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.

  • Brief Communication
  • Published:

miR-92a regulates expression of synaptic GluA1-containing AMPA receptors during homeostatic scaling

A Corrigendum to this article was published on 21 November 2014

This article has been updated

Abstract

We investigated whether microRNAs could regulate AMPA receptor expression during activity blockade. miR-92a strongly repressed the translation of GluA1 receptors by binding the 3′ untranslated region of rat GluA1 (also known as Gria1) mRNA and was downregulated in rat hippocampal neurons after treatment with tetrodotoxin and AP5. Deleting the seed region in GluA1 or overexpressing miR-92a blocked homeostatic scaling, indicating that miR-92a regulates the translation and synaptic incorporation of new GluA1-containing AMPA receptors.

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

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

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

Figure 1: Regulation of GluA1 mRNA translation and miRNA levels on homeostatic scaling.
Figure 2: miR-92a represses recombinant GluA1 expression.
Figure 3: miR-92a regulates endogenous GluA1-containing AMPAR expression on homeostatic scaling.

Similar content being viewed by others

Accession codes

Primary accessions

NCBI Reference Sequence

Change history

  • 04 August 2014

    In the version of this article initially published, the e-mail address for author O.T. was given as othoumin@u-bordeaux.fr and a protein in the last paragraph of the main text was given as SVA2. These should be, respectively, olivier.thoumine@u-bordeaux.fr and SV2A. The errors have been corrected in the HTML and PDF versions of the article.

