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:

Epigenetic basis of opiate suppression of Bdnf gene expression in the ventral tegmental area

Abstract

Brain-derived neurotrophic factor (BDNF) has a crucial role in modulating neural and behavioral plasticity to drugs of abuse. We found a persistent downregulation of exon-specific Bdnf expression in the ventral tegmental area (VTA) in response to chronic opiate exposure, which was mediated by specific epigenetic modifications at the corresponding Bdnf gene promoters. Exposure to chronic morphine increased stalling of RNA polymerase II at these Bdnf promoters in VTA and altered permissive and repressive histone modifications and occupancy of their regulatory proteins at the specific promoters. Furthermore, we found that morphine suppressed binding of phospho-CREB (cAMP response element binding protein) to Bdnf promoters in VTA, which resulted from enrichment of trimethylated H3K27 at the promoters, and that decreased NURR1 (nuclear receptor related-1) expression also contributed to Bdnf repression and associated behavioral plasticity to morphine. Our findings suggest previously unknown epigenetic mechanisms of morphine-induced molecular and behavioral neuroadaptations.

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: Opiate-induced downregulation of Bdnf expression in human, rat and mouse VTA.
Figure 2: Effect of chronic morphine on expression of Bdnf exons and on binding of Pol II to the Bdnf gene in rat VTA.
Figure 3: Morphine-induced histone modifications at Bdnf promoters in rat VTA.
Figure 4: Chronic morphine regulation of CREB binding to Bdnf promoters in VTA.
Figure 5: Epigenetic regulation of Bdnf by NURR1 and its concomitant behavioral effects.

Similar content being viewed by others

References

  1. Lobo, M.K. et al. Cell type–specific loss of BDNF signaling mimics optogenetic control of cocaine reward. Science 330, 385–390 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  2. Pu, L., Liu, Q.S. & Poo, M.M. BDNF-dependent synaptic sensitization in midbrain dopamine neurons after cocaine withdrawal. Nat. Neurosci. 9, 605–607 (2006).

    CAS  PubMed  Google Scholar 

  3. Russo, S.J., Mazei-Robison, M.S., Ables, J.L. & Nestler, E.J. Neurotrophic factors and structural plasticity in addiction. Neuropharmacology 56 (suppl. 1), 73–82 (2009).

    CAS  PubMed  Google Scholar 

  4. Filip, M. et al. Alterations in BDNF and trkB mRNAs following acute or sensitizing cocaine treatments and withdrawal. Brain Res. 1071, 218–225 (2006).

    CAS  PubMed  Google Scholar 

  5. Graham, D.L. et al. Dynamic BDNF activity in nucleus accumbens with cocaine use increases self-administration and relapse. Nat. Neurosci. 10, 1029–1037 (2007).

    CAS  PubMed  Google Scholar 

  6. Grimm, J.W. et al. Time-dependent increases in brain-derived neurotrophic factor protein levels within the mesolimbic dopamine system after withdrawal from cocaine: implications for incubation of cocaine craving. J. Neurosci. 23, 742–747 (2003).

    CAS  PubMed  PubMed Central  Google Scholar 

  7. Graham, D.L. et al. Tropomyosin-related kinase B in the mesolimbic dopamine system: region-specific effects on cocaine reward. Biol. Psychiatry 65, 696–701 (2009).

    CAS  PubMed  Google Scholar 

  8. Hall, F.S., Drgonova, J., Goeb, M. & Uhl, G.R. Reduced behavioral effects of cocaine in heterozygous brain-derived neurotrophic factor (BDNF) knockout mice. Neuropsychopharmacology 28, 1485–1490 (2003).

    CAS  PubMed  Google Scholar 

  9. Horger, B.A. et al. Enhancement of locomotor activity and conditioned reward to cocaine by brain-derived neurotrophic factor. J. Neurosci. 19, 4110–4122 (1999).

    CAS  PubMed  PubMed Central  Google Scholar 

  10. Lu, L., Dempsey, J., Liu, S.Y., Bossert, J.M. & Shaham, Y. A single infusion of brain-derived neurotrophic factor into the ventral tegmental area induces long-lasting potentiation of cocaine seeking after withdrawal. J. Neurosci. 24, 1604–1611 (2004).

    CAS  PubMed  PubMed Central  Google Scholar 

  11. Koo, J.W. et al. BDNF is a negative modulator of morphine action. Science 338, 124–128 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  12. Berhow, M.T. et al. Influence of neurotrophic factors on morphine- and cocaine-induced biochemical changes in the mesolimbic dopamine system. Neuroscience 68, 969–979 (1995).

