Skip to main content
Log in

G9a-Mediated Regulation of OXT and AVP Expression in the Basolateral Amygdala Mediates Stress-Induced Lasting Behavioral Depression and Its Reversal by Exercise

  • Published:
Molecular Neurobiology Aims and scope Submit manuscript

Abstract

Chronic stress produces behavioral depression. Conversely, physical exercise is held to be beneficial in the treatment of depression. Although genomic mechanisms are likely involved in these behavioral changes, underlying mechanisms are not clearly understood. In the present study, we investigated whether stress effects and their reversal by exercise occur via genomic mechanisms in the amygdala, a core part of the limbic system important for regulating mood states. Mice treated with chronic restraint showed lasting depression-like behaviors, which were counteracted by treatment with scheduled forceful exercise. Microarray analysis identified a number of genes whose expression in the amygdala was either upregulated or downregulated after repeated stress, and these changes were reversed by exercise. Of these genes, the neuropeptides oxytocin (OXT) and arginine vasopressin (AVP) were selected as representative stress-induced and exercise-responded genes in the BLA. Stereotaxic injection of OXT or AVP receptor agonists within the BLA in normal mice produced depression-like behaviors, whereas small interfering RNA (siRNA)-mediated suppression of the OXT or AVP transcripts in the BLA was sufficient to block stress-induced depressive behaviors. Stress-induced depression-like behaviors were accompanied by a global reduction of G9a histone methyltransferase and H3K9me2 at the OXT and AVP promoters. Conversely, repeated exercise increased the levels of G9a and H3K9me2 at the OXT and AVP promoters in the BLA, which was associated with the suppression of OXT and AVP expressions. These results identify G9a-induced histone methylation at the OXT and AVP promoters in the BLA as a mechanism for mediating stress-induced lasting behavioral depression and its reversal by exercise.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2
Fig. 3
Fig. 4
Fig. 5
Fig. 6

Similar content being viewed by others

References

  1. Nestler EJ, Hyman SE (2010) Animal models of neuropsychiatric disorders. Nat Neurosci 13:1161–1169

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Strohle A (2009) Physical activity, exercise, depression and anxiety disorders. J Neural Transm 116:777–784

    Article  PubMed  Google Scholar 

  3. Krogh J, Nordentoft M, Sterne JA, Lawlor DA (2011) The effect of exercise in clinically depressed adults: systematic review and meta-analysis of randomized controlled trials. J Clin Psychiatry 72:529–538

    Article  PubMed  Google Scholar 

  4. Josefsson T, Lindwall M, Archer T (2014) Physical exercise intervention in depressive disorders: meta-analysis and systematic review. Scand J Med Sci Sports 24:259–272

    Article  CAS  PubMed  Google Scholar 

  5. Mammen G, Faulkner G (2013) Physical activity and the prevention of depression: a systematic review of prospective studies. Am J Prev Med 45:649–657

    Article  PubMed  Google Scholar 

  6. Gimpl G, Fahrenholz F (2001) The oxytocin receptor system: structure, function, and regulation. Physiol Rev 81:629–683

    CAS  PubMed  Google Scholar 

  7. Koshimizu TA, Nakamura K, Egashira N, Hiroyama M, Nonoguchi H, Tanoue A (2012) Vasopressin V1a and V1b receptors: from molecules to physiological systems. Physiol Rev 92:1813–1864

    Article  CAS  PubMed  Google Scholar 

  8. Arletti R, Bertolini A (1987) Oxytocin acts as an antidepressant in two animal models of depression. Life Sci 41:1725–1730

    Article  CAS  PubMed  Google Scholar 

  9. Yan Y, Wang YL, Su Z, Zhang Y, Guo SX, Liu AJ, Wang CH, Sun FJ et al (2014) Effect of oxytocin on the behavioral activity in the behavioral despair depression rat model. Neuropeptides 48:83–89

