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:

Antidepressive effects of targeting ELK-1 signal transduction

Abstract

Depression, a devastating psychiatric disorder, is a leading cause of disability worldwide. Current antidepressants address specific symptoms of the disease, but there is vast room for improvement1. In this respect, new compounds that act beyond classical antidepressants to target signal transduction pathways governing synaptic plasticity and cellular resilience are highly warranted2,3,4. The extracellular signal–regulated kinase (ERK) pathway is implicated in mood regulation5,6,7, but its pleiotropic functions and lack of target specificity prohibit optimal drug development. Here, we identified the transcription factor ELK-1, an ERK downstream partner8, as a specific signaling module in the pathophysiology and treatment of depression that can be targeted independently of ERK. ELK1 mRNA was upregulated in postmortem hippocampal tissues from depressed suicides; in blood samples from depressed individuals, failure to reduce ELK1 expression was associated with resistance to treatment. In mice, hippocampal ELK-1 overexpression per se produced depressive behaviors; conversely, the selective inhibition of ELK-1 activation prevented depression-like molecular, plasticity and behavioral states induced by stress. Our work stresses the importance of target selectivity for a successful approach for signal-transduction-based antidepressants, singles out ELK-1 as a depression-relevant transducer downstream of ERK and brings proof-of-concept evidence for the druggability of ELK-1.

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: Increased hippocampal ELK1 mRNA expression in depression.
Fig. 2: Reduction of blood ELK1 mRNA levels reflects antidepressant efficacy.
Fig. 3: Selective inhibition of p-ELK-1 reverses stress-induced behavioral, molecular and plasticity changes.
Fig. 4: ELK-1 overexpression in the hippocampus induces depressive-like behaviors.

Similar content being viewed by others

References

  1. Perlis, R. H. Abandoning personalization to get to precision in the pharmacotherapy of depression. World Psychiatry 15, 228–235 (2016).

    Article  Google Scholar 

  2. Ionescu, D. F. & Papakostas, G. I. Experimental medication treatment approaches for depression. Transl. Psychiatry 7, e1068 (2017).

    Article  CAS  Google Scholar 

  3. Yuan, L. L., Wauson, E. & Duric, V. Kinase-mediated signaling cascades in mood disorders and antidepressant treatment. J. Neurogenet. 30, 178–184 (2016).

    Article  CAS  Google Scholar 

  4. Jeon, S. W. & Kim, Y. K. Molecular neurobiology and promising new treatment in depression. Int. J. Mol. Sci. 17, 381 (2016).

    Article  Google Scholar 

  5. Duman, C. H., Schlesinger, L., Kodama, M., Russell, D. S. & Duman, R. S. A role for MAP kinase signaling in behavioral models of depression and antidepressant treatment. Biol. Psychiatry 61, 661–670 (2007).

    Article  CAS  Google Scholar 

  6. Einat, H. et al. The role of the extracellular signal–regulated kinase signaling pathway in mood modulation. J. Neurosci. 23, 7311–7316 (2003).

    Article  CAS  Google Scholar 

  7. Labonté, B. et al. Sex-specific transcriptional signatures in human depression. Nat. Med. 23, 1102–1111 (2017).

    Article  Google Scholar 

  8. Janknecht, R., Ernst, W. H., Pingoud, V. & Nordheim, A. Activation of ternary complex factor Elk-1 by MAP kinases. EMBO J. 12, 5097–5104 (1993).

    Article  CAS  Google Scholar 

  9. Gould, T. D. & Manji, H. K. Signaling networks in the pathophysiology and treatment of mood disorders. J. Psychosom. Res. 53, 687–697 (2002).

    Article  Google Scholar 

  10. Duric, V. et al. A negative regulator of MAP kinase causes depressive behavior. Nat. Med. 16, 1328–1332 (2010).

    Article  CAS  Google Scholar 

  11. Popoli, M., Yan, Z., McEwen, B. S. & Sanacora, G. The stressed synapse: the impact of stress and glucocorticoids on glutamate transmission. Nat. Rev. Neurosci. 13, 22–37 (2011).