    CAS  PubMed  Google Scholar 

  13. Russo, S.J. et al. IRS2-Akt pathway in midbrain dopamine neurons regulates behavioral and cellular responses to opiates. Nat. Neurosci. 10, 93–99 (2007).

    CAS  PubMed  Google Scholar 

  14. Sklair-Tavron, L. et al. Chronic morphine induces visible changes in the morphology of mesolimbic dopamine neurons. Proc. Natl. Acad. Sci. USA 93, 11202–11207 (1996).

    CAS  PubMed  PubMed Central  Google Scholar 

  15. Mazei-Robison, M.S. et al. Role for mTOR signaling and neuronal activity in morphine-induced adaptations in ventral tegmental area dopamine neurons. Neuron 72, 977–990 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  16. Aid, T., Kazantseva, A., Piirsoo, M., Palm, K. & Timmusk, T. Mouse and rat BDNF gene structure and expression revisited. J. Neurosci. Res. 85, 525–535 (2007).

    CAS  PubMed  Google Scholar 

  17. Vanderschuren, L.J. et al. Morphine-induced long-term sensitization to the locomotor effects of morphine and amphetamine depends on the temporal pattern of the pretreatment regimen. Psychopharmacology (Berl.) 131, 115–122 (1997).

    CAS  Google Scholar 

  18. Komarnitsky, P., Cho, E.J. & Buratowski, S. Different phosphorylated forms of RNA polymerase II and associated mRNA processing factors during transcription. Genes Dev. 14, 2452–2460 (2000).

    CAS  PubMed  PubMed Central  Google Scholar 

  19. Laherty, C.D. et al. Histone deacetylases associated with the mSin3 corepressor mediate mad transcriptional repression. Cell 89, 349–356 (1997).

    CAS  PubMed  Google Scholar 

  20. Shi, X. et al. ING2 PHD domain links histone H3 lysine 4 methylation to active gene repression. Nature 442, 96–99 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  21. Ythier, D. et al. Sumoylation of ING2 regulates the transcription mediated by Sin3A. Oncogene 29, 5946–5956 (2010).

    CAS  PubMed  Google Scholar 

  22. Czermin, B. et al. Drosophila enhancer of Zeste/ESC complexes have a histone H3 methyltransferase activity that marks chromosomal Polycomb sites. Cell 111, 185–196 (2002).

    CAS  PubMed  Google Scholar 

  23. Müller, J. et al. Histone methyltransferase activity of a Drosophila Polycomb group repressor complex. Cell 111, 197–208 (2002).

    PubMed  Google Scholar 

  24. Simon, J.A. & Kingston, R.E. Mechanisms of polycomb gene silencing: knowns and unknowns. Nat. Rev. Mol. Cell Biol. 10, 697–708 (2009).

    CAS  PubMed  Google Scholar 

  25. Cao, R., Tsukada, Y. & Zhang, Y. Role of Bmi-1 and Ring1A in H2A ubiquitylation and Hox gene silencing. Mol. Cell 20, 845–854 (2005).

    CAS  PubMed  Google Scholar 

  26. Tao, X., West, A.E., Chen, W.G., Corfas, G. & Greenberg, M.E. A calcium-responsive transcription factor, CaRF, that regulates neuronal activity-dependent expression of BDNF. Neuron 33, 383–395 (2002).

    CAS  PubMed  Google Scholar 

  27. Walters, C.L., Kuo, Y.C. & Blendy, J.A. Differential distribution of CREB in the mesolimbic dopamine reward pathway. J. Neurochem. 87, 1237–1244 (2003).

    CAS  PubMed  Google Scholar 

  28. Olson, V.G. et al. Regulation of drug reward by cAMP response element-binding protein: evidence for two functionally distinct subregions of the ventral tegmental area. J. Neurosci. 25, 5553–5562 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  29. Xiong, Y. et al. Polycomb antagonizes p300/CREB-binding protein-associated factor to silence FOXP3 in a Kruppel-like factor-dependent manner. J. Biol. Chem. 287, 34372–34385 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  30. Volpicelli, F. et al. Direct regulation of Pitx3 expression by Nurr1 in culture and in developing mouse midbrain. PLoS ONE 7, e30661 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  31. Volpicelli, F. et al. Bdnf gene is a downstream target of Nurr1 transcription factor in rat midbrain neurons in vitro. J. Neurochem. 102, 441–453 (2007).