    Article  CAS  PubMed  Google Scholar 

  10. Winslow JT, Insel TR (2002) The social deficits of the oxytocin knockout mouse. Neuropeptides 36:221–229

    Article  CAS  PubMed  Google Scholar 

  11. Lee HJ, Caldwell HK, Macbeth AH, Tolu SG, Young WS 3rd (2008) A conditional knockout mouse line of the oxytocin receptor. Endocrinology 149:3256–3263

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Yang J, Pan YJ, Yin ZK, Hai GF, Lu L, Zhao Y, Wang DX, Wang H et al (2012) Effect of arginine vasopressin on the behavioral activity in the behavior despair depression rat model. Neuropeptides 46:141–149

    Article  CAS  PubMed  Google Scholar 

  13. van Londen L, Goekoop JG, van Kempen GM, Frankhuijzen-Sierevogel AC, Wiegant VM, van der Velde EA, De Wied D (1997) Plasma levels of arginine vasopressin elevated in patients with major depression. Neuropsychopharmacology 17:284–292

    Article  PubMed  Google Scholar 

  14. Griebel G, Simiand J, Serradeil-Le Gal C, Wagnon J, Pascal M, Scatton B, Maffrand JP, Soubrie P (2002) Anxiolytic- and antidepressant-like effects of the non-peptide vasopressin V1b receptor antagonist, SSR149415, suggest an innovative approach for the treatment of stress-related disorders. Proc Natl Acad Sci U S A 99:6370–6375

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Mlynarik M, Zelena D, Bagdy G, Makara GB, Jezova D (2007) Signs of attenuated depression-like behavior in vasopressin deficient Brattleboro rats. Horm Behav 51:395–405

    Article  CAS  PubMed  Google Scholar 

  16. Caldwell HK, Wersinger SR, Young WS 3rd (2008) The role of the vasopressin 1b receptor in aggression and other social behaviours. Prog Brain Res 170:65–72

    Article  CAS  PubMed  Google Scholar 

  17. Neumann ID, Landgraf R (2012) Balance of brain oxytocin and vasopressin: implications for anxiety, depression, and social behaviors. Trends Neurosci 35:649–659

    Article  CAS  PubMed  Google Scholar 

  18. Tsankova N, Renthal W, Kumar A, Nestler EJ (2007) Epigenetic regulation in psychiatric disorders. Nat Rev Neurosci 8:355–367

    Article  CAS  PubMed  Google Scholar 

  19. Vialou V, Feng J, Robison AJ, Nestler EJ (2013) Epigenetic mechanisms of depression and antidepressant action. Annu Rev Pharmacol Toxicol 53:59–87

    Article  CAS  PubMed  Google Scholar 

  20. Zhang X, Wen H, Shi X (2012) Lysine methylation: beyond histones. Acta Biochim Biophys Sin 44:14–27

    Article  PubMed  Google Scholar 

  21. Tachibana M, Sugimoto K, Nozaki M, Ueda J, Ohta T, Ohki M, Fukuda M, Takeda N et al (2002) G9a histone methyltransferase plays a dominant role in euchromatic histone H3 lysine 9 methylation and is essential for early embryogenesis. Genes 16:1779–1791

    Article  CAS  Google Scholar 

  22. Covington HE 3rd, Maze I, Sun H, Bomze HM, DeMaio KD, Wu EY, Dietz DM, Lobo MK et al (2011) A role for repressive histone methylation in cocaine-induced vulnerability to stress. Neuron 71:656–670

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Shinkai Y, Tachibana M (2011) H3K9 methyltransferase G9a and the related molecule GLP. Genes Dev 25:781–788

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Krishnan V, Nestler EJ (2011) Linking molecules to mood: new insight into the biology of depression. Am J Psychiatry 167:1305–1320

    Article  Google Scholar 

  25. Murray EA, Wise SP, Drevets WC (2011) Localization of dysfunction in major depressive disorder: prefrontal cortex and amygdala. Biol Psychiatry 69:e43–e54

