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:

G9a is essential for epigenetic silencing of K+ channel genes in acute-to-chronic pain transition

Abstract

Neuropathic pain is a debilitating clinical problem and difficult to treat. Nerve injury causes a long-lasting reduction in K+ channel expression in the dorsal root ganglion (DRG), but little is known about the epigenetic mechanisms involved. We found that nerve injury increased dimethylation of Lys9 on histone H3 (H3K9me2) at Kcna4, Kcnd2, Kcnq2 and Kcnma1 promoters but did not affect levels of DNA methylation on these genes in DRGs. Nerve injury increased activity of euchromatic histone-lysine N-methyltransferase-2 (G9a), histone deacetylases and enhancer of zeste homolog-2 (EZH2), but only G9a inhibition consistently restored K+ channel expression. Selective knockout of the gene encoding G9a in DRG neurons completely blocked K+ channel silencing and chronic pain development after nerve injury. Remarkably, RNA sequencing analysis revealed that G9a inhibition not only reactivated 40 of 42 silenced genes associated with K+ channels but also normalized 638 genes down- or upregulated by nerve injury. Thus G9a has a dominant function in transcriptional repression of K+ channels and in acute-to-chronic pain transition after nerve injury.

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: Nerve injury reduces expression levels of Kcna4, Kcnd2, Kcnq2 and Kcnma1 and increases expression and activity of HDACs, G9a and EZH2 in the DRG.
Figure 2: Cellular distribution of HDACs, G9a and EZH2 in control and injured DRGs.
Figure 3: Nerve injury increases H3K9me2 levels at K+ channel promoters but does not change DNA methylation status at their promoter regions in the DRG.
Figure 4: Inhibition of G9a activity normalizes K+ channel expression in the DRG diminished by nerve injury.
Figure 5: Inhibition of G9a activity restores Kv channel currents in DRG neurons reduced by nerve injury.
Figure 6: Differential effects of inhibition of G9a, HDACs and EZH2 on pain hypersensitivity induced by nerve injury.
Figure 7: Deletion of Ehmt2 in DRG neurons prevents nerve injury–induced pain hypersensitivity and reduction in K+ channel expression in the DRG.
Figure 8: G9a inhibition normalizes the expression of genes encoding K+ channels and a subset of other genes modified by nerve injury.

Similar content being viewed by others

Accession codes

Primary accessions

Gene Expression Omnibus

References

  1. Amir, R., Michaelis, M. & Devor, M. Burst discharge in primary sensory neurons: triggered by subthreshold oscillations, maintained by depolarizing afterpotentials. J. Neurosci. 22, 1187–1198 (2002).

    CAS  PubMed  PubMed Central  Google Scholar 

  2. Campbell, J.N., Raja, S.N., Meyer, R.A. & Mackinnon, S.E. Myelinated afferents signal the hyperalgesia associated with nerve injury. Pain 32, 89–94 (1988).

    CAS  PubMed  Google Scholar 

  3. Wang, H. et al. Chronic neuropathic pain is accompanied by global changes in gene expression and shares pathobiology with neurodegenerative diseases. Neuroscience 114, 529–546 (2002).

    CAS  PubMed  Google Scholar 

  4. Xiao, H.S. et al. Identification of gene expression profile of dorsal root ganglion in the rat peripheral axotomy model of neuropathic pain. Proc. Natl. Acad. Sci. USA 99, 8360–8365 (2002).

    CAS  PubMed  PubMed Central  Google Scholar 

  5. Cao, X.H., Byun, H.S., Chen, S.R., Cai, Y.Q. & Pan, H.L. Reduction in voltage-gated K+ channel activity in primary sensory neurons in painful diabetic neuropathy: role of brain-derived neurotrophic factor. J. Neurochem. 114, 1460–1475 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  6. Chen, S.R., Cai, Y.Q. & Pan, H.L. Plasticity and emerging role of BKCa channels in nociceptive control in neuropathic pain. J. Neurochem. 110, 352–362 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  7. Rasband, M.N. et al. Distinct potassium channels on pain-sensing neurons. Proc. Natl. Acad. Sci. USA 98, 13373–13378 (2001).

