphorylation of the histone chaperone DAXX regulates H3.3 loading and transcription upon neuronal activation. Neuron 74, 122–135 (2012)

    Article  CAS  Google Scholar 

  11. Bargaje, R. et al. Proximity of H2A.Z containing nucleosome to the transcription start site influences gene expression levels in the mammalian liver and brain. Nucleic Acids Res. 40, 8965–8978 (2012)

    Article  CAS  Google Scholar 

  12. Schauer, T. et al. CAST-ChIP maps cell-type-specific chromatin states in the Drosophila central nervous system. Cell Rep 5, 271–282 (2013)

    Article  CAS  Google Scholar 

  13. Jones, P. A. Functions of DNA methylation: islands, start sites, gene bodies and beyond. Nature Rev. Genet. 13, 484–492 (2012)

    Article  CAS  Google Scholar 

  14. Bonisch, C. & Hake, S. B. Histone H2A variants in nucleosomes and chromatin: more or less stable? Nucleic Acids Res. 40, 10719–10741 (2012)

    Article  Google Scholar 

  15. Weber, C. M., Ramachandran, S. & Henikoff, S. Nucleosomes are context-specific, H2A.Z-modulated barriers to RNA polymerase. Mol. Cell 53, 819–830 (2014)

    Article  CAS  Google Scholar 

  16. Bellucci, L., Dalvai, M., Kocanova, S., Moutahir, F. & Bystricky, K. Activation of p21 by HDAC inhibitors requires acetylation of H2A.Z. PLoS ONE 8, e54102 (2013)

    Article  ADS  CAS  Google Scholar 

  17. Valdes-Mora, F. et al. Acetylation of H2A.Z is a key epigenetic modification associated with gene deregulation and epigenetic remodeling in cancer. Genome Res. 22, 307–321 (2012)

    Article  CAS  Google Scholar 

  18. Hardy, S. et al. The euchromatic and heterochromatic landscapes are shaped by antagonizing effects of transcription on H2A.Z deposition. PLoS Genet. 5, e1000687 (2009)

    Article  Google Scholar 

  19. Gevry, N., Chan, H. M., Laflamme, L., Livingston, D. M. & Gaudreau, L. p21 transcription is regulated by differential localization of histone H2A.Z. Genes Dev. 21, 1869–1881 (2007)

    Article  CAS  Google Scholar 

  20. Chauhan, S. & Boyd, D. D. Regulation of u-PAR gene expression by H2A.Z is modulated by the MEK-ERK/AP-1 pathway. Nucleic Acids Res. 40, 600–613 (2012)

    Article  CAS  Google Scholar 

  21. Nock, A., Ascano, J. M., Barrero, M. J. & Malik, S. Mediator-regulated transcription through the +1 nucleosome. Mol. Cell 48, 837–848 (2012)

    Article  CAS  Google Scholar 

  22. Smith, A. P. et al. Histone H2A.Z regulates the expression of several classes of phosphate starvation response genes but not as a transcriptional activator. Plant Physiol. 152, 217–225 (2010)

    Article  CAS  Google Scholar 

  23. Millar, C. B., Xu, F., Zhang, K. & Grunstein, M. Acetylation of H2AZ Lys 14 is associated with genome-wide gene activity in yeast. Genes Dev. 20, 711–722 (2006)

    Article  CAS  Google Scholar 

  24. Watanabe, S., Radman-Livaja, M., Rando, O. J. & Peterson, C. L. A histone acetylation switch regulates H2A.Z deposition by the SWR-C remodeling enzyme. Science 340, 195–199 (2013)

    Article  ADS  CAS  Google Scholar 

  25. Conerly, M. L. et al. Changes in H2A.Z occupancy and DNA methylation during B-cell lymphomagenesis. Genome Res. 20, 1383–1390 (2010)

    Article  CAS  Google Scholar 

  26. Baptista, T. et al. Regulation of histone H2A.Z expression is mediated by sirtuin 1 in prostate cancer. Oncotarget 4, 1673–1685 (2013)

    Article  Google Scholar 

  27. Gao, J. et al. A novel pathway regulates memory and plasticity via SIRT1 and miR-134. Nature 466, 1105–1109 (2010)

    Article  ADS  CAS  Google Scholar 

  28. Paxinos, G. & Franklin, K. The Mouse Brain in Stereotaxic Coordinates (Academic Press, 1997)

    Google Scholar 

Download references

Acknowledgements

The authors’ work is supported by DARPA grant HR0011-12-1-0015 and NIH grants MH091122, MH57014 (J.D.S.) and NSERC-PDF grant PDF 387473-10 (I.B.Z.). We would like to thank F. Sultan for providing RNA primers and K. Alison Margolies for providing the immunohistochemistry images.