    Article  Google Scholar 

  12. Tse, Y. C., Bagot, R. C., Hutter, J. A., Wong, A. S. & Wong, T. P. Modulation of synaptic plasticity by stress hormone associates with plastic alteration of synaptic NMDA receptor in the adult hippocampus. PLoS One 6, e27215 (2011).

    Article  CAS  Google Scholar 

  13. Revest, J. M. et al. The MAPK pathway and Egr-1 mediate stress-related behavioral effects of glucocorticoids. Nat. Neurosci. 8, 664–672 (2005).

    Article  CAS  Google Scholar 

  14. Gutièrrez-Mecinas, M. et al. Long-lasting behavioral responses to stress involve a direct interaction of glucocorticoid receptors with ERK1/2-MSK1-Elk-1 signaling. Proc. Natl. Acad. Sci. USA 108, 13806–13811 (2011).

    Article  Google Scholar 

  15. Belzeaux, R. et al. Responder and nonresponder patients exhibit different peripheral transcriptional signatures during major depressive episode. Transl. Psychiatry 2, e185 (2012).

    Article  CAS  Google Scholar 

  16. Menke, A. et al. Dexamethasone stimulated gene expression in peripheral blood is a sensitive marker for glucocorticoid receptor resistance in depressed patients. Neuropsychopharmacology 37, 1455–1464 (2012).

    Article  CAS  Google Scholar 

  17. Vythilingam, M. et al. Childhood trauma associated with smaller hippocampal volume in women with major depression. Am. J. Psychiatry 159, 2072–2080 (2002).

    Article  Google Scholar 

  18. Turecki, G., Ernst, C., Jollant, F., Labonté, B. & Mechawar, N. The neurodevelopmental origins of suicidal behavior. Trends Neurosci. 35, 14–23 (2012).

    Article  CAS  Google Scholar 

  19. Farley, S., Apazoglou, K., Witkin, J. M., Giros, B. & Tzavara, E. T. Antidepressant-like effects of an AMPA receptor potentiator under a chronic mild stress paradigm. Int. J. Neuropsychopharmacol. 13, 1207–1218 (2010).

    Article  CAS  Google Scholar 

  20. Tsankova, N. M. et al. Sustained hippocampal chromatin regulation in a mouse model of depression and antidepressant action. Nat. Neurosci. 9, 519–525 (2006).

    Article  CAS  Google Scholar 

  21. Krishnan, V. et al. Molecular adaptations underlying susceptibility and resistance to social defeat in brain reward regions. Cell 131, 391–404 (2007).

    Article  CAS  Google Scholar 

  22. Lopez, J. P. et al. miR-1202 is a primate-specific and brain-enriched microRNA involved in major depression and antidepressant treatment. Nat. Med. 20, 764–768 (2014).

    Article  CAS  Google Scholar 

  23. Nilsson, M. et al. Elk1 and SRF transcription factors convey basal transcription and mediate glucose response via their binding sites in the human LXRB gene promoter. Nucleic Acids Res. 35, 4858–4868 (2007).

    Article  CAS  Google Scholar 

  24. Chen, E., Miller, G. E., Kobor, M. S. & Cole, S. W. Maternal warmth buffers the effects of low early-life socioeconomic status on pro-inflammatory signaling in adulthood. Mol. Psychiatry 16, 729–737 (2011).

    Article  CAS  Google Scholar 

  25. Yang, C. H., Huang, C. C. & Hsu, K. S. A critical role for protein tyrosine phosphatase nonreceptor type 5 in determining individual susceptibility to develop stress-related cognitive and morphological changes. J. Neurosci. 32, 7550–7562 (2012).

    Article  CAS  Google Scholar 

  26. Lavaur, J. et al. A TAT-DEF-Elk-1 peptide regulates the cytonuclear trafficking of Elk-1 and controls cytoskeleton dynamics. J. Neurosci. 27, 14448–14458 (2007).