    CAS  PubMed  PubMed Central  Google Scholar 

  8. Rose, K. et al. Transcriptional repression of the M channel subunit Kv7.2 in chronic nerve injury. Pain 152, 742–754 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  9. Mucha, M. et al. Transcriptional control of KCNQ channel genes and the regulation of neuronal excitability. J. Neurosci. 30, 13235–13245 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  10. Vydyanathan, A., Wu, Z.Z., Chen, S.R. & Pan, H.L. A-type voltage-gated K+ currents influence firing properties of isolectin B4-positive but not isolectin B4-negative primary sensory neurons. J. Neurophysiol. 93, 3401–3409 (2005).

    CAS  PubMed  Google Scholar 

  11. Cao, X.H., Chen, S.R., Li, L. & Pan, H.L. Nerve injury increases brain-derived neurotrophic factor levels to suppress BK channel activity in primary sensory neurons. J. Neurochem. 121, 944–953 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  12. Chien, L.Y., Cheng, J.K., Chu, D., Cheng, C.F. & Tsaur, M.L. Reduced expression of A-type potassium channels in primary sensory neurons induces mechanical hypersensitivity. J. Neurosci. 27, 9855–9865 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  13. Delmas, P. & Brown, D.A. Pathways modulating neural KCNQ/M (Kv7) potassium channels. Nat. Rev. Neurosci. 6, 850–862 (2005).

    CAS  PubMed  Google Scholar 

  14. Marrion, N.V. & Tavalin, S.J. Selective activation of Ca2+-activated K+ channels by co-localized Ca2+ channels in hippocampal neurons. Nature 395, 900–905 (1998).

    CAS  PubMed  Google Scholar 

  15. Sah, P. & Faber, E.S. Channels underlying neuronal calcium-activated potassium currents. Prog. Neurobiol. 66, 345–353 (2002).

    CAS  PubMed  Google Scholar 

  16. Riccio, A. Dynamic epigenetic regulation in neurons: enzymes, stimuli and signaling pathways. Nat. Neurosci. 13, 1330–1337 (2010).

    CAS  PubMed  Google Scholar 

  17. Guan, J.S. et al. HDAC2 negatively regulates memory formation and synaptic plasticity. Nature 459, 55–60 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  18. Kouzarides, T. Chromatin modifications and their function. Cell 128, 693–705 (2007).

    CAS  PubMed  Google Scholar 

  19. Ronan, J.L., Wu, W. & Crabtree, G.R. From neural development to cognition: unexpected roles for chromatin. Nat. Rev. Genet. 14, 347–359 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  20. Everill, B. & Kocsis, J.D. Reduction in potassium currents in identified cutaneous afferent dorsal root ganglion neurons after axotomy. J. Neurophysiol. 82, 700–708 (1999).

    CAS  PubMed  Google Scholar 

  21. Passmore, G.M. et al. KCNQ/M currents in sensory neurons: significance for pain therapy. J. Neurosci. 23, 7227–7236 (2003).

    CAS  PubMed  PubMed Central  Google Scholar 

  22. Kim, S.H. & Chung, J.M. An experimental model for peripheral neuropathy produced by segmental spinal nerve ligation in the rat. Pain 50, 355–363 (1992).

    CAS  Google Scholar 

  23. Broide, R.S. et al. Distribution of histone deacetylases 1–11 in the rat brain. J. Mol. Neurosci. 31, 47–58 (2007).

    CAS  PubMed  Google Scholar 

  24. Rivera, C. et al. The K+/Cl- co-transporter KCC2 renders GABA hyperpolarizing during neuronal maturation. Nature 397, 251–255 (1999).

    CAS  PubMed  Google Scholar 

  25. Vedadi, M. et al. A chemical probe selectively inhibits G9a and GLP methyltransferase activity in cells. Nat. Chem. Biol. 7, 566–574 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  26. Béguelin, W. et al. EZH2 is required for germinal center formation and somatic EZH2 mutations promote lymphoid transformation. Cancer Cell 23, 677–692 (2013).

    PubMed  PubMed Central  Google Scholar 

  27. Dokmanovic, M., Clarke, C. & Marks, P.A. Histone deacetylase inhibitors: overview and perspectives. Mol. Cancer Res. 5, 981–989 (2007).

    CAS  PubMed  Google Scholar 

  28. Cai, Y.Q. et al. Role of M2, M3, and M4 muscarinic receptor subtypes in the spinal cholinergic control of nociception revealed using siRNA in rats. J. Neurochem. 111, 1000–1010 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  29. Luo, M.C. et al. An efficient intrathecal delivery of small interfering RNA to the spinal cord and peripheral neurons. Mol. Pain 1, 29 (2005).