    CAS  PubMed  Google Scholar 

  32. Barneda-Zahonero, B. et al. Nurr1 protein is required for N-methyl-d-aspartic acid (NMDA) receptor-mediated neuronal survival. J. Biol. Chem. 287, 11351–11362 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  33. Kadkhodaei, B. et al. Nurr1 is required for maintenance of maturing and adult midbrain dopamine neurons. J. Neurosci. 29, 15923–15932 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  34. McEvoy, A.N. et al. Activation of nuclear orphan receptor NURR1 transcription by NF-kappa B and cyclic adenosine 5′-monophosphate response element-binding protein in rheumatoid arthritis synovial tissue. J. Immunol. 168, 2979–2987 (2002).

    PubMed  Google Scholar 

  35. Chu, N.N. et al. Peripheral electrical stimulation reversed the cell size reduction and increased BDNF level in the ventral tegmental area in chronic morphine-treated rats. Brain Res. 1182, 90–98 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  36. Vargas-Perez, H. et al. Ventral tegmental area BDNF induces an opiate-dependent-like reward state in naive rats. Science 324, 1732–1734 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  37. Fujii, S., Ito, K., Ito, Y. & Ochiai, A. Enhancer of zeste homologue 2 (EZH2) down-regulates RUNX3 by increasing histone H3 methylation. J. Biol. Chem. 283, 17324–17332 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  38. van der Vlag, J. & Otte, A.P. Transcriptional repression mediated by the human polycomb-group protein EED involves histone deacetylation. Nat. Genet. 23, 474–478 (1999).

    CAS  PubMed  Google Scholar 

  39. Milne, T.A. et al. MLL targets SET domain methyltransferase activity to Hox gene promoters. Mol. Cell 10, 1107–1117 (2002).

    CAS  PubMed  Google Scholar 

  40. Xia, Z.B., Anderson, M., Diaz, M.O. & Zeleznik-Le, N.J. MLL repression domain interacts with histone deacetylases, the polycomb group proteins HPC2 and BMI-1, and the co-repressor C terminal–binding protein. Proc. Natl. Acad. Sci. USA 100, 8342–8347 (2003).

    CAS  PubMed  PubMed Central  Google Scholar 

  41. Rios, M. et al. Conditional deletion of brain-derived neurotrophic factor in the postnatal brain leads to obesity and hyperactivity. Mol. Endocrinol. 15, 1748–1757 (2001).

    CAS  PubMed  Google Scholar 

  42. Covington, H.E. III et al. A role for repressive histone methylation in cocaine-induced vulnerability to stress. Neuron 71, 656–670 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  43. Anderson, S.A. et al. Impaired periamygdaloid-cortex prodynorphin is characteristic of opiate addiction and depression. J. Clin. Invest. 123, 5334–5341 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  44. 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 

  45. Dietz, D.M. et al. Rac1 is essential in cocaine-induced structural plasticity of nucleus accumbens neurons. Nat. Neurosci. 15, 891–896 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  46. Liu, Q.R. et al. Rodent BDNF genes, novel promoters, novel splice variants, and regulation by cocaine. Brain Res. 1067, 1–12 (2006).

    CAS  PubMed  Google Scholar 

  47. Livak, K.J. & Schmittgen, T.D. Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) Method. Methods 25, 402–408 (2001).

    CAS  PubMed  Google Scholar 

  48. Schmidt, H.D. et al. Increased brain-derived neurotrophic factor (BDNF) expression in the ventral tegmental area during cocaine abstinence is associated with increased histone acetylation at BDNF exon I-containing promoters. J. Neurochem. 120, 202–209 (2012).

    CAS  PubMed  Google Scholar 

  49. Xu, Y.X. & Manley, J.L. Pin1 modulates RNA polymerase II activity during the transcription cycle. Genes Dev. 21, 2950–2962 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  50. Sandoval, J. et al. RNAPol-ChIP: a novel application of chromatin immunoprecipitation to the analysis of real-time gene transcription. Nucleic Acids Res. 32, e88 (2004).

    PubMed  PubMed Central  Google Scholar 

  51. Egloff, S., Al-Rawaf, H., O'Reilly, D. & Murphy, S. Chromatin structure is implicated in “late” elongation checkpoints on the U2 snRNA and beta-actin genes. Mol. Cell. Biol. 29, 4002–4013 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  52. Khobta, A., Anderhub, S., Kitsera, N. & Epe, B. Gene silencing induced by oxidative DNA base damage: association with local decrease of histone H4 acetylation in the promoter region. Nucleic Acids Res. 38, 4285–4295 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  53. Weishaupt, H. & Attema, J.L. A method to study the epigenetic chromatin states of rare hematopoietic stem and progenitor cells: MiniChIP-Chip. Biol. Proced. Online 12, 1–17 (2010).