    Article  CAS  Google Scholar 

  27. Jacobs, D., Glossip, D., Xing, H., Muslin, A. J. & Kornfeld, K. Multiple docking sites on substrate proteins form a modular system that mediates recognition by ERK MAP kinase. Genes Dev. 13, 163–175 (1999).

    Article  CAS  Google Scholar 

  28. Sharrocks, A. D., Yang, S. H. & Galanis, A. Docking domains and substrate-specificity determination for MAP kinases. Trends Biochem. Sci. 25, 448–453 (2000).

    Article  CAS  Google Scholar 

  29. Besnard, A. et al. Alterations of molecular and behavioral responses to cocaine by selective inhibition of Elk-1 phosphorylation. J. Neurosci. 31, 14296–14307 (2011).

    Article  CAS  Google Scholar 

  30. Crozatier, C. et al. Calcineurin (protein phosphatase 2B) is involved in the mechanisms of action of antidepressants. Neuroscience 144, 1470–1476 (2007).

    Article  CAS  Google Scholar 

  31. Svenningsson, P. et al. Involvement of striatal and extrastriatal DARPP-32 in biochemical and behavioral effects of fluoxetine (Prozac). Proc. Natl. Acad. Sci. USA 99, 3182–3187 (2002).

    Article  CAS  Google Scholar 

  32. Mouri, A. et al. Involvement of a dysfunctional dopamine-D1/N-methyl-d-aspartate-NR1 and Ca2+/calmodulin-dependent protein kinase II pathway in the impairment of latent learning in a model of schizophrenia induced by phencyclidine. Mol. Pharmacol. 71, 1598–1609 (2007).

    Article  CAS  Google Scholar 

  33. Manji, H. K. et al. Enhancing neuronal plasticity and cellular resilience to develop novel, improved therapeutics for difficult-to-treat depression. Biol. Psychiatry 53, 707–742 (2003).

    Article  CAS  Google Scholar 

  34. Duman, R. S., Aghajanian, G. K., Sanacora, G. & Krystal, J. H. Synaptic plasticity and depression: new insights from stress and rapid-acting antidepressants. Nat. Med. 22, 238–249 (2016).

    Article  CAS  Google Scholar 

  35. Nasca, C. et al. Stress dynamically regulates behavior and glutamatergic gene expression in hippocampus by opening a window of epigenetic plasticity. Proc. Natl. Acad. Sci. USA 112, 14960–14965 (2015).

    Article  CAS  Google Scholar 

  36. Guilloux, J. P., Seney, M., Edgar, N. & Sibille, E. Integrated behavioral z-scoring increases the sensitivity and reliability of behavioral phenotyping in mice: relevance to emotionality and sex. J. Neurosci. Methods 197, 21–31 (2011).

    Article  Google Scholar 

  37. Dwivedi, Y. et al. Reduced activation and expression of ERK1/2 MAP kinase in the post-mortem brain of depressed suicide subjects. J. Neurochem. 77, 916–928 (2001).

    Article  CAS  Google Scholar 

  38. Qi, X. et al. Fluoxetine increases the activity of the ERK-CREB signal system and alleviates the depressive-like behavior in rats exposed to chronic forced swim stress. Neurobiol. Dis. 31, 278–285 (2008).

    Article  CAS  Google Scholar 

  39. Svenningsson, P. et al. Biochemical and behavioral evidence for antidepressant-like effects of 5-HT6 receptor stimulation. J. Neurosci. 27, 4201–4209 (2007).

    Article  CAS  Google Scholar 

  40. Brami-Cherrier, K. et al. Parsing molecular and behavioral effects of cocaine in mitogen- and stress-activated protein kinase-1-deficient mice. J. Neurosci. 25, 11444–11454 (2005).

    Article  CAS  Google Scholar 

  41. Nolte, J. The Human Brain: An Introduction to its Functional Anatomy (Mosby/Elsevier, Philadelphia, 2009).

  42. Uher, R. et al. An inflammatory biomarker as a differential predictor of outcome of depression treatment with escitalopram and nortriptyline. Am. J. Psychiatry 171, 1278–1286 (2014).