    Article  PubMed  Google Scholar 

  26. Kim KS, Han PL (2009) Mice lacking adenylyl cyclase-5 cope badly with repeated restraint stress. J Neurosci Res 87:2983–2993

    Article  CAS  PubMed  Google Scholar 

  27. Greenwood BN, Strong PV, Loughridge AB, Day HE, Clark PJ, Mika A, Hellwinkel JE, Spence KG et al (2012) 5-HT2C receptors in the basolateral amygdala and dorsal striatum are a novel target for the anxiolytic and antidepressant effects of exercise. PLoS ONE 7:e46118

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Sah P, Faber ES, Lopez De Armentia M, Power J (2003) The amygdaloid complex: anatomy and physiology. Physiol Rev 83:803–834

    Article  CAS  PubMed  Google Scholar 

  29. Seo JS, Park JY, Choi J, Kim TK, Shin JH, Lee JK, Han PL (2012) NADPH oxidase mediates depressive behavior induced by chronic stress in mice. J Neurosci 32:9690–9699

    Article  CAS  PubMed  Google Scholar 

  30. Park JY, Kim TK, Choi J, Lee JE, Kim H, Lee EH, Han PL (2014) Implementation of a two-dimensional behavior matrix to distinguish individuals with differential depression states in a rodent model of depression. Exp Neurobiol 23:215–223

    Article  PubMed  PubMed Central  Google Scholar 

  31. Kim TK, Park JY, Han PL (2015) Physiological parameters in the blood of a murine stress-induced depression model before and after repeated passive exercise. Endocrinol Metab 30:e2

  32. Kim KS, Lee KW, Baek IS, Lim CM, Krishnan V, Lee JK, Nestler EJ, Han PL (2008) Adenylyl cyclase-5 activity in the nucleus accumbens regulates anxiety-related behavior. J Neurochem 107:105–115

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Lee KW, Kim JB, Seo JS, Kim TK, Im JY, Baek IS, Kim KS, Lee JK et al (2009) Behavioral stress accelerates plaque pathogenesis in the brain of Tg2576 mice via generation of metabolic oxidative stress. J Neurochem 108:165–175

    Article  CAS  PubMed  Google Scholar 

  34. Irizarry RA, Bolstad BM, Collin F, Cope LM, Hobbs B, Speed TP (2003) Summaries of Affymetrix GeneChip probe level data. Nucleic Acids Res 31:e15

    Article  PubMed  PubMed Central  Google Scholar 

  35. Gentleman RC, Carey VJ, Bates DM, Bolstad B, Dettling M, Dudoit S, Ellis B, Gautier L et al (2004) Bioconductor: open software development for computational biology and bioinformatics. Genome Biol 5:R80

    Article  PubMed  PubMed Central  Google Scholar 

  36. Kuo MH, Allis CD (1999) In vivo cross-linking and immunoprecipitation for studying dynamic Protein: DNA associations in a chromatin environment. Methods 19:425–433

    Article  CAS  PubMed  Google Scholar 

  37. Chakrabarti SK, James JC, Mirmira RG (2002) Quantitative assessment of gene targeting in vitro and in vivo by the pancreatic transcription factor, Pdx1. Importance of chromatin structure in directing promoter binding. J Biol Chem 277:13286–13293

    Article  CAS  PubMed  Google Scholar 

  38. Ogryzko VV, Schiltz RL, Russanova V, Howard BH, Nakatani Y (1996) The transcriptional coactivators p300 and CBP are histone acetyltransferases. Cell 87:953–959

    Article  CAS  PubMed  Google Scholar 

  39. Tachibana M, Matsumura Y, Fukuda M, Kimura H, Shinkai Y (2008) G9a/GLP complexes independently mediate H3K9 and DNA methylation to silence transcription. EMBO J 27:2681–2690