    PubMed  PubMed Central  Google Scholar 

  54. Nitzsche, A., Steinhausser, C., Mucke, K., Paulus, C. & Nevels, M. Histone H3 lysine 4 methylation marks postreplicative human cytomegalovirus chromatin. J. Virol. 86, 9817–9827 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  55. Gilfillan, G.D. et al. Limitations and possibilities of low cell number ChIP-seq. BMC Genomics 13, 645 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  56. Marchesi, I., Fiorentino, F.P., Rizzolio, F., Giordano, A. & Bagella, L. The ablation of EZH2 uncovers its crucial role in rhabdomyosarcoma formation. Cell Cycle 11, 3828–3836 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  57. Lee, T.I. et al. Control of developmental regulators by Polycomb in human embryonic stem cells. Cell 125, 301–313 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  58. Schaefer, A. et al. Control of cognition and adaptive behavior by the GLP/G9a epigenetic suppressor complex. Neuron 64, 678–691 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  59. Ren, G. et al. Polycomb protein EZH2 regulates tumor invasion via the transcriptional repression of the metastasis suppressor RKIP in breast and prostate cancer. Cancer Res. 72, 3091–3104 (2012).

    CAS  PubMed  Google Scholar 

  60. Rotman, N., Guex, N., Gouranton, E. & Wahli, W. PPARbeta interprets a chromatin signature of pluripotency to promote embryonic differentiation at gastrulation. PLoS ONE 8, e83300 (2013).

    PubMed  PubMed Central  Google Scholar 

  61. Basu, A., Wilkinson, F.H., Colavita, K., Fennelly, C. & Atchison, M.L. YY1 DNA binding and interaction with YAF2 is essential for Polycomb recruitment. Nucleic Acids Res. 42, 2208–2223 (2014).

    CAS  PubMed  Google Scholar 

  62. Richly, H. et al. Transcriptional activation of polycomb-repressed genes by ZRF1. Nature 468, 1124–1128 (2010).

    CAS  PubMed  Google Scholar 

  63. Mao, L. et al. Cyclin E1 is a common target of BMI1 and MYCN and a prognostic marker for neuroblastoma progression. Oncogene 31, 3785–3795 (2012).

    CAS  PubMed  Google Scholar 

  64. Ellison-Zelski, S.J., Solodin, N.M. & Alarid, E.T. Repression of ESR1 through actions of estrogen receptor alpha and Sin3A at the proximal promoter. Mol. Cell. Biol. 29, 4949–4958 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  65. Pellegrini, M. et al. Expression profile of CREB knockdown in myeloid leukemia cells. BMC Cancer 8, 264 (2008).

    PubMed  PubMed Central  Google Scholar 

  66. Sakurada, K., Ohshima-Sakurada, M., Palmer, T.D. & Gage, F.H. Nurr1, an orphan nuclear receptor, is a transcriptional activator of endogenous tyrosine hydroxylase in neural progenitor cells derived from the adult brain. Development 126, 4017–4026 (1999).

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank T. Abel (University of Pennsylvania) for helpful discussions. This work was supported by grants from the National Institute on Drug Abuse (E.J.N. and M.S.M.-R.).

Author information

Authors and Affiliations

Authors

Contributions

J.W.K., M.S.M.-R., S.J.R., Y.L.H. and E.J.N. designed the study. J.W.K., M.S.M.-R., Q.L., G.E., K.M.B., D.N.A., D.F., J.F., H.S., K.N.S., D.M.D.-W., E.R., C.J.P., D.W., R.C.B., M.E.C., S.A.R.A., B.L., G.E.H., H.B., B.C., A.J.R., V.F.V., C.D., Z.L., E.M., M.K.L. and D.M.D. performed the experiments. J.W.K. and R.L.N. generated viral vectors. J.W.K. and Q.L. analyzed data. J.W.K. and E.J.N. wrote the paper.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 Validation of opiate treatment regimens used in this study.

