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:

Silencing microRNA-134 produces neuroprotective and prolonged seizure-suppressive effects

Abstract

Temporal lobe epilepsy is a common, chronic neurological disorder characterized by recurrent spontaneous seizures. MicroRNAs (miRNAs) are small, noncoding RNAs that regulate post-transcriptional expression of protein-coding mRNAs, which may have key roles in the pathogenesis of neurological disorders. In experimental models of prolonged, injurious seizures (status epilepticus) and in human epilepsy, we found upregulation of miR-134, a brain-specific, activity-regulated miRNA that has been implicated in the control of dendritic spine morphology. Silencing of miR-134 expression in vivo using antagomirs reduced hippocampal CA3 pyramidal neuron dendrite spine density by 21% and rendered mice refractory to seizures and hippocampal injury caused by status epilepticus. Depletion of miR-134 after status epilepticus in mice reduced the later occurrence of spontaneous seizures by over 90% and mitigated the attendant pathological features of temporal lobe epilepsy. Thus, silencing miR-134 exerts prolonged seizure-suppressant and neuroprotective actions; determining whether these are anticonvulsant effects or are truly antiepileptogenic effects requires additional experimentation.

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: MiR-134 upregulation after status epilepticus and in epilepsy.
Figure 2: Antagomir-mediated silencing of miR-134 in mouse hippocampus.
Figure 3: Antagomir silencing of miR-134 reduces hippocampal CA3 spine density in vivo.
Figure 4: Antagomir silencing of miR-134 reduces seizure severity during status epilepticus.
Figure 5: Antagomir silencing of miR-134 protects against status epilepticus in vivo and kainic acid toxicity in vitro.
Figure 6: Antagomir silencing of miR-134 after status epilepticus reduces the number of epileptic seizures and protects against progressive TLE pathology.

Similar content being viewed by others

References

  1. Pitkänen, A. & Lukasiuk, K. Mechanisms of epileptogenesis and potential treatment targets. Lancet Neurol. 10, 173–186 (2011).

    Article  Google Scholar 

  2. DeFelipe, J. Chandelier cells and epilepsy. Brain 122, 1807–1822 (1999).

    Article  Google Scholar 

  3. McNamara, J.O., Huang, Y.Z. & Leonard, A.S. Molecular signaling mechanisms underlying epileptogenesis. Sci. STKE 2006, re12 (2006).

    Article  Google Scholar 

  4. Wetherington, J., Serrano, G. & Dingledine, R. Astrocytes in the epileptic brain. Neuron 58, 168–178 (2008).

    Article  CAS  Google Scholar 

  5. Vezzani, A., French, J., Bartfai, T. & Baram, T.Z. The role of inflammation in epilepsy. Nat. Rev. Neurol. 7, 31–40 (2011).

    Article  CAS  Google Scholar 

  6. Eacker, S.M., Dawson, T.M. & Dawson, V.L. Understanding microRNAs in neurodegeneration. Nat. Rev. Neurosci. 10, 837–841 (2009).

    Article  CAS  Google Scholar 

  7. Saugstad, J.A. MicroRNAs as effectors of brain function with roles in ischemia and injury, neuroprotection, and neurodegeneration. J. Cereb. Blood Flow Metab. 30, 1564–1576 (2010).

    Article  CAS  Google Scholar 

  8. Aronica, E. et al. Expression pattern of miR-146a, an inflammation-associated microRNA, in experimental and human temporal lobe epilepsy. Eur. J. Neurosci. 31, 1100–1107 (2010).

    Article  CAS  Google Scholar 

  9. Song, Y.J. et al. Temporal lobe epilepsy induces differential expression of hippocampal miRNAs including let-7e and miR-23a/b. Brain Res. 1387, 134–140 (2011).

    Article  CAS  Google Scholar 

  10. Ambros, V. The functions of animal microRNAs. Nature 431, 350–355 (2004).

    Article  CAS  Google Scholar 

  11. Bartel, D.P. MicroRNAs: genomics, biogenesis, mechanism, and function. Cell 116, 281–297 (2004).

    Article  CAS  Google Scholar 

  12. Lagos-Quintana, M. et al. Identification of tissue-specific microRNAs from mouse. Curr. Biol. 12, 735–739 (2002).

    Article  CAS  Google Scholar 

  13. Schratt, G.M. et al. A brain-specific microRNA regulates dendritic spine development. Nature 439, 283–289 (2006).

    Article  CAS  Google Scholar 

  14. Matsuzaki, M., Honkura, N., Ellis-Davies, G.C. & Kasai, H. Structural basis of long-term potentiation in single dendritic spines. Nature 429, 761–766 (2004).