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Young WS 3rd, Gainer H (2003) Transgenesis and the study of expression, cellular targeting and function of oxytocin, vasopressin and their receptors. Neuroendocrinology 78:185–203

    Article  CAS  PubMed  Google Scholar 

  41. Viviani D, Stoop R (2008) Opposite effects of oxytocin and vasopressin on the emotional expression of the fear response. Prog Brain Res 170:207–218

    Article  CAS  PubMed  Google Scholar 

  42. Sessoms-Sikes S, Honse Y, Lovinger DM, Colbran RJ (2005) CaMKIIalpha enhances the desensitization of NR2B-containing NMDA receptors by an autophosphorylation-dependent mechanism. Mol Cell Neurosci 29:139–147

    Article  CAS  PubMed  Google Scholar 

  43. Raveendran R, Devi Suma Priya S, Mayadevi M, Steephan M, Santhoshkumar TR, Cheriyan J, Sanalkumar R, Pradeep KK et al (2009) Phosphorylation status of the NR2B subunit of NMDA receptor regulates its interaction with calcium/calmodulin-dependent protein kinase II. J Neurochem 110:92–105

    Article  CAS  PubMed  Google Scholar 

  44. Barria A, Muller D, Derkach V, Griffith LC, Soderling TR (1997) Regulatory phosphorylation of AMPA-type glutamate receptors by CaM-KII during long-term potentiation. Science 276:2042–2045

    Article  CAS  PubMed  Google Scholar 

  45. Peng S, Zhang Y, Zhang J, Wang H, Ren B (2010) ERK in learning and memory: a review of recent research. Int J Mol Sci 11:222–232

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Carlezon WA Jr, Duman RS, Nestler EJ (2005) The many faces of CREB. Trends Neurosci 28:436–445

    Article  CAS  PubMed  Google Scholar 

  47. Thomas MG, Huganir RL (2004) MAPK cascade signalling and synaptic plasticity. Nat Rev Neurosci 5:173–183

    Article  CAS  PubMed  Google Scholar 

  48. Davis S, Vanhoutte P, Pages C, Caboche J, Laroche S (2000) The MAPK/ERK cascade targets both Elk-1 and cAMP response element-binding protein to control long-term potentiation-dependent gene expression in the dentate gyrus in vivo. J Neurosci 20:4563–4572

    CAS  PubMed  Google Scholar 

  49. Landgraf R, Neumann ID (2004) Vasopressin and oxytocin release within the brain: a dynamic concept of multiple and variable modes of neuropeptide communication. Front Neuroendocrinol 25:150–176

    Article  CAS  PubMed  Google Scholar 

  50. Schaefer A, Sampath SC, Intrator A, Min A, Gertler TS, Surmeier DJ, Tarakhovsky A, Greengard P (2009) Control of cognition and adaptive behavior by the GLP/G9a epigenetic suppressor complex. Neuron 64:678–691

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Babyak M, Blumenthal JA, Herman S, Khatri P, Doraiswamy M, Moore K, Craighead WE, Baldewicz TT et al (2000) Exercise treatment for major depression: maintenance of therapeutic benefit at 10 months. Psychosom Med 62:633–638

    Article  CAS  PubMed  Google Scholar 

  52. Erickson KI, Miller DL, Roecklein KA (2012) The aging hippocampus: interactions between exercise, depression, and BDNF. Neuroscientist 18:82–97

    Article  CAS  PubMed  Google Scholar 

  53. Danielsson L, Noras AM, Waern M, Carlsson J (2013) Exercise in the treatment of major depression: a systematic review grading the quality of evidence. Physiother Theory Pract 29:573–585

    Article  PubMed  Google Scholar 

  54. Kmietowicz Z (2013) Evidence that exercise helps in depression is still weak, finds review. BMJ 13:347

    Google Scholar 

  55. Cooney GM, Dwan K, Greig CA, Lawlor DA, Rimer J, Waugh FR, McMurdo M, Mead GE (2013) Exercise for depression. Cochrane Database Syst Rev 12:9

    Google Scholar 

Download references

Acknowledgments

This research was supported by a grant (2012R1A2A1A03010177) from the Ministry of Science, ICT and Future Planning, Republic of Korea.