Author information

Authors and Affiliations

Authors

Contributions

J.D.S. and I.B.Z. conceived the experiments. I.B.Z. conducted the experiments and B.S.P. and D.M.E. assisted in performing the experiments. J.J.D. analysed the next-generation sequencing data.

Corresponding author

Correspondence to J. David Sweatt.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Extended data figures and tables

Extended Data Figure 1 Hippocampal H2A.Z is expressed throughout the hippocampus and is inhibited 30 min after fear conditioning.

a, b, Chromogenic staining of H2A.Z (a) and negative control (b). c, Fluorescent staining of H2A.Z (red) and DAPI (blue) shows H2A.Z distribution in CA1 and dentate gyrus (DG). d, e, H2afz mRNA expression (d; n mice per group: N = 4; C = 7; S = 2; CS = 5) and H2A.Z protein expression (e; n mice per group: N = 3; C = 3; S = 4; CS = 3) 30 min after training. f, DNA methylation at the H2afz promoter 30 min after fear conditioning (n mice per group: N = 7; C = 9; S = 5; CS = 4). N, naive; C, context; S, shock; CS, context plus shock. Data expressed as mean ± s.e.m. *Follow-up comparisons with P < 0.05.

Extended Data Figure 2 H2A.Z exchange in CA1.

H2A.Z binding at −1 nucleosome (first column for each time point) and +1 nucleosome (second column for each time point) of Egr1, Fos, Bdnf IV and Ppp1cc 30 min (left; n mice per group for Egr1 and Bdnf IV: N = 7; C = 5; S = 4; CS = 6; Ppp3ca: N = 4, C = 3, S = 3, CS = 3; n mice per group for Fos and Ppp1cc: N = 4, C = 3, S = 3, CS = 3) or 2 h (right; n mice per group: N = 10; C = 2; CS = 4; S = 6) after training. Gene expression is shown in the third column for each time point (n mice per group: N = 5, C = 6; S = 2; CS = 6; for Fos and Ppp1cc: N = 3; C = 3; S = 2; CS = 2). Data are expressed as mean ± s.e.m. *Follow-up comparisons with P < 0.05.

Extended Data Figure 3 Acetylated H2A.Z binding at the −1 and +1 nucleosomes 30 min after fear conditioning in CA1.

H2A.Zac binding was investigated at the −1 nucleosome (displayed in the first column for each set of genes) and the +1 nucleosome (displayed in the second column for each set of genes) of Npas4, Egr2, Arc and Ppp3ca (left) and Egr1, Fos, Bdnf IV and Ppp1cc genes (right) 30 min after fear conditioning. n mice per group: N = 3, C = 2; S = 4; CS = 3. N, naive; C, context; S, shock; CS, context plus shock. Data are expressed as mean ± s.e.m. *Follow-up comparisons with P < 0.05.

Extended Data Figure 4 H2A.Z expression in the mPFC after training.

a, b, H2afz expression was investigated in the mPFC 30 min (a; n mice per group: N = 2; C = 3; S = 3; CS = 3) or 2 h (b; n mice per group: N = 8; C = 5; S = 4; CS = 8) after fear conditioning. N, naive; C, context; S, shock; CS, context plus shock. Data are expressed as mean ± s.e.m.

Extended Data Figure 5 H2A.Z exchange in the mPFC.