    Article  CAS  Google Scholar 

  15. Zhou, Q., Homma, K.J. & Poo, M.M. Shrinkage of dendritic spines associated with long-term depression of hippocampal synapses. Neuron 44, 749–757 (2004).

    Article  CAS  Google Scholar 

  16. Noguchi, J., Matsuzaki, M., Ellis-Davies, G.C. & Kasai, H. Spine-neck geometry determines NMDA receptor-dependent Ca2+ signaling in dendrites. Neuron 46, 609–622 (2005).

    Article  CAS  Google Scholar 

  17. Müller, M., Gahwiler, B.H., Rietschin, L. & Thompson, S.M. Reversible loss of dendritic spines and altered excitability after chronic epilepsy in hippocampal slice cultures. Proc. Natl. Acad. Sci. USA 90, 257–261 (1993).

    Article  Google Scholar 

  18. Rensing, N. et al. In vivo imaging of dendritic spines during electrographic seizures. Ann. Neurol. 58, 888–898 (2005).

    Article  Google Scholar 

  19. Zeng, L.H. et al. Kainate seizures cause acute dendritic injury and actin depolymerization in vivo. J. Neurosci. 27, 11604–11613 (2007).

    Article  CAS  Google Scholar 

  20. Penzes, P., Cahill, M.E., Jones, K.A., VanLeeuwen, J.E. & Woolfrey, K.M. Dendritic spine pathology in neuropsychiatric disorders. Nat. Neurosci. 14, 285–293 (2011).

    Article  CAS  Google Scholar 

  21. Bhatt, D.H., Zhang, S. & Gan, W.B. Dendritic spine dynamics. Annu. Rev. Physiol. 71, 261–282 (2009).

    Article  CAS  Google Scholar 

  22. Halpain, S., Hipolito, A. & Saffer, L. Regulation of F-actin stability in dendritic spines by glutamate receptors and calcineurin. J. Neurosci. 18, 9835–9844 (1998).

    Article  CAS  Google Scholar 

  23. Saneyoshi, T., Fortin, D.A. & Soderling, T.R. Regulation of spine and synapse formation by activity-dependent intracellular signaling pathways. Curr. Opin. Neurobiol. 20, 108–115 (2010).

    Article  CAS  Google Scholar 

  24. Meng, Y. et al. Abnormal spine morphology and enhanced LTP in LIMK-1 knockout mice. Neuron 35, 121–133 (2002).

    Article  CAS  Google Scholar 

  25. Christensen, M., Larsen, L.A., Kauppinen, S. & Schratt, G. Recombinant adeno-associated virus-mediated microRNA delivery into the postnatal mouse brain reveals a role for miR-134 in dendritogenesis in vivo. Front Neural Circuits 3, 16 (2010).

    Google Scholar 

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

    Article  CAS  Google Scholar 

  27. Stark, K.L. et al. Altered brain microRNA biogenesis contributes to phenotypic deficits in a 22q11-deletion mouse model. Nat. Genet. 40, 751–760 (2008).

    Article  CAS  Google Scholar 

  28. Segal, M. Dendritic spines, synaptic plasticity and neuronal survival: activity shapes dendritic spines to enhance neuronal viability. Eur. J. Neurosci. 31, 2178–2184 (2010).

    Article  Google Scholar 

  29. Sierra-Paredes, G., Oreiro-Garcia, T., Nunez-Rodriguez, A., Vazquez-Lopez, A. & Sierra-Marcuno, G. Seizures induced by in vivo latrunculin a and jasplakinolide microperfusion in the rat hippocampus. J. Mol. Neurosci. 28, 151–160 (2006).

    Article  CAS  Google Scholar 

  30. Meller, R. et al. Ubiquitin proteasome-mediated synaptic reorganization: a novel mechanism underlying rapid ischemic tolerance. J. Neurosci. 28, 50–59 (2008).

    Article  CAS  Google Scholar 

  31. Mouri, G. et al. Unilateral hippocampal CA3-predominant damage and short latency epileptogenesis after intra-amygdala microinjection of kainic acid in mice. Brain Res. 1213, 140–151 (2008).

    Article  CAS  Google Scholar 

  32. Murphy, B.M. et al. Contrasting patterns of Bim induction and neuroprotection in Bim-deficient mice between hippocampus and neocortex after status epilepticus. Cell Death Differ. 17, 459–468 (2010).