Competing Interests

The authors declare that they have no competing financial interests.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Pyung-Lim Han.

Additional information

Tae-Kyung Kim and Jung-Eun Lee contributed equally to this work.

Electronic Supplementary Material

Below is the link to the electronic supplementary material.

Supplemental Table 1

A list of the genes that were upregulated or downregulated in the amygdala after the 2 h × 14 days RST treatment. A list of the genes whose expressions were upregulated or downregulated in the amygdala by more than 1.2-fold after the 2 h × 14 days RST treatment (CON vs. RST), and follow-up unsupervised clusterings of the respective genes in animal groups treated with imipramine or exercise were overlaid with respect to RST (RST vs. RST + IMI; RST vs. RST + EXE) or control (CON vs. RST + IMI; CON vs. RST + EXE). Average fold changes were presented. CON vs. RST, comparison between control and RST group (the genes that were upregulated or downregulated in the amygdala after the 2 h × 14 days RST); RST vs. RST + IMI, comparison between RST and RST + IMI group (the genes that were downregulated or upregulated by imipramine); RST vs. RST + EXE, comparison between RST and RST + EXE group (the genes that were downregulated or upregulated by exercise); CON vs. RST + IMI, comparison between control and RST + IMI group (the genes that were downregulated or upregulated by imipramine compared to control); CON vs. RST + EXE, comparison between control and RST + EXE group (the genes that were downregulated or upregulated by exercise compared to control). (XLSX 143 kb)

Supplemental Table 2

A list of the genes that were upregulated or downregulated in the amygdala after the 2 h × 14 days RST treatment but completely reversed in their expression by exercise. A list of the genes that were upregulated or downregulated in the amygdala by more than 1.2-fold after the 2 h × 14 days RST treatment (CON vs. RST), and then were reversed in their expression in opposite directions beyond the control level after treatment with exercise (E; CON vs. RST + EXE). These groups of the genes correspond to the 3-2 and 4-2 patterns among hypothetical gene expression alterations (G) induced after treatment with exercise. Average fold changes were presented. (XLSX 36 kb)

Supplemental Table 3

A list of the genes that were upregulated or downregulated in the amygdala after the 2 h × 14 days RST treatment, but completely reversed in their expression by imipramine. A list of the genes that were upregulated or downregulated in the amygdala by more than 1.2-fold after the 2 h × 14 days RST treatment (CON vs. RST), and then were reversed in their expression in opposite directions beyond the control level after treatment with imipramine (CON vs. RST + IMI). These groups of the genes correspond to the 3-2 and 4-2 patterns among hypothetical gene expression alterations (G) induced after treatment with imipramine. Average fold changes were presented. (XLSX 20.1 kb)

Supplemental Fig. 1

Real-time PCR analysis for expression levels of HATs, HDACs and HMTs in the amygdala after the 2 h × 14 days RST treatment. (A) Experimental design for treatment with 2 h × 14 days RST and following tissue preparation (arrow). (B) Real-time PCR analysis for expression levels of HATs, HDACs and HMTs in the amygdala after the 2 h × 14 days RST treatment. HAT, histone acetyltransferases (CREB-binding protein(CBP) and p300); HDAC2 and HDAC5, histone deacetyltransferase-2 and deacetyltransferase-5; HMT, histone methyltransferase (G9a and GLP). Data are presented as the mean ± SEM (n = 6 animals for each). Student t test. *p < 0.05 for the difference between indicated group. Primers used were 5′-CGCAACATCACCCATCTG-3′ and 5′-TCATACCAGCATCGGATACT-3′ for G9a, 5′-GACTGAGCAGCGATAATG-3′ and 5′-CAAGGTGTCTCTAGTGTATG-3′for p300, 5′-GGTTGCCTATGCTAAGAAAGT-3′ and 5′-GATGCCTTGCTTATGTAAACG-3′ for CBP, 5′-GGGACAGGCTTGGTTGTTTC-3′ and 5′-GAGCATCAGCAATGGCAAGT-3′ for HDAC2, 5′-TGTCACCGCCAGATGTTTTG-3′ and 5′-TGAGCAGAGCCGAGACACAG-3′ for HDAC5, 5′-CCAAGCAAGAGACCAAGCAG-3′ and 5′-CTTCCTGTGGGCTAGCTCTT-3′ for GLP, 5′-AGAAGGTGGTGAAGCAGGCATC-3′ and 5′-CGAAGGTGGAAGAGTGGGAGTTG-3′ for GAPDH, and 5′-GCTGCCATCTGTTTTACGG-3′ and 5′-TGACTGGTGCCTGATGAACT-3′ for L32. (GIF 32 kb)