    PubMed  PubMed Central  Google Scholar 

  30. Gupta-Agarwal, S. et al. G9a/GLP histone lysine dimethyltransferase complex activity in the hippocampus and the entorhinal cortex is required for gene activation and silencing during memory consolidation. J. Neurosci. 32, 5440–5453 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  31. Zhou, X. et al. Deletion of PIK3C3/Vps34 in sensory neurons causes rapid neurodegeneration by disrupting the endosomal but not the autophagic pathway. Proc. Natl. Acad. Sci. USA 107, 9424–9429 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  32. Mortazavi, A., Williams, B.A., McCue, K., Schaeffer, L. & Wold, B. Mapping and quantifying mammalian transcriptomes by RNA-Seq. Nat. Methods 5, 621–628 (2008).

    Article  CAS  PubMed  Google Scholar 

  33. Kim, D.S., Choi, J.O., Rim, H.D. & Cho, H.J. Downregulation of voltage-gated potassium channel alpha gene expression in dorsal root ganglia following chronic constriction injury of the rat sciatic nerve. Brain Res. Mol. Brain Res. 105, 146–152 (2002).

    CAS  PubMed  Google Scholar 

  34. Zhao, X. et al. A long noncoding RNA contributes to neuropathic pain by silencing Kcna2 in primary afferent neurons. Nat. Neurosci. 16, 1024–1031 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  35. Tsantoulas, C. et al. Sensory neuron downregulation of the Kv9.1 potassium channel subunit mediates neuropathic pain following nerve injury. J. Neurosci. 32, 17502–17513 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  36. Fan, L. et al. Impaired neuropathic pain and preserved acute pain in rats overexpressing voltage-gated potassium channel subunit Kv1.2 in primary afferent neurons. Mol. Pain 10, 8 (2014).

    PubMed  PubMed Central  Google Scholar 

  37. Ma, C., Rosenzweig, J., Zhang, P., Johns, D.C. & LaMotte, R.H. Expression of inwardly rectifying potassium channels by an inducible adenoviral vector reduced the neuronal hyperexcitability and hyperalgesia produced by chronic compression of the spinal ganglion. Mol. Pain 6, 65 (2010).

    PubMed  PubMed Central  Google Scholar 

  38. Uchida, H., Sasaki, K., Ma, L. & Ueda, H. Neuron-restrictive silencer factor causes epigenetic silencing of Kv4.3 gene after peripheral nerve injury. Neuroscience 166, 1–4 (2010).

    CAS  PubMed  Google Scholar 

  39. Rice, J.C. et al. Histone methyltransferases direct different degrees of methylation to define distinct chromatin domains. Mol. Cell 12, 1591–1598 (2003).

    CAS  PubMed  Google Scholar 

  40. Tachibana, M. et al. G9a histone methyltransferase plays a dominant role in euchromatic histone H3 lysine 9 methylation and is essential for early embryogenesis. Genes Dev. 16, 1779–1791 (2002).

    CAS  PubMed  PubMed Central  Google Scholar 

  41. Tachibana, M. et al. Histone methyltransferases G9a and GLP form heteromeric complexes and are both crucial for methylation of euchromatin at H3–K9. Genes Dev. 19, 815–826 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  42. Chiechio, S. et al. Epigenetic modulation of mGlu2 receptors by histone deacetylase inhibitors in the treatment of inflammatory pain. Mol. Pharmacol. 75, 1014–1020 (2009).

    CAS  PubMed  Google Scholar 

  43. Denk, F. et al. HDAC inhibitors attenuate the development of hypersensitivity in models of neuropathic pain. Pain 154, 1668–1679 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  44. Zhang, Y., Laumet, G., Chen, S.R., Hittelman, W.N. & Pan, H.L. Pannexin-1 up-regulation in the dorsal root ganglion contributes to neuropathic pain development. J. Biol. Chem. 290, 14647–14655 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  45. Ghare, S.S. et al. Coordinated histone H3 methylation and acetylation regulate physiologic and pathologic fas ligand gene expression in human CD4+ T cells. J. Immunol. 193, 412–421 (2014).

    CAS  PubMed  Google Scholar 

  46. Park, J.A. et al. Deacetylation and methylation at histone H3 lysine 9 (H3K9) coordinate chromosome condensation during cell cycle progression. Mol. Cells 31, 343–349 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  47. Chaturvedi, C.P. et al. Maintenance of gene silencing by the coordinate action of the H3K9 methyltransferase G9a/KMT1C and the H3K4 demethylase Jarid1a/KDM5A. Proc. Natl. Acad. Sci. USA 109, 18845–18850 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  48. Lee, D.Y., Northrop, J.P., Kuo, M.H. & Stallcup, M.R. Histone H3 lysine 9 methyltransferase G9a is a transcriptional coactivator for nuclear receptors. J. Biol. Chem. 281, 8476–8485 (2006).