(a) Scatter plot of Bdnf exon IX mRNA levels in VTA of human heroin addicts compared to control subjects. Student’s t-test, *p < 0.05 compared to control subjects, n = 5,9. (b) Acquisition of heroin self administration. Rats robustly self-administered heroin (30 μg/kg/infusion on a designated “active” lever), but not saline. After 10 days of chronic heroin self-administration, animals were killed 24 hrs after last drug use. Repeated measures two-way ANOVA (drug effect: p < 0.001; day effect: p < 0.001; drug×day effect: p < 0.001), Fisher’s post hoc tests, *p < 0.05, **p < 0.01, ***p < 0.001 compared with heroin inactive group; #p < 0.05, ###p < 0.001 compared with saline active group, n = 9,14. (c) Locomotor activity in a standard morphine treatment regimen where rats are injected daily with saline or morphine (5 mg/kg, IP) for 14 days and, two weeks later, are given a challenge dose of saline or morphine. S/S, saline for 14 days, saline challenge; S/M, saline for 14 days, morphine challenge; M/S, morphine for 14 days, saline challenge; and M/M, morphine for 14 days, morphine challenge. Locomotor assays confirmed that this morphine treatment regimen produced the expected behavioral sensitization18. For day 0 to 14, repeated measures two-way ANOVA (drug effect: p = n.s., day effect: p < 0.001; drug×day effect: p < 0.001), Fisher’s post hoc tests, ***p < 0.001 compared with S/S control group, n = 18. On day 28, morphine challenge 14 days after chronic morphine (M/M) elicited hyperactivity compared to S/S, S/M, and M/S groups. Acute morphine exposure 14 days after chronic saline (S/M) showed lower levels of locomotor activity compared to saline controls (S/S) as observed in morphine injected rats on day 1. One-way ANOVA (p < 0.001), Fisher’s post hoc tests, **p < 0.01, ***p < 0.001 compared with S/S group; ###p < 0.001 compared with M/M group, n = 9. (d) Subchronic morphine exposure (15 mg/kg, IP) during 3 day-CPP training induced high levels of CPP scores compared to saline-injected controls. t-test, ***p < 0.001, n = 12. (e) Effect of chronic morphine on gene expression of Bdnf exons III, V, VII, and VIII in rat VTA. qPCR showed that Bdnf transcript levels of exons III (Ct = 33.393 ± 0.295, n = 8), V (Ct = not detectable, n = 8), VII (Ct = 34.776 ± 0.226, n = 8), and VIII (Ct = 34.363 ± 0.392, n = 7,8) are rarely, if ever, detected, and when detected are not significantly altered by morphine. Student's t-tests, t14= 0.573 for exons III; t14= 0.0163 for exons VII, all p’s = n.s.; Mann-Whitney U test, U = 26 for exons VIII, p = n.s. Individual n, p values and degrees of freedom are available in the Supplementary Methods Checklist.

Supplementary Figure 2 Chronic morphine-induced modifications of histones and histone-regulatory proteins at Bdnf gene promoters in rat VTA.

(a-g) Two-way ANOVA followed by Fisher’s post hoc tests showed that chronic morphine (as in Fig. 1c) alters levels of acH4 at Bdnf promoter 4 (-p4) (b) and of H3K27me3 at Bdnf-p2 (f) in VTA. However, there was no morphine-induced alteration in acH3 (a, drug effect: p = n.s.; region effect: p = n.s.; drug×region effect: p = n.s., n = 3,4), H3K4me3 (c, drug effect: p = n.s.; region effect: p = n.s.; drug×region effect: p = n.s., n = 4,5), H3K9me2 (d, drug effect: p = n.s.; region effect: p = n.s.; drug×region effect: p = n.s., n = 4), H3K9me3 (e, drug effect: p = n.s.; region effect: p = n.s.; drug×region effect: p = n.s., n = 5), and H3K36me3 (g, drug effect: p = n.s.; region effect: p = n.s.; drug×region effect: p = n.s., n = 6,5). Additional post hoc analyses with Student’s t-tests showed that chronic morphine changes levels of acH3 (a) and of H3K4me3 (c) at Bdnf-p2. #p < 0.05 compared to S/S controls. (h-p) Chronic morphine also changes occupancy of proteins such as mSIN3a (h), ING2 (i), MLL1 (j), SUZ12 (l), EZH2 (m), RING1A (n), and BMI1 (p) primarily at Bdnf-p2 in VTA (see Fig. 3). However, there was no morphine-induced alteration in G9a (k) and RING1B (o). Fisher’s post hoc tests, *p < 0.05, **p < 0.01, ***p < 0.001 compared with S/S control group. (q) Summary of epigenetic alterations at the Bdnf genes in VTA by chronic morphine. Green thick lines indicate the relative position of amplicons generated by primers used to quantify immunoprecipitated chromatin-DNA. Exons are represented as boxes and the introns as lines. Numbers of the exons are indicated in roman numerals. The positions of the CREB binding sites at Bdnf promoter regions are indicated as red circles. Filled red arrows indicate significant increases; filled blue arrows indicate significant decreases in response to chronic morphine. Individual n, p values and degrees of freedom are available in the Supplementary Methods Checklist.

