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:

mGluR5 in the nucleus accumbens is critical for promoting resilience to chronic stress

Abstract

Resilience to aversive events has a central role in determining whether stress leads to the development of depression. mGluR5 has been implicated in the pathophysiology of depression, but the effect of mGluR5 activity on stress resilience remains unexplored. We found that mGluR5−/− (also known as Grm5−/−) mice displayed more depression-like behaviors (for example, learned helplessness, social withdrawal and anhedonia) than control mice following exposure to various stressful stimuli. Lentiviral 'rescue' of mGluR5 in the nucleus accumbens (NAc) decreased these depression-like behaviors in mGluR5−/− mice. In the NAc, ΔFosB, whose induction promotes stress resilience, failed to be upregulated by stress in mGluR5−/− mice. Notably, targeted pharmacological activation of mGluR5 in the NAc increased ΔFosB expression. Our findings point to an essential role for mGluR5 in promoting stress resilience and suggest that a defect in mGluR5-mediated signaling in the NAc may represent an endophenotype for stress-induced depression.

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: mGluR5−/− mice exhibit depression-like behaviors following exposure to stress in a learned helplessness procedure.
Figure 2: mGluR5−/− mice exhibit enhanced susceptibility to stress-induced social avoidance and anhedonia.
Figure 3: Pharmacological modulation of mGluR5 activity influences depression-like behaviors.
Figure 4: Lentiviral mGluR5 expression in the NAc shell prevents social avoidance in mGluR5−/− mice following a 3-d social defeat stress.
Figure 5: Social avoidance levels after chronic defeat stress are correlated with mGluR5 levels in the NAc shell.
Figure 6: mGluR5 activation mediates the induction of ΔFosB.
Figure 7: Phospho-SRF (S103) mediates the induction of ΔFosB by mGluR5.

Similar content being viewed by others

References

  1. Kendler, K.S., Karkowski, L.M. & Prescott, C.A. Causal relationship between stressful life events and the onset of major depression. Am. J. Psychiatry 156, 837–841 (1999).

    CAS  PubMed  Google Scholar 

  2. Nestler, E.J. et al. Neurobiology of depression. Neuron 34, 13–25 (2002).

    CAS  PubMed  Google Scholar 

  3. Russo, S.J., Murrough, J.W., Han, M.H., Charney, D.S. & Nestler, E.J. Neurobiology of resilience. Nat. Neurosci. 15, 1475–1484 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  4. Southwick, S.M. & Charney, D.S. The science of resilience: implications for the prevention and treatment of depression. Science 338, 79–82 (2012).

    CAS  PubMed  Google Scholar 

  5. Feder, A., Nestler, E.J. & Charney, D.S. Psychobiology and molecular genetics of resilience. Nat. Rev. Neurosci. 10, 446–457 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  6. Franklin, T.B., Saab, B.J. & Mansuy, I.M. Neural mechanisms of stress resilience and vulnerability. Neuron 75, 747–761 (2012).

    CAS  PubMed  Google Scholar 

  7. Barrot, M. et al. CREB activity in the nucleus accumbens shell controls gating of behavioral responses to emotional stimuli. Proc. Natl. Acad. Sci. USA 99, 11435–11440 (2002).

    CAS  PubMed  PubMed Central  Google Scholar 

  8. Perrotti, L.I. et al. Induction of deltaFosB in reward-related brain structures after chronic stress. J. Neurosci. 24, 10594–10602 (2004).

    CAS  PubMed  PubMed Central  Google Scholar 

  9. Vialou, V. et al. Serum response factor promotes resilience to chronic social stress through the induction of DeltaFosB. J. Neurosci. 30, 14585–14592 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  10. Vialou, V. et al. DeltaFosB in brain reward circuits mediates resilience to stress and antidepressant responses. Nat. Neurosci. 13, 745–752 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  11. Niswender, C.M. & Conn, P.J. Metabotropic glutamate receptors: physiology, pharmacology, and disease. Annu. Rev. Pharmacol. Toxicol. 50, 295–322 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  12. Kim, C.H., Lee, J., Lee, J.Y. & Roche, K.W. Metabotropic glutamate receptors: phosphorylation and receptor signaling. J. Neurosci. Res. 86, 1–10 (2008).