H2A.Z binding was investigated at the −1 nucleosome (displayed in the first column for each time point) and the +1 nucleosome (displayed in the second column for each time point) of Egr1, Egr2, Arc and Ppp3ca genes 2 h (left; n mice per group: N = 4; C = 4; S = 3; CS = 5), 7 days (middle; n = 4 mice per group; n for −1 Arc and +1 Ppp3ca: N = 7; C = 6; S = 4; CS = 8) or 30 days (right; n mice per group: N = 2; C = 3; S = 3; CS = 3) after fear conditioning. N, naive; C, context; S, shock; CS, context plus shock. Data are expressed as mean ± s.e.m. *Follow-up comparisons with P < 0.05.

Extended Data Figure 6 H2A.Z exchange in the mPFC.

H2A.Z binding was investigated at the −1 nucleosome (displayed in the first column for each time point) and the +1 nucleosome (displayed in the second column for each time point) of Npas4, Fos, Bdnf IV and Ppp1cc genes 2 h (left; n mice per group: N = 4; C = 2; S = 4; CS = 6), 7 days (middle; n = 4 mice per group) or 30 days (right; n mice per group: N = 2; C = 3; S = 3; CS = 3) after fear conditioning. N, naive; C, context; S, shock; CS, context plus shock. Data are expressed as mean ± s.e.m. *Follow-up comparisons with P < 0.05.

Extended Data Figure 7 Acetylated H2A.Z binding at the −1 and +1 nucleosomes 2 h after fear conditioning in the mPFC.

H2A.Zac binding was investigated at the −1 nucleosome (displayed in the first column for each set of genes) and the +1 nucleosome (displayed in the second column for each set of genes) of Egr1, Egr2, Arc and Ppp3ca (left) and Npas4, Fos, Bdnf IV and Ppp1cc genes (right) 2 h after fear conditioning; n mice per group: N = 2; C = 4; S = 3; CS = 5. N, naive; C, context; S, shock; CS, context plus shock. Data are expressed as mean ± s.e.m. *Follow-up comparisons with P < 0.05.

Extended Data Figure 8 Acetylated H2A.Z binding at the −1 and +1 nucleosomes 7 days after fear conditioning in the mPFC.

H2A.Zac binding was investigated at the −1 nucleosome (displayed in the first column for each set of genes) and the +1 nucleosome (displayed in the second column for each set of genes) of Egr1, Egr2, Arc and Ppp3ca (left) and Npas4, Fos, Bdnf IV and Ppp1cc genes (right) 7 days after fear conditioning; n mice per group: N = 4; C = 3; S = 4; CS = 4. N, naive; C, context; S, shock; CS, context plus shock. Data are expressed as mean ± s.e.m. *Follow-up comparisons with P < 0.05.

Extended Data Figure 9 Open field test in mice receiving intra-cortical scramble or H2A.Z AAV.

a, Summary of experimental design. b, There were no differences in locomotor activity between H2A.Z mice and scramble controls. c, No group differences were found in movement velocity. d, No differences were found in vertical activity. e, There were no differences in the time spent in the centre, a widely used index of anxiety (n = 8 mice per group). Data are expressed as mean ± s.e.m.

Extended Data Table 1 The list of qPCR primers for genomic and complementary DNA

Supplementary information

Supplementary Table 1

A list of differentially expressed genes in untrained mice 2 weeks after receiving intra-CA1 injections of scramble AAV or H2A.Z AAV. The results of directional, PolyA+ RNA sequencing identified 451 differentially expressed genes, of which 272 were increased and 179 were decreased in response to H2A.Z depletion. (XLSX 56 kb)

Supplementary Table 2

A list of differentially expressed genes in mice receiving intra-CA1 H2A.Z AAV injections with and without training. The results of directional, PolyA+ RNA sequencing identified 202 differentially expressed genes, of which 66 were increased and 136 were decreased 30 min after fear conditioning. (XLSX 32 kb)

PowerPoint slides

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Zovkic, I., Paulukaitis, B., Day, J. et al. Histone H2A.Z subunit exchange controls consolidation of recent and remote memory. Nature 515, 582–586 (2014). https://doi.org/10.1038/nature13707

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nature13707

This article is cited by

Comments

By submitting a comment you agree to abide by our Terms and Community Guidelines. If you find something abusive or that does not comply with our terms or guidelines please flag it as inappropriate.

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