    CAS  PubMed  Google Scholar 

  49. Vakoc, C.R., Mandat, S.A., Olenchock, B.A. & Blobel, G.A. Histone H3 lysine 9 methylation and HP1gamma are associated with transcription elongation through mammalian chromatin. Mol. Cell 19, 381–391 (2005).

    CAS  PubMed  Google Scholar 

  50. Laedermann, C.J., Pertin, M., Suter, M.R. & Decosterd, I. Voltage-gated sodium channel expression in mouse DRG after SNI leads to re-evaluation of projections of injured fibers. Mol. Pain 10, 19 (2014).

    PubMed  PubMed Central  Google Scholar 

  51. Chaplan, S.R., Bach, F.W., Pogrel, J.W., Chung, J.M. & Yaksh, T.L. Quantitative assessment of tactile allodynia in the rat paw. J. Neurosci. Methods 53, 55–63 (1994).

    CAS  PubMed  Google Scholar 

  52. Renn, C.L. et al. Multimodal assessment of painful peripheral neuropathy induced by chronic oxaliplatin-based chemotherapy in mice. Mol. Pain 7, 29 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  53. Bangaru, M.L., Park, F., Hudmon, A., McCallum, J.B. & Hogan, Q.H. Quantification of gene expression after painful nerve injury: validation of optimal reference genes. J. Mol. Neurosci. 46, 497–504 (2012).

    CAS  PubMed  Google Scholar 

  54. Chen, S.R. & Pan, H.L. Antinociceptive effect of morphine, but not mu opioid receptor number, is attenuated in the spinal cord of diabetic rats. Anesthesiology 99, 1409–1414 (2003).

    CAS  PubMed  Google Scholar 

  55. Subbanna, S. et al. G9a-mediated histone methylation regulates ethanol-induced neurodegeneration in the neonatal mouse brain. Neurobiol. Dis. 54, 475–485 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  56. Wilting, R.H. et al. Overlapping functions of Hdac1 and Hdac2 in cell cycle regulation and haematopoiesis. EMBO J. 29, 2586–2597 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  57. Zhang, Z., Cai, Y.Q., Zou, F., Bie, B. & Pan, Z.Z. Epigenetic suppression of GAD65 expression mediates persistent pain. Nat. Med. 17, 1448–1455 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  58. Mandal, M. et al. Epigenetic repression of the Igk locus by STAT5-mediated recruitment of the histone methyltransferase Ezh2. Nat. Immunol. 12, 1212–1220 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  59. Jelinek, J. et al. Conserved DNA methylation patterns in healthy blood cells and extensive changes in leukemia measured by a new quantitative technique. Epigenetics 7, 1368–1378 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  60. Mitchell, N.C. et al. Hfp inhibits Drosophila myc transcription and cell growth in a TFIIH/Hay-dependent manner. Development 137, 2875–2884 (2010).

    CAS  PubMed  Google Scholar 

  61. Trapnell, C., Pachter, L. & Salzberg, S.L. TopHat: discovering splice junctions with RNA-Seq. Bioinformatics 25, 1105–1111 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  62. Trapnell, C. et al. Transcript assembly and quantification by RNA-Seq reveals unannotated transcripts and isoform switching during cell differentiation. Nat. Biotechnol. 28, 511–515 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  63. Malin, S.A., Davis, B.M. & Molliver, D.C. Production of dissociated sensory neuron cultures and considerations for their use in studying neuronal function and plasticity. Nat. Protoc. 2, 152–160 (2007).

    CAS  PubMed  Google Scholar 

  64. Worley, B., Halouska, S. & Powers, R. Utilities for quantifying separation in PCA/PLS-DA scores plots. Anal. Biochem. 433, 102–104 (2013).

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We wish to thank C.L. Creasy from GlaxoSmithKline Laboratories for generously providing GSK503 and W.N. Hittleman at MD Anderson Cancer Center for assisting with the confocal microscopy. We also thank Z. Pan (MD Anderson Cancer Center) and F. Wang (Duke University) for providing Ehmt2flox/flox and AdvillinCre mice, respectively. This study was supported by the NIH Blueprint for Neuroscience Research–Grand Challenge on Chronic Pain (R01 DE022015) and the N.G. and Helen T. Hawkins endowment (H.-L.P.).