    CAS  PubMed  Google Scholar 

  13. Nicoletti, F. et al. Metabotropic glutamate receptors: from the workbench to the bedside. Neuropharmacology 60, 1017–1041 (2011).

    CAS  PubMed  Google Scholar 

  14. Chiamulera, C. et al. Reinforcing and locomotor stimulant effects of cocaine are absent in mGluR5 null mutant mice. Nat. Neurosci. 4, 873–874 (2001).

    CAS  PubMed  Google Scholar 

  15. 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).

    CAS  PubMed  Google Scholar 

  16. Tatarczyńska, E. et al. Potential anxiolytic- and antidepressant-like effects of MPEP, a potent, selective and systemically active mGlu5 receptor antagonist. Br. J. Pharmacol. 132, 1423–1430 (2001).

    PubMed  PubMed Central  Google Scholar 

  17. Kovacčevic´, T., Skelin, I., Minuzzi, L., Rosa-Neto, P. & Diksic, M. Reduced metabotropic glutamate receptor 5 in the Flinders Sensitive Line of rats, an animal model of depression: an autoradiographic study. Brain Res. Bull. 87, 406–412 (2012).

    Google Scholar 

  18. Deschwanden, A. et al. Reduced metabotropic glutamate receptor 5 density in major depression determined by [(11)C]ABP688 PET and postmortem study. Am. J. Psychiatry 168, 727–734 (2011).

    PubMed  PubMed Central  Google Scholar 

  19. Belozertseva, I.V., Kos, T., Popik, P., Danysz, W. & Bespalov, A.Y. Antidepressant-like effects of mGluR1 and mGluR5 antagonists in the rat forced swim and the mouse tail suspension tests. Eur. Neuropsychopharmacol. 17, 172–179 (2007).

    CAS  PubMed  Google Scholar 

  20. Cryan, J.F. & Slattery, D.A. Animal models of mood disorders: Recent developments. Curr. Opin. Psychiatry 20, 1–7 (2007).

    PubMed  Google Scholar 

  21. Lim, B.K., Huang, K.W., Grueter, B.A., Rothwell, P.E. & Malenka, R.C. Anhedonia requires MC4R-mediated synaptic adaptations in nucleus accumbens. Nature 487, 183–189 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  Google Scholar 

  23. Nestler, E.J. & Hyman, S.E. Animal models of neuropsychiatric disorders. Nat. Neurosci. 13, 1161–1169 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  24. Misra, R.P. et al. L-type voltage-sensitive calcium channel activation stimulates gene expression by a serum response factor-dependent pathway. J. Biol. Chem. 269, 25483–25493 (1994).

    CAS  PubMed  Google Scholar 

  25. Janknecht, R., Hipskind, R.A., Houthaeve, T., Nordheim, A. & Stunnenberg, H.G. Identification of multiple SRF N-terminal phosphorylation sites affecting DNA binding properties. EMBO J. 11, 1045–1054 (1992).

    CAS  PubMed  PubMed Central  Google Scholar 

  26. Ulery, P.G., Rudenko, G. & Nestler, E.J. Regulation of DeltaFosB stability by phosphorylation. J. Neurosci. 26, 5131–5142 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  27. Hughes, Z.A. et al. Negative allosteric modulation of metabotropic glutamate receptor 5 results in broad spectrum activity relevant to treatment resistant depression. Neuropharmacology 66, 202–214 (2013).

    CAS  PubMed  Google Scholar 

  28. Donahue, R.J., Muschamp, J.W., Russo, S.J., Nestler, E.J. & Carlezon, W.A. Jr. Effects of striatal DeltaFosB overexpression and ketamine on social defeat stress-induced anhedonia in mice. Biol. Psychiatry 76, 550–558 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  29. Nestler, E.J. & Fos, B. A transcriptional regulator of stress and antidepressant responses. Eur. J. Pharmacol. 753, 66–72 (2014).