    Article  Google Scholar 

  43. Bell, J. A., Kivimäki, M., Bullmore, E. T., Steptoe, A. & Carvalho, L. A. Repeated exposure to systemic inflammation and risk of new depressive symptoms among older adults. Transl. Psychiatry 7, e1208 (2017).

    Article  CAS  Google Scholar 

  44. Consoloni, J. L. et al. Serotonin transporter gene expression predicts the worsening of suicidal ideation and suicide attempts along a long-term follow-up of a major depressive episode. Eur. Neuropsychopharmacol. 28, 401–414 (2018).

    Article  CAS  Google Scholar 

  45. Hervé, M. et al. Translational identification of transcriptional signatures of major depression and antidepressant response. Front. Mol. Neurosci. 10, 248 (2017).

    Article  Google Scholar 

  46. Tzavara, E. T. et al. M4 muscarinic receptors regulate the dynamics of cholinergic and dopaminergic neurotransmission: relevance to the pathophysiology and treatment of related CNS pathologies. FASEB J. 18, 1410–1412 (2004).

    Article  CAS  Google Scholar 

  47. Paxinos, G. & Franklin, K. B. J. The Mouse Brain In Stereotaxic Coordinates (Gulf Professional Publishing, Houston, 2003).

    Google Scholar 

  48. Li, X., Need, A. B., Baez, M. & Witkin, J. M. Metabotropic glutamate 5 receptor antagonism is associated with antidepressant-like effects in mice. J. Pharmacol. Exp. Ther. 319, 254–259 (2006).

    Article  CAS  Google Scholar 

  49. Mombereau, C. et al. Genetic and pharmacological evidence of a role for GABA(B) receptors in the modulation of anxiety- and antidepressant-like behavior. Neuropsychopharmacology 29, 1050–1062 (2004).

    Article  CAS  Google Scholar 

  50. Dulawa, S. C. & Hen, R. Recent advances in animal models of chronic antidepressant effects: the novelty-induced hypophagia test. Neurosci. Biobehav. Rev. 29, 771–783 (2005).

    Article  CAS  Google Scholar 

  51. Gur, T. L. et al. cAMP response element-binding protein deficiency allows for increased neurogenesis and a rapid onset of antidepressant response. J. Neurosci. 27, 7860–7868 (2007).

    Article  CAS  Google Scholar 

  52. Kruk-Slomka, M., Michalak, A., Budzynska, B. & Biala, G. A comparison of mecamylamine and bupropion effects on memory-related responses induced by nicotine and scopolamine in the novel object recognition test in mice. Pharmacol. Rep. 66, 638–646 (2014).

    Article  CAS  Google Scholar 

  53. Tzavara, E. T. et al. Endocannabinoids activate transient receptor potential vanilloid 1 receptors to reduce hyperdopaminergia-related hyperactivity: therapeutic implications. Biol. Psychiatry 59, 508–515 (2006).

    Article  CAS  Google Scholar 

  54. Hancock, C. N. et al. Identification of novel extracellular signal-regulated kinase docking domain inhibitors. J. Med. Chem. 48, 4586–4595 (2005).

    Article  CAS  Google Scholar 

  55. Dournes, C., Beeské, S., Belzung, C. & Griebel, G. Deep brain stimulation in treatment-resistant depression in mice: comparison with the CRF1 antagonist, SSR125543. Prog. Neuropsychopharmacol. Biol. Psychiatry 40, 213–220 (2013).

    Article  CAS  Google Scholar 

  56. Moutsimilli, L. et al. Antipsychotics increase vesicular glutamate transporter 2 (VGLUT2) expression in thalamolimbic pathways. Neuropharmacology 54, 497–508 (2008).

    Article  CAS  Google Scholar 

  57. Meffre, D. et al. Liver X receptors alpha and beta promote myelination and remyelination in the cerebellum. Proc. Natl. Acad. Sci. USA 112, 7587–7592 (2015).