High resolution image (TIFF 1332 kb)

Supplemental Fig. 2

Representative amplification plots for ChIP assays quantified by real-time PCR and relative enrichments of G9a and H3K9me2 at the OXT and AVP promoters. (A, B) The promoter regions (P1 and P2) of the OXT(D) and AVP(E) genes used for ChIP analysis were indicated. tss, transcription start site of each gene. (C–F) Representative amplification plots for ChIP assays quantified by real-time PCR. Relative fluorescence levels were plotted over PCR cycle numbers. The Tc value of immunoprecipitated products with normal anti-IgG was observed later than the Tc value of immunoprecipitated products with anti-DimeH3K9 or anti-G9a in all promoter cases examined. These results indicate that ChIPs reacted with DimeH3K9 or G9a antibody were enriched for the OXT and AVP promoters, thus were dissociated from ChIPs produced by normal IgG in the test condition. (G–J) Relative enrichments of G9a and H3K9me2 at the P1 and P2 promoter regions of the OXT (G, H) and AVP (I, J). ChIP reactions were conducted with anti-DimeH3K9, anti-G9a, or normal IgG for each experiment group, and obtained ChIP products in each reaction were quantified for the indicated promoter region by real-time PCR. The data presented were normalized by the input DNA. CON, control; RST, treatment with 2 h × 14 days restraint; RST + IMI, treatment with RST and imipramine; RST + EXE, treatment with RST and exercise. Data are presented as the mean ± SEM (n = 18 animals for each, 3 repeats of ChIP). One-way ANOVA and Newman-Keuls post hoc test. *p < 0.05 and **p < 0.01 for the difference between the control and indicated groups. # p < 0.05 and ## p < 0.01, respectively for the difference between RST and indicated groups. (K–R) Representative amplification plots for DimeH3K or G9a ChIP assays showing the differential enrichments of H3K9me2 (K–N) or G9a (O–R) at the P1 and P2 promoter regions of the OXT (K, L, O, P) and AVP (M, N, Q, R) in different experimental groups. Compared to the control at all promoter regions examined, exercise group was detected earlier than the control, while stress group appeared later than the control. The higher the occupancy of DimeH3K or G9a at each promoter region, the earlier the PCR cycle at which the amplification reaches threshold fluorescence. CON, naïve control; RST, mice treated with 2 h × 14 days restraint; RST + IMI, mice treated with 2 h × 14 days RST, followed by imipramine; RST + EXE, mice treated with 2 h × 14 days RST, followed by exercise (GIF 93 kb)

High resolution image (TIFF 2792 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Kim, TK., Lee, JE., Kim, JE. et al. G9a-Mediated Regulation of OXT and AVP Expression in the Basolateral Amygdala Mediates Stress-Induced Lasting Behavioral Depression and Its Reversal by Exercise. Mol Neurobiol 53, 2843–2856 (2016). https://doi.org/10.1007/s12035-015-9160-z

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s12035-015-9160-z

Keywords

Navigation