    PubMed  PubMed Central  Google Scholar 

  30. Britt, J.P. et al. Synaptic and behavioral profile of multiple glutamatergic inputs to the nucleus accumbens. Neuron 76, 790–803 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  31. Jia, Z. et al. Selective abolition of the NMDA component of long-term potentiation in mice lacking mGluR5. Learn. Mem. 5, 331–343 (1998).

    CAS  PubMed  PubMed Central  Google Scholar 

  32. Bougarel, L., Guitton, J., Zimmer, L., Vaugeois, J.M. & El Yacoubi, M. Behavior of a genetic mouse model of depression in the learned helplessness paradigm. Psychopharmacology (Berl.) 215, 595–605 (2011).

    CAS  Google Scholar 

  33. Chourbaji, S. et al. Learned helplessness: validity and reliability of depressive-like states in mice. Brain Res. Brain Res. Protoc. 16, 70–78 (2005).

    CAS  PubMed  Google Scholar 

  34. Golden, S.A., Covington, H.E. III., Berton, O. & Russo, S.J. A standardized protocol for repeated social defeat stress in mice. Nat. Protoc. 6, 1183–1191 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  35. Guo, M. et al. Forebrain glutamatergic neurons mediate leptin action on depression-like behaviors and synaptic depression. Transl. Psychiatry 2, e83 (2012).

    PubMed  PubMed Central  Google Scholar 

  36. Iñiguez, S.D. et al. Extracellular signal-regulated kinase-2 within the ventral tegmental area regulates responses to stress. J. Neurosci. 30, 7652–7663 (2010).

    PubMed  PubMed Central  Google Scholar 

  37. Porsolt, R.D., Brossard, G., Hautbois, C. & Roux, S. Rodent models of depression: forced swimming and tail suspension behavioral despair tests in rats and mice. Curr. Protoc. Neurosci. 8, 8.10A (2001).

    Google Scholar 

  38. Kutner, R.H., Zhang, X.Y. & Reiser, J. Production, concentration and titration of pseudotyped HIV-1-based lentiviral vectors. Nat. Protoc. 4, 495–505 (2009).

    CAS  PubMed  Google Scholar 

  39. Fowler, C.D., Lu, Q., Johnson, P.M., Marks, M.J. & Kenny, P.J. Habenular alpha5 nicotinic receptor subunit signalling controls nicotine intake. Nature 471, 597–601 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  40. Ko, S.J. et al. PKC phosphorylation regulates mGluR5 trafficking by enhancing binding of Siah-1A. J. Neurosci. 32, 16391–16401 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  41. Berton, O. et al. Essential role of BDNF in the mesolimbic dopamine pathway in social defeat stress. Science 311, 864–868 (2006).

    CAS  PubMed  Google Scholar 

  42. Bruchas, M.R. et al. Selective p38alpha MAPK deletion in serotonergic neurons produces stress resilience in models of depression and addiction. Neuron 71, 498–511 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank the Yonsei-Carl Zeiss Advanced Imaging Center for technical assistance. We thank G. Panagiotakos for her careful and critical reading. This study was supported by the Basic Science Research Program through the National Research Foundation of Korea (NRF) funded by the Ministry of Education, Science and Technology (NRF-2014R1A2A1A11051372 & no. 2007-0056092 to C.H.K., no. 2013054004 to D.G.K.), the DGIST R&D Program of the Ministry of Science, ICT and Future Planning (14-BD-16 to C.H.K.), and a faculty research grant from Yonsei University College of Medicine for 2012 (no. 6-2012-0018 to D.G.K.).

Author information

Authors and Affiliations

Authors

Contributions

S.S., O.K., C.H.K. and D.G.K. designed the experiments and analyzed the data. C.H.K. and D.G.K. prepared the manuscript. S.S., O.K., S.O. and J.C. performed the experiments. J.I.K. and S.K. provided scientific input and helped edit the manuscript.

Corresponding authors

Correspondence to Chul Hoon Kim or Dong Goo Kim.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 Basic characteristics of mGluR5−/− mice.