    Article  CAS  Google Scholar 

  33. Jimenez-Mateos, E.M., Mouri, G., Conroy, R.M. & Henshall, D.C. Epileptic tolerance is associated with enduring neuroprotection and uncoupling of the relationship between CA3 damage, neuropeptide Y rearrangement and spontaneous seizures following intra-amygdala kainic acid-induced status epilepticus in mice. Neuroscience 171, 556–565 (2010).

    Article  CAS  Google Scholar 

  34. Peters, L. & Meister, G. Argonaute proteins: mediators of RNA silencing. Mol. Cell 26, 611–623 (2007).

    Article  CAS  Google Scholar 

  35. Karginov, F.V. et al. A biochemical approach to identifying microRNA targets. Proc. Natl. Acad. Sci. USA 104, 19291–19296 (2007).

    Article  CAS  Google Scholar 

  36. Jimenez-Mateos, E.M. et al. MicroRNA expression profile after status epilepticus and hippocampal neuroprotection by targeting miR-132. Am. J. Pathol. 179, 2519–2532 (2011).

    Article  CAS  Google Scholar 

  37. Krützfeldt, J. et al. Silencing of microRNAs in vivo with 'antagomirs'. Nature 438, 685–689 (2005).

    Article  Google Scholar 

  38. Krützfeldt, J. et al. Specificity, duplex degradation and subcellular localization of antagomirs. Nucleic Acids Res. 35, 2885–2892 (2007).

    Article  Google Scholar 

  39. Elmén, J. et al. LNA-mediated microRNA silencing in non-human primates. Nature 452, 896–899 (2008).

    Article  Google Scholar 

  40. Engel, T. et al. Reduced hippocampal damage and epileptic seizures after status epilepticus in mice lacking proapoptotic Puma. FASEB J. 24, 853–861 (2010).

    Article  CAS  Google Scholar 

  41. O'Tuathaigh, C.M. et al. Phenotypic characterization of spatial cognition and social behavior in mice with 'knockout' of the schizophrenia risk gene neuregulin 1. Neuroscience 147, 18–27 (2007).

    Article  CAS  Google Scholar 

  42. Merino-Serrais, P., Knafo, S., Alonso-Nanclares, L., Fernaud-Espinosa, I. & Defelipe, J. Layer-specific alterations to CA1 dendritic spines in a mouse model of Alzheimer's disease. Hippocampus 21, 1037–1044 (2011).

    Article  CAS  Google Scholar 

  43. Ballesteros-Yáñez, I., Benavides-Piccione, R., Bourgeois, J.P., Changeux, J.P. & DeFelipe, J. Alterations of cortical pyramidal neurons in mice lacking high-affinity nicotinic receptors. Proc. Natl. Acad. Sci. USA 107, 11567–11572 (2010).

    Article  Google Scholar 

  44. Engel, T. et al. Loss of p53 results in protracted electrographic seizures and development of an aggravated epileptic phenotype following status epilepticus. Cell Death Dis. 1, e79 (2010).

    Article  CAS  Google Scholar 

  45. Shinoda, S. et al. Development of a model of seizure-induced hippocampal injury with features of programmed cell death in the BALB/c mouse. J. Neurosci. Res. 76, 121–128 (2004).

    Article  CAS  Google Scholar 

  46. Pitkänen, A., Kharatishvili, I., Narkilahti, S., Lukasiuk, K. & Nissinen, J. Administration of diazepam during status epilepticus reduces development and severity of epilepsy in rat. Epilepsy Res. 63, 27–42 (2005).

    Article  Google Scholar 

  47. Nairismägi, J. et al. Progression of brain damage after status epilepticus and its association with epileptogenesis: a quantitative MRI study in a rat model of temporal lobe epilepsy. Epilepsia 45, 1024–1034 (2004).

    Article  Google Scholar 

  48. Mathern, G.W., Babb, T.L. & Armstrong, D.L. Hippocampal sclerosis. in Epilepsy: A Comprehensive Textbook Vol. 13 (eds. Engel, J.J. & Pedley, T.A.) 133–155 (Lippincott-Raven Publishers, Philadelphia, 1997).

  49. Cavazos, J.E., Golarai, G. & Sutula, T.P. Mossy fiber synaptic reorganization induced by kindling: time course of development, progression, and permanence. J. Neurosci. 11, 2795–2803 (1991).

    Article  CAS  Google Scholar 

  50. Hu, K. et al. Expression profile of microRNAs in rat hippocampus following lithium-pilocarpine-induced status epilepticus. Neurosci. Lett. 488, 252–257 (2011).