    Article  CAS  Google Scholar 

  58. Viereckel, T. et al. Midbrain gene screening identifies a new mesoaccumbal glutamatergic pathway and a marker for dopamine cells neuroprotected in Parkinson's disease. Sci. Rep. 6, 35203 (2016).

    Article  CAS  Google Scholar 

  59. Morice, E. et al. Defective synaptic transmission and structure in the dentate gyrus and selective fear memory impairment in the Rsk2 mutant mouse model of Coffin-Lowry syndrome. Neurobiol. Dis. 58, 156–168 (2013).

    Article  CAS  Google Scholar 

  60. Errington, M. L., Bliss, T. V., Morris, R. J., Laroche, S. & Davis, S. Long-term potentiation in awake mutant mice. Nature 387, 666–667 (1997).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank P. Greengard, G. G. Nomikos and R. H. Perlis for critical reading of the manuscript. We want to acknowledge M. -J. Brisorgueil and A. Besnard for help with immunohistochemistry experiments, C. Tecker for qPCR experiments in mice, P. Vanhoutte for TDE design and the Imaging Platform at the Institut de la Vision (Paris, France) for slide scanning. This research was supported in part by ERA-NET NEURON (Grant WM2NA; N.M., C.M., E.T.T.), Labex-Biopsy (AIM: E.T.T.; SignAddict: J.C.), FRC (Fondation Recherche pour le Cerveau; E.T.T.), the National Hospital Clinical Research Program (Assistance Publique-Hopitaux de Marseille; PHRC no. 2010-19: R.B.), Conseil Régional d’Aquitaine (L.M., L.G.) and the Canadian Institute for Health Research (B.G. and G.T.). R.B. was supported by a FondaMental Servier postdoctoral fellowship, and K.A. was supported by an MRT (Ministère Recherche Technologie, Ecole Doctorale MTCI) graduate award. B.G. is the holder of the Graham Boeckh Chair, and E.T.T. is a past recipient of the Bodossakis Foundation Young Scientist Award.

Author information

Authors and Affiliations

Authors

Contributions

E.T.T. and B.G. designed the study with the help of V.G. and J.C. for mouse studies and R.B. and G.T. for clinical studies; K.A., S.F., V.G. and M.-A.E.K. performed experiments and analyzed data in mouse studies; R.M., A. Barbé, C.E.A.d.M., W.J., A. Bochereau, A.O., E.I., C.G., L.M., F.L., S.G., L.G. and V.V. performed experiments in mice; R.B., N.M. and G.T. contributed clinical and postmortem samples; R.B., J.P.L. and E.C.I. generated and analyzed clinical and postmortem data; E.M. and F.M. generated in vivo electrophysiological data; Y.C.T. and T.P.W. performed electrophysiological experiments; J.G., E.C.I. and C.M. performed qPCR experiments; S.D. and S.F.  performed FISH experiments; F.Y. designed and provided viral vectors; J.C. designed and provided pharmacological tools; E.T.T. also participated in analyses of behavioral and clinical data; V.G., R.B., J.G., E.C.I., N.M., F.M., B.G. and E.T.T. wrote the paper.

Corresponding authors

Correspondence to Bruno Giros or Eleni T. Tzavara.

Ethics declarations

Competing interests

The TDE peptide used in the present article and its application in depression are protected by the published patents WO 2006/087242 and WO2010/037841, respectively, filed in Europe, the United States, Canada and Japan. This does not alter our adherence to Nature Publishing Group policies on sharing data and materials. J.C., B.G. and E.T.T. are founding shareholders of Melkin Pharmaceuticals, a biotech company, which has developed the TDE peptide as a drug candidate. The company did not have any role in study design, data collection and analysis, decision to publish or preparation of the manuscript.

Additional information

Publisher’s note: Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–21, Supplementary Table 1 and Supplementary Note

Reporting Summary

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Apazoglou, K., Farley, S., Gorgievski, V. et al. Antidepressive effects of targeting ELK-1 signal transduction. Nat Med 24, 591–597 (2018). https://doi.org/10.1038/s41591-018-0011-0

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41591-018-0011-0

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