References

  1. Bartel, D.P. Cell 136, 215–233 (2009).

    Article  CAS  Google Scholar 

  2. Kosik, K.S. Nat. Rev. Neurosci. 10, 754–759 (2009).

    Article  CAS  Google Scholar 

  3. Edbauer, D. et al. Neuron 65, 373–384 (2010).

    Article  CAS  Google Scholar 

  4. Lambert, T.J., Storm, D.R. & Sullivan, J.M. PLoS ONE 5, e15182 (2010).

    Article  CAS  Google Scholar 

  5. Cohen, J.E., Lee, P.R., Chen, S., Li, W. & Fields, R.D. Proc. Natl. Acad. Sci. USA 108, 11650–11655 (2011).

    Article  CAS  Google Scholar 

  6. Karr, J. et al. J. Cell Biol. 185, 685–697 (2009).

    Article  CAS  Google Scholar 

  7. Pozo, K. & Goda, Y. Neuron 66, 337–351 (2010).

    Article  CAS  Google Scholar 

  8. Turrigiano, G.G. Cell 135, 422–435 (2008).

    Article  CAS  Google Scholar 

  9. Turrigiano, G.G., Leslie, K.R., Desai, N.S., Rutherford, L.C. & Nelson, S.B. Nature 391, 892–896 (1998).

    Article  CAS  Google Scholar 

  10. Sutton, M.A. et al. Cell 125, 785–99 (2006).

    Article  CAS  Google Scholar 

  11. Cajigas, I.J. et al. Neuron 74, 453–466 (2012).

    Article  CAS  Google Scholar 

  12. Ju, W. et al. Nat. Neurosci. 7, 244–253 (2004).

    Article  CAS  Google Scholar 

  13. Aoto, J., Nam, C.I., Poon, M.M., Ting, P. & Chen, L. Neuron 60, 308–320 (2008).

    Article  CAS  Google Scholar 

  14. Poon, M.M. & Chen, L. Proc. Natl. Acad. Sci. USA 105, 20303–20308 (2008).

    Article  CAS  Google Scholar 

  15. Maghsoodi, B. et al. Proc. Natl. Acad. Sci. USA 105, 16015–16020 (2008).

    Article  CAS  Google Scholar 

  16. Heine, M. et al. Proc. Natl. Acad. Sci. USA 105, 20947–20952 (2008).

    Article  CAS  Google Scholar 

  17. Jalvy-Delvaille, S. et al. Nucleic Acids Res. 40, 1356–1365 (2012).

    Article  CAS  Google Scholar 

  18. Nowakowski, T.J. et al. Proc. Natl. Acad. Sci. USA 110, 7056–7061 (2013).

    Article  CAS  Google Scholar 

  19. Thiagarajan, T.C., Lindskog, M. & Tsien, R.W. Neuron 47, 725–737 (2005).

    Article  CAS  Google Scholar 

  20. Groth, R.D., Lindskog, M., Thiagarajan, T.C., Li, L. & Tsien, R.W. Proc. Natl. Acad. Sci. USA 108, 828–833 (2011).

    Article  CAS  Google Scholar 

  21. Lewis, B.P., Burge, C.B. & Bartel, D.P. Cell 120, 15–20 (2005).

    Article  CAS  Google Scholar 

  22. Livak, K.J. & Schmittgen, T.D. Methods 25, 402–408 (2001).

    Article  CAS  Google Scholar 

  23. Favereaux, A. et al. EMBO J. 30, 3830–3841 (2011).

    Article  CAS  Google Scholar 

  24. Kaech, S. & Banker, G. Nat. Protoc. 1, 2406–2415 (2006).

    Article  CAS  Google Scholar 

  25. Mondin, M. et al. J. Neurosci. 31, 13500–13515 (2011).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We acknowledge B. Tessier, D. Bouchet, C. Breillat, N. Retailleau, C. Genuer, F. Neca and R. Sterling for cell culture and technical assistance, and the Bordeaux Imaging Center for microscopy support. We acknowledge Y. Goda and Y. Le Feuvre for helpful comments on the manuscript. This work received funding from the Centre National de la Recherche Scientifique, Agence Nationale pour la Recherche (grant MirPAIN), Conseil Régional Aquitaine and Fondation pour la Recherche Médicale.

Author information

Authors and Affiliations

Authors

Contributions

M. Letellier, S.E., M.M., O.T. and A.F. designed the experiments and wrote the manuscript. M. Letellier, S.E., M.M., A.S., A.P., O.T. and A.F. performed the experiments and data analysis. M. Letellier, A.S., D.C., M. Landry, O.T. and A.F. revised the manuscript.

Corresponding authors

Correspondence to Olivier Thoumine or Alexandre Favereaux.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 Homeostatic scaling of GluA1 is transcription independent.

(a) In situ hybridization of GluA1 mRNAs in 15 DIV neurons. (b) GluA1 mRNAs quantified by qRT-PCR in TTX/AP5-treated and untreated neurons (Wilcoxon rank sum test, U = 9, P = 0.5317, data are represented as mean + SEM). (c) 15 DIV neurons were treated with TTX/AP5 for 4 h or untreated, and endogenous AMPARs were live labeled with an anti-GluA1 antibody. (d) Quantification of GluA1 surface staining in dendrites (Student t-test, t(97) = 4.120, *** P = 0.0001, data are represented as mean + SEM and the number of experiments is indicated in the bars).

Supplementary Figure 2 Expression level of miRNAs targeting GluA2.

Four brain-specific miRNAs were also predicted to target the GluA2 mRNA (Gria2 gene). The levels of these miRNAs were quantified by QRT-PCR analysis on extracts of neurons treated with TTX/AP5, and normalized to untreated cultures. None of the four miRNAs tested was significantly regulated in response to TTX/AP5 treatment, ANOVA followed by Newman-Keuls multiple comparison test (F(7,28) = 0.2288, P = 0.9749, data are represented as mean + SEM and the number of experiments is indicated in the bars).

Supplementary Figure 3 miRNA-mRNA interaction prediction for miR-92a/b and GluA1 mRNA.

Hybridization energies between GluA1 mRNA and miR-92a or miR-92b were predicted with intRNA tool (v1.2.5). Thermodynamic parameters are moderately in favor of a miR-92a-GluA1 interaction.

Supplementary Figure 4 Homeostatic scaling of GluA1 is local and translation dependent.