Supplementary Figure 3 Validation of HSV-EZH2 and of LV-shRNA-EZH2.

(a) Infusion of HSV-EZH2 into VTA of wildtype c57BL/6 mice increased Ezh2, but not Th and Gria1 mRNA expression compared to HSV-GFP controls. Student’s t-tests, t16= 3.044 for Ezh2, ** p < 0.01, n = 9. Mann-Whitney U tests, U = 25 for Th, U = 30 for Gria1, all p’s = n.s., all n’s = 9. (b-d) Validation of LV-shRNA-EZH2. (b) Representative images (scale bar, 25 μm) demonstrating localized LV-mediated shRNA-EZH2 expression (green) in TH-positive (red) cells (white arrows) in rat VTA. (c) Infusion of LV-shRNA-EZH2 into VTA of Sprague Dawley rats decreased Ezh2 mRNA levels compared to LV-GFP-scramble controls. Student’s t-test, t20= 2.193, *p < 0.05, n = 11. (d) Morphine-treated rats that were injected with LV-shRNA-scramble lower Bdnf mRNA levels of compared to saline-treated rats injected with LV-shRNA-scramble. LV-mediated repression of EZH2 in VTA blocked the morphine-induced Bdnf mRNA reduction (one-way ANOVA, F2,13 = 5.003, p < 0.05, n = 5,4,5). Fisher’s post hoc tests, *p < 0.05 compared to saline+LV-shRNA-scramble; #p < 0.05 compared to Morphine+LV-shRNA-scramble.

Supplementary Figure 4 Western blot analysis for validation of HSV-EZH2, HSV-CREB, and HSV-NURR1 in rat VTA.

The viruses were infused into VTA, and protein levels of EZH2 (a), CREB (b), and NURR1(c) were measured in VTA and in anatomical controls, substantia nigra (SN) and red nucleus (RN). The immunoblots show viral-mediated overexpression of the respective protein in VTA (a, HSV-EZH2, Student’s t-test, *p < 0.05 compared to HSV-GFP, n = 7; b, HSV-CREB, one-way ANOVA, F2,14 = 3.764, p < 0.05, n = 5,6,6; c, HSV-NURR1, one-way ANOVA, F2,14 = 5.092, p < 0.05, n = 5,6,6), but not in other brain areas. Fisher’s post hoc tests, *p < 0.05, ** p < 0.01 compared to HSV-TMT controls. Each protein’s expression was normalized to β-tubulin levels, which were not affected by the treatments. Individual n, p values, and degrees of freedom are available in the Supplementary Methods Checklist. For full-length western blots, see in Supplementary Figure 10.

Supplementary Figure 5 Validation of HSV-CREB and HSV-Cre.

(a) Infusion of HSV-CREB into VTA of wildtype c57BL/6 mice increased Creb1 mRNA expression compared to HSV-TMT (tdTomato) controls. Student’s t-test, t16= 2.208, *p < 0.05, n = 9. (b) In contrast, infusion of HSV-Cre into the VTA of floxed CREB mice decreased Creb1 mRNA expression. t-test, t18= 4.344, ***p < 0.001, n = 10. (c,d) Intra-VTA infusion of HSV-CREB up-regulated both total- (c, t-test, t8= 3.813, **p < 0.01, n = 5) and phospho-CREB binding to Nurr1 promoter (d, Mann-Whitney U test, U = 2, *p < 0.05, n = 5) compared to HSV-TMT controls.

Supplementary Figure 6 Effect of opiates on levels of phospho-/total-CREB binding and H3K27me3 at Bdnf promoters or at promoters of other CREB targets, Th and Gria1 in rat VTA.