(a) At 16 week of age, mGluR5−/− mice showed reduced body weights compared with wild-type mice (n = 14 mice for mGluR5+/+ and 12 for each mGluR5+/– and mGluR5−/−, respectively). One-way ANOVA (F(2,35) = 6.661, P = 0.004) followed by Bonferroni post hoc test for multiple comparisons; **P = 0.003 compared with wild-type mice. (b) Locomotor activity was monitored for 15 min and analyzed in 3 min bins (n = 10 mice for each mGluR5+/+ and mGluR5+/– and 11 for mGluR5−/−, respectively). (c) Total distance traveled during the 15 min test period. No significant difference in locomotor activity was observed between genotypes. (d) Rearing counts (vertical activity) were unchanged in mGluR5+/– and mGluR5−/− mice (n = 8 mice for mGluR5+/+ and 7 for each mGluR5+/– and mGluR5−/−, respectively). Data are expressed as the mean ± SEM. (e) Western blot analysis of whole brain lysates and crude synaptosomal fractions from mGluR5+/+, mGluR5+/–, and mGluR5−/− mice. mGluR5−/− mice showed normal levels of other subtypes of mGluRs (mGluR1, mGluR2/3, and mGluR7), ionotropic glutamate receptors (GluR1, GluR2, NR2A, and NR2B), and synaptic scaffold proteins (PSD-95, PICK1, Pan-MAGUK, Chapsyn-110, and SAP-102). The experiment was successfully repeated three times. Full-length blots are presented in Supplementary Figure 9.

Supplementary Figure 2 mGluR5−/− mice exhibit depression-like behaviors in two-day FST and TST.

(a,b) FST over two consecutive days. Immobility (a) and struggling time (b) were measured during the last 4 min of the 6-min swim test period on day 1 and day 2 respectively (n = 8 mice for mGluR5+/+ and 7 for mGluR5/–). Two-sided t-test (day2 mGluR5+/+ vs mGluR5/–, t(13) = –2.504 in a and t(13) = 2.899 in b); P < 0.05 compared with wild-type mice on day 2. (c) Tail suspension test (TST) over two consecutive days. Immobility time was measured during the last 4 min of the 6-min suspension test period on day 1 and day 2 respectively. Two-sided t-test (day2 mGluR5+/+ vs mGluR5/–, t(10) = –2.236); *P = 0.049 compared with wild-type mice on day 2. (d,e) Time spent immobile per 1-min interval within the 6-min TST session on day 1 (d) and day 2 (e) (n = 7 mice for mGluR5+/+ and 5 for mGluR5/–). Two-way RM ANOVA (in e, genotype × time interaction F(5,50) = 1.552, P = 0.191) followed by Bonferroni post hoc test for multiple comparisons; *P < 0.05 compared with wild-type mice at each time point. Data are expressed as the mean ± SEM.

Supplementary Figure 3 Validation of mGluR5-expressing lentivirus and verification of correct injection site in the NAc shell and core.

(a) Diagram of the lentiviral vector used to rescue mGluR5. (b) Validation of the mGluR5-lentivirus. Western blot analysis of mGluR5 expression in HEK293FT cells 6 days after transduction of the control-LV (lentivirus) or mGluR5-LV with low, medium, and high concentrations. Full-length blots are presented in Supplementary Figure 10. The experiment was successfully repeated two times. (c) Immunohistochemistry of mGluR5 overlapping ZsGreen in the NAc shell of mice injected with mGluR5-LV. Scale bars, 50 μm. The experiment was successfully repeated three times. (d) Immunofluorescence of ZsGreen in the NAc shell (left) and NAc core (right) of mice injected with control-LV. ac, anterior commissure; AcbC, nucleus accumbens core; Acbsh, nucleus accumbens shell. Scale bars, 100 μm. The experiment was successfully repeated three times.

Supplementary Figure 4 ‘Rescue’ of mGluR5 in the NAc shell normalizes sucrose preference.