(a) Dendrites from 15 DIV neurons expressing SEP-GluA1-3'UTR-WT were transected using a micropipette (arrows), then neurons were placed back in the incubator for 4 h, with or without TTX/AP5. Surface GFP immunolabeling is shown in false colors. (b) The level of anti-GFP staining in transected dendrites was normalized to untreated condition. TTX/AP5 treatment induces a strong increase in GluA1 surface expression, suggesting a local effect (Student t-test, t(18) = 2.333, * P = 0.0314). (c) 15 DIV neurons expressing SEP-GluA1-3'UTR-WT were treated with TTX/AP5, or TTX/AP5 + cycloheximide, or left untreated for 4 h, then processed for anti-GFP surface immunostaining. The SEP (white) and anti-GFP (false colors) images are shown. (d) Protein synthesis blockade with cycloheximide occludes GluA1 scaling in response to TTX/AP5 treatment, demonstrating the requirement for protein translation (Kruskal-Wallis test, Kruskal-Wallis statistic = 14.23, P = 0.0008, followed by Dunn's multiple comparisons test, * p < 0.05, *** p < 0.001). All data are represented as mean + SEM and the number of experiments is indicated in the bars.

Supplementary Figure 5 Modulating miR-92a basal levels moderately regulates GluA1 expression.

Neurons were transfected at DIV 12 with exogenous miR-92a, miR-Ctl, or LNAs against endogenous miR-92a, and surface immunostained for GluA1 at DIV 15. The immunostaining levels were quantified for the three conditions. There is a significant difference between the miR-92a and LNA conditions but not between the miR-92a, or the LNA, and the control condition (Kruskal-Wallis test, Kruskal-Wallis statistic = 7.101, P = 0.0287, followed by Dunn's multiple comparisons test, * p < 0.05). The small and not significant effect of exogenous miR-92a on GluA1 level indicates that the basal level of endogenous is high and tightly controlled in neurons. The small and not significant effect of the LNA to endogenous miR-92a on the surface GluA1 level may be due to the fact that LNA directed against miR-92a do not only target the interaction between miR-92a and GluA1 but also have an impact on all other miR-92a targets. In addition, this might explain the toxic effects of the LNAs on neurons (not shown). Indeed, since only neurons showing no gross morphological signs of alteration were selected, it is possible that those neurons are the ones that take up the less LNAs. Such small quantities of LNAs might then not be sufficient to compete with endogenous miR-92a.

Supplementary Figure 6 Influence of TTX/AP5 treatment on mEPSC kinetics.

(a) AMPAR-mediated mEPSC frequency measured by electrophysiology. Neither TTX/AP5 treatment nor miRNA expression have any effect (Kruskal-Wallis test, Kruskal-Wallis statistic = 10.25, P = 0.0685). (b) Summary of mean mEPSC 10-90% rise-time, TTX/AP5 treatment induced a faster rise-time in neurons expressing empty vector but not for neurons overexpressing miR-92a (ANOVA, F(3, 50) = 3.637, P = 0.0188, followed by Newman-Keuls multiple comparison test, ** p < 0.01). (c) Average scaled traces of AMPA mEPSCs in the different conditions. Note a faster decay after TTX/AP5 treatment for neurons expressing empty vector but not for neurons expressing miR-92a. (d) mEPSC decay time constants for the different conditions. Since the contribution of series resistance and whole-cell capacitance to the time constant of the voltage clamp was invariant across recording conditions (Table S2), we rule out this effect as a potential cause of our measured rise time and decay kinetics. Kruskal-Wallis (Kruskal-Wallis statistic = 8.117, P = 0.0134) followed by Dunn's multiple comparisons test (* p < 0.05). All data are represented as mean + SEM, and the number of cells examined is indicated in the bars.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–6 and Supplementary Tables 1 and 2 (PDF 34505 kb)

Supplementary Methods Checklist

(PDF 364 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Letellier, M., Elramah, S., Mondin, M. et al. miR-92a regulates expression of synaptic GluA1-containing AMPA receptors during homeostatic scaling. Nat Neurosci 17, 1040–1042 (2014). https://doi.org/10.1038/nn.3762

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nn.3762

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