(a) In contrast to phospho-CREB binding to Bdnf promoters, phospho-CREB binding to Th and Gria1 promoters was increased in morphine-treated rats (Student’s t-tests, for Th, t14= 2.165, n = 9,7; for Gria1, t7= 2.489, n = 5,4, *p < 0.05). (b,c) However, chronic morphine exposure had no effect on levels of total-CREB binding (for Th, t-test, t7= 0.380; for Gria1, Mann-Whitney U test, U = 9; all p’s = n.s., all n’s = 5,4) and H3K27me3 (for Th, t-test, t8= 0.0297; for Gria1, Mann-Whitney U test, U = 7; all p’s = n.s., all n’s = 5) at Th and Gria1 promoters. (d) Self-administered heroin significantly reduced phospho-CREB binding to all Bdnf promoters (two-way ANOVA, drug effect: F1,32 = 32.004, p < 0.001; region effect: F3,32 = 0.132, p = n.s.; drug×region effect: F3,32 = 0.132, p = n.s., n = 5). (e,f) In contrast, heroin self-administration increased total-CREB binding to Bdnf-p2 and -p6 (e, two-way ANOVA, drug effect: F1,20 = 5.683, p < 0.05; region effect: F3,20 = 2.175, p = n.s.; drug×region effect: F3,20 = 2.175, p = n.s., n = 3,4) and H3K27me3 levels (f, two-way ANOVA, drug effect: F1,36 = 19.593, p < 0.001; region effect: F3,36 = 0.542, p = n.s.; drug×region effect: F3,36 = 0.542, p = n.s., n = 5,6) at Bdnf-p2 and -p4. Fisher’s post hoc tests, *p < 0.05, **p < 0.01 compared with each control group. Additional post hoc analyses with Student’s t-tests showed that self-administered heroin induced H3K27me3 at Bdnf-p1,-p2, and -p4 (Bdnf-p1, t9 = 2.789; Bdnf-p2, t9 = 2.495; Bdnf-p4, t9 = 2.549; Bdnf-p6, t9 = 1.262, #p < 0.05).

Supplementary Figure 7 Validation of HSV-NURR1 and NURR1 ChIP antibody.

(a) Infusion of HSV-NURR1 into rat VTA increases Nurr1 mRNA expression in this region. Student’s t-test, t17= 2.166, *p < 0.05, n = 9,10. (b) Effect of Nurr1 overexpression on morphine re-exposure induced hyper-locomotor activity. Repeated injection of morphine (5 mg/kg, IP) for 14 days induced high levels of locomotion (repeated measures two-way ANOVA, drug effect: F1,36 = 7.087, p < 0.05; day effect: F1,36 = 54.272, p < 0.001; drug×day effect: F1,36 = 13.080, p < 0.001, n = 11,27). Fisher’s post hoc tests, ***p < 0.001 compared with saline group. 10 days later, HSV-NURR1 or HSV-TMT were infused into VTA. Four days after the intra-VTA infusion of HSVs (day 28), re-exposure of HSV-TMT infused group (MM-TMT) to morphine challenge dose (5 mg/kg, IP) induced hyper-locomotion compared to saline exposed HSV-TMT infused group (MS-TMT). Importantly, intra-VTA infusion of HSV-NURR1 blocked this morphine re-exposure induced hyper-locomotion (see Fig. 5g,h). Nurr1 overexpression in saline-treated group (i.e., without morphine) had no effect on locomotor activity (Inset). One-way ANOVA, F4,37 = 3.031, p < 0.05, n = 5-10. (c-e) In order to verify the specificity of NURR1 antibody used for ChIP experiments, western blotting was performed with (c) anti-NURR1, (d) anti-NOR1, or (e) anti-NUR77. The analysis shows an enrichment of NURR1 in the anti-NURR1-ChIP’ed samples but not in the corresponding input samples. Student’s t-test, t4= 12.297, ***p < 0.001 compared to input, n = 3 (10 rat VTA punches pooled per sample). No immunoreactivity was detected by anti-NOR1, anti-NUR77, and anti-β-tubulin in the immunoprecipitated samples with anti-NURR1 (all n’s = 3). For full-length western blots, see in Supplementary Figure 10.

Supplementary Figure 8 Schematic model of the complex epigenetic mechanisms underlying suppression of the Bdnf gene in VTA by chronic morphine.