(a) Localized expression of mGluR5 within the NAc shell reversed the 3-day restraint stress-induced anhedonia observed in mGluR5−/− mice (n = 9, 5, and 6 mice for WT-control LV, KO-control LV and KO-mGluR5 LV, respectively). Two-way RM ANOVA (group × stress interaction F(2,17) = 9.042, P = 0.002) followed by Fisher LSD post hoc test for multiple comparisons; ##P = 0.001 compared with the WT-control LV following restraint stress; ††P = 0.004 compared with the respective pre-stressed mice; §P = 0.028 compared with the KO-control LV following restraint stress. (b) Rescue of mGluR5 in the NAc shell did not alter total liquid intake (water plus sucrose solution) before and after the 3-day restraint stress. Data are expressed as the mean ± SEM.

Supplementary Figure 5 Learned helplessness level after electric foot shock is correlated with mGluR5 level in NAc shell.

(a) Susceptible mice showed higher mean escape latency compared to non-defeat control mice, while resilient mice did not (n = 5, 8, and 12 mice for unshocked, susceptible, and resilient group, respectively). One-way ANOVA (F(2,22) = 72.955, P < 0.001) followed by Fisher LSD post hoc test for multiple comparisons; ***P < 0.001 compared with control group; †††P < 0.001 compared with resilient group. (b) Mean escape latency during 5-trial blocks. Two-way RM ANOVA (group × number of block interaction F(10,110) = 1.440, P = 0.172) followed by Fisher LSD post hoc test for multiple comparisons; ***P < 0.001 compared with unshocked mice; †††P < 0.001 compared with resilient group at each trial block. (c) Western blot images and quantification analysis of mGluR5 level in the NAc of susceptible mice following learned helplessness paradigm showed lower level compared to that of control mice, while resilient mice did not. One-way ANOVA (F(2,22) = 4.042, P = 0.032) followed by Fisher LSD post hoc test for multiple comparisons; *P = 0.046 compared with control group; P = 0.014 compared with resilient group. Data are expressed as the mean ± SEM. Full-length blots are presented in Supplementary Figure 10. (d) There was a significant correlation between mean escape latency and mGluR5 level in the NAc; Pearson correlation.

Supplementary Figure 6 Lentiviral ΔFosB expression in the NAc shell prevents social avoidance in mGluR5−/− mice following a 3-day social defeat stress.

(a) Diagram of the lentiviral vector used to express ΔFosB. Scale bar, 50 μm. For each mouse used, precise delivery of the virus was confirmed by visualizing ZsGreen1-expressing cells. (b) Validation of the ΔFosB-lentivirus. Western blot analysis of ΔFosB expression in HEK293FT cells 6 days after transduction of the control-LV (lentivirus) or ΔFosB-LV. Full-length blots are presented in Supplementary Figure 10. (c,d) mGluR5−/− mice that received ΔFosB LV in the NAc shell exhibited times spent in the interaction zone (c) and the corner zone (d) that were comparable to those of control LV-injected mGluR5+/+ mice (n = 4, 6, and 9 mice for WT-control LV, KO-control LV and KO-ΔFosB LV, respectively). Two-way RM ANOVA (in c, group × target interaction F(2,16) = 3.821, P = 0.044) followed by Bonferroni post hoc test for multiple comparisons; #P = 0.012, ##P = 0.006 compared with the WT-control LV in the presence of a target; P = 0.046, ††P = 0.004 compared with the respective no-target conditions; §P = 0.028, §§P = 0.002 compared with the KO-control LV in the presence of a target. Data are expressed as the mean ± SEM.

Supplementary Figure 7 Phospho-SRF (Ser103) mediates the induction of ΔFosB by mGluR5 after 3-day social defeat stress.

Representative double immunohistochemistry of the NAc shell of wild-type and mGluR5/– mice following a 3-day social defeat stress (lower 2 rows) or not (upper 2 rows). Scale bars, 50 μm. The experiment was successfully repeated three times.

Supplementary Figure 8 Un-cropped blot images in the main figures.

Supplementary Figure 9 Un-cropped blot images in the supplementary figure 1.

Supplementary Figure 10 Un-cropped blot images in the supplementary figures 3,5,6.

Supplementary information

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Shin, S., Kwon, O., Kang, J. et al. mGluR5 in the nucleus accumbens is critical for promoting resilience to chronic stress. Nat Neurosci 18, 1017–1024 (2015). https://doi.org/10.1038/nn.4028

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nn.4028

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