    Article  CAS  Google Scholar 

  51. Tao, J. et al. Deletion of astroglial dicer causes non–cell-autonomous neuronal dysfunction and degeneration. J. Neurosci. 31, 8306–8319 (2011).

    Article  CAS  Google Scholar 

  52. Araki, T., Simon, R.P., Taki, W., Lan, J.Q. & Henshall, D.C. Characterization of neuronal death induced by focally evoked limbic seizures in the C57BL/6 mouse. J. Neurosci. Res. 69, 614–621 (2002).

    Article  CAS  Google Scholar 

  53. Lee, B. et al. The CREB/CRE transcriptional pathway: protection against oxidative stress-mediated neuronal cell death. J. Neurochem. 108, 1251–1265 (2009).

    Article  CAS  Google Scholar 

  54. Brandt, C., Potschka, H., Loscher, W. & Ebert, U. N-methyl-D-aspartate receptor blockade after status epilepticus protects against limbic brain damage but not against epilepsy in kainate model of temporal lobe epilepsy. Neuroscience 118, 727–740 (2003).

    Article  CAS  Google Scholar 

  55. Jimenez-Mateos, E.M. et al. Hippocampal transcriptome after status epilepticus in mice rendered seizure damage-tolerant by epileptic preconditioning features suppressed calcium and neuronal excitability pathways. Neurobiol. Dis. 32, 442–453 (2008).

    Article  CAS  Google Scholar 

  56. Nägerl, U.V., Eberhorn, N., Cambridge, S.B. & Bonhoeffer, T. Bidirectional activity-dependent morphological plasticity in hippocampal neurons. Neuron 44, 759–767 (2004).

    Article  Google Scholar 

  57. Kim, C.H. & Lisman, J.E. A role of actin filament in synaptic transmission and long-term potentiation. J. Neurosci. 19, 4314–4324 (1999).

    Article  CAS  Google Scholar 

  58. Pavlowsky, A. et al. A postsynaptic signaling pathway that may account for the cognitive defect due to IL1RAPL1 mutation. Curr. Biol. 20, 103–115 (2010).

    Article  CAS  Google Scholar 

  59. Wermeling, D.P. Intranasal delivery of antiepileptic medications for treatment of seizures. Neurotherapeutics 6, 352–358 (2009).

    Article  CAS  Google Scholar 

  60. Paxinos, G. & Franklin, K.B.J. The Mouse Brain in Stereotaxic Coordinates 2nd edn. (Elsevier, San Diego, California, 2001).

Download references

Acknowledgements

We would like to thank J. Varley, J. Phillips and members of the Epilepsy Programme, Beaumont Hospital. We thank S. Miller-Delaney for assistance and N. Plesnila, J. Prehn and R. Simon for helpful suggestions on the manuscript. We thank the Brain and Tissue Bank for Developmental Disorders at the University of Maryland, Baltimore, Maryland. This work was supported by funding from Science Foundation Ireland awards 08/IN1/B1875 (D.C.H., E.M.J.-M., K.T., G.M. and T.S.), 11/TIDA/B1988 (D.C.H.) and 07/IN.1/B960 (J.W. and C.O.), US National Institute of Neurological Disorders and Stroke award R56 073714 (D.C.H.), an Irish Research Council for Science, Engineering and Technology postdoctoral fellowship (E.M.J.-M.), Irish Health Research Board grant PHD/2007/11 (R.C.M.) and the Spanish Ministry of Education, Science and Innovation grant SAF2009-09394 (J.D.).

Author information

Authors and Affiliations

Authors

Contributions

E.M.J.-M. performed expression analyses, tissue culture, spine imaging, histology and epilepsy monitoring. T.E., K.T., G.M. and T.S. performed mouse modeling and telemetry. P.M.-S. and J.D. performed spine injections and data analysis. R.C.M. and S.P. performed expression studies. C.O. and J.L.W. conducted and analyzed the behavioral studies. N.D., D.F.O. and M.A.F. organized the human studies. R.M.C. performed statistical analyses. R.L.S. contributed to study design and analysis. D.C.H. and E.M.J.-M. conceived of the study, analyzed data and wrote the manuscript.

Corresponding author

Correspondence to David C Henshall.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–8 (PDF 852 kb)

Rights and permissions

Reprints and permissions

About this article

Cite this article

Jimenez-Mateos, E., Engel, T., Merino-Serrais, P. et al. Silencing microRNA-134 produces neuroprotective and prolonged seizure-suppressive effects. Nat Med 18, 1087–1094 (2012). https://doi.org/10.1038/nm.2834

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nm.2834

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