Chronic morphine stalls transcriptional activity of polymerase II (Pol II) at Bdnf by increasing phospho-Ser5-Pol II (Ser5-p) at promoter regions, while reducing phospho-Ser2-Pol II (Ser2-p) at Bdnf exons. Chronic morphine also decreases permissive histone acetylation (e.g., AcH3), and increases repressive histone methylation (H3K27me3), at Bdnf-p2, the latter part of a repressive complex (PRC2, Polycomb repressive complex 2) which includes increased binding of SUZ12 and EZH2 (an H3K27 methyltransferase). As well, chronic morphine paradoxically increases levels of a permissive histone mark (H3K4me3) and its methytransferase (MLL1) at Bdnf-p2. However, these paradoxical increases are associated with morphine induction of ING2 and mSIN3a binding to Bdnf-p2. ING2, a subunit of the repressive mSIN3a-HDAC complex, binds with high specificity to H3K4me3 at particular genes and thereby represses their transcription. In addition, the morphine-induced repression of Bdnf involves reduced binding of two key transcription factors, phospho-CREB (cAMP response element binding protein) and NURR1 (nuclear receptor related 1), which normally induce Bdnf gene expression. In particular, phospho-CREB binding, which is increased at other target genes in VTA by chronic morphine (not shown), is antagonized at Bdnf promoters by morphine-induced increases in H3K27me3. These studies, which provide one of the most comprehensive analyses of epigenetic regulation of a target gene in the brain yet available, reveal novel epigenetic mechanisms for Bdnf down-regulation in VTA in response to chronic morphine, adaptations that contribute to enhanced behavioral responses to the drug.

Supplementary Figure 9 Validation of antibodies used for qChIP experiments.

qPCR amplification was performed with primer sets that were designed based on previous studies validating ChIP antibodies in other systems or on validation information provided by manufacturers (for details, see Supplementary Table 4). Levels of immunoprecipitated DNA are presented as a percentage of input DNA. (a-c) Pol II antibodies. One-way ANOVA, p < 0.05 (a, Total Pol II; c, Ser2-p-Pol II); p < 0.01 (b, Ser5-p-Pol II). Fisher’s post hoc tests, *p < 0.05, **p < 0.01, ***p < 0.001 compared to Gapdh; #p < 0.05, ##p < 0.01, ###p < 0.001 compared to Actb-1, n = 3. (d-j) Antibodies for histone markers. One-way ANOVA, all p’s < 0.001 (d, acH3; f, H3K4me3; g, H3K9me2; h, H3K9me3; i, H3K27me3; j, H3K36me3); p < 0.05 (e, acH4). Fisher’s post hoc tests, *p < 0.05, **p < 0.01, ***p < 0.001 compared to Gapdh; #p < 0.05, ##p < 0.01, ###p < 0.001 compared to Rpl30, n = 2-6. (k,l) HDAC-related proteins. One-way ANOVA, all p’s < 0.05. Fisher’s post hoc tests, *p < 0.05, **p < 0.01 compared to Esr1; #p < 0.05 compared to Rpl30, n = 3-4. (m) MLL1. One-way ANOVA, p < 0.05. Fisher’s post hoc tests, *p < 0.05, **p < 0.01 compared to Esr1, n = 4. (n) G9a. Student’s t-test, *p < 0.05 compared to Rpl30, n = 4,3. (o-s) Polycomb group proteins. t-tests, *p < 0.05 compared to Gapdh (o, SUZ12; p, EZH2); one-way ANOVA, p < 0.05 (q, RING1A); p < 0.001 (r, RING1B); p < 0.01 (s, BMI1). Fisher’s post hoc tests, *p < 0.05, **p < 0.01, ***p < 0.001 compared to Gapdh; ###p < 0.001, compared to Myod1, n = 3-4. (t,u) CREB. t-test, *p < 0.05 compared to Nr4a3. n = 5-6. (v) NURR1. One-way ANOVA, p < 0.01. Fisher’s post hoc tests, *p < 0.05, **p < 0.01, ***p < 0.001 compared to Afm; #p < 0.05, ##p < 0.01 compared to Myod1, n = 2-3. Individual n, p values and degrees of freedom from all ChIP data analyses are available in the Supplementary Methods Checklist.

Supplementary Figure 10 Full-length images of blots from the Supplementary Figures.

(a-c) Immunoblots of all individuals from Supplementary Figure 4. and (d,e) from Supplementary Figure 7. #One blot is excluded from analysis due to highly variable expression of the internal control (c, β-tubulin).

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–10 and Supplementary Tables 1–4 (PDF 2135 kb)

Supplementary Methods Checklist (PDF 661 kb)

Supplementary Table 5

Summary of experimental design. (XLSX 16 kb)

Source data

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Koo, J., Mazei-Robison, M., LaPlant, Q. et al. Epigenetic basis of opiate suppression of Bdnf gene expression in the ventral tegmental area. Nat Neurosci 18, 415–422 (2015). https://doi.org/10.1038/nn.3932

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nn.3932

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