Author information

Authors and Affiliations

Authors

Contributions

G.L. performed biochemical and behavioral experiments and mouse breeding. S.-R.C. performed all surgeries, tissue extractions, and immunocytochemical experiments. Y.Z. performed ChIP-PCR experiments. J.G. and J.J. performed bisulfite sequencing experiments. J.G. and M.C. performed RNA sequencing experiments. D.-P.L. and T.M.S. performed DRG neuron dissociation and electrophysiological recordings. Y.D. performed some immunocytochemical and behavioral experiments. G.L., J.G., S.-R.C., D.-P.L., T.M.S., J.J., M.C., J.-P.I. and H.-L.P. analyzed the data. H.-L.P. and J.-P.I. conceived the project and supervised the study. G.L. and H.-L.P. wrote the manuscript.

Corresponding author

Correspondence to Hui-Lin Pan.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 Evidence for a new upstream exon in Kcna4 in the rat DRG.

(a) The UCSC Genome browser annotation indicating that rat Kcna4 is monoexonic and that the transcriptional start site (TSS) is around Chr3: 92781000. (a) Bisulfite sequencing data showing the annotated promoter region was hypermethylated in the DRG (open circle: unmethylated CpG sites; closed circle: methylated CpG sites). However, this gene is highly expressed in the DRG (Figure 1a). Because many species have an upstream exon and this region is very conserved (see http://genome.ucsc.edu/cgi-bin/hgGateway genome “rat” search “Kcna4”), we hypothesized that the rat genome also has this upstream exon. Using rat-mouse homology, we saw that the rat DNA sequence of Kcna4 was exactly the same as mouse DNA sequence. (a) We designed 3 pairs of primers (A, B, and C) to determine if rat Kv1.4 mRNA is the same as mouse Kv1.4 mRNA in control DRG samples. Two percent agarose gel image showing RT-PCR byproducts after reverse transcription (RT+) or without reverse transcription (RT-). Total RNAs were treated with DNase, and heat-inactivated cDNA were treated with RNase. We were able to amplify cDNA with these three pairs of primers, which suggested that rat and mouse Kcna4 genes have the similar structure within two exons. (a) Gene annotation map showing the location of the exons in rat, mouse, and human Kcna4 in the original database and the three primers used for panel a. (a) Evidence of an upstream exon in Kcna4 in the rat DRG. Kcna4 has a similar structure in the rat and mouse. Moreover, the new promoter region was not hypermethylated according to its mRNA level (Figure 3i).

Supplementary Figure 2 Nerve injury has no effect on expression of Kcna4, Kcnd2, Kcnq2, Kcnma1, H3K9me2 or G9a in the dorsal spinal cord.

(a) Mean data show the mRNA level of Kcna4, Kcnd2, Kcnq2, Kcnma1 and G9a in the dorsal spinal cord of sham control and SNL rats at 21 days after surgery (n = 11 rats in each group). The mRNA level was quantified by using real-time PCR and normalized to a housekeeping gene (Gapdh). (b) Representative gel images and quantification data show the protein level of G9a and H3K9me2 in the dorsal spinal cord of sham control and SNL rats at 21 days after surgery (n = 7 in each group). GAPDH and histone H3 were used as loading controls. Data are shown as mean ± s.e.m. Supplementary Figure S11 contains the uncropped version of the gel images.

Supplementary Figure 3 Changes in the expression of Hdac, Ehmt2, Ezh2 and Pcna in the DRG after nerve injury.

(a-f) Time course of changes in the mRNA levels of G9a (a), Ezh2 (b), Hdac1 (c), Hdac2 (d), Hdac4 (e), and Hdac5 (f) in L5 and L6 DRG tissues obtained from sham and SNL rats before and 5, 10 and 21 days after surgery (n = 6 in each group). mRNA data were quantified using real-time PCR and normalized to Gapdh. (g) mRNA levels of proliferation cell nuclear antigen (Pcna) in L5 and L6 DRGs obtained from sham and SNL rats before and 5, 10 and 21 days after surgery (n = 6 in each group). Data are presented as mean ± SEM. Statistical analysis was performed using repeated measures ANOVA followed by Dunnett’s post hoc test. * P < 0.05, ** P < 0.01, compared with baseline control (time 0).

Supplementary Figure 4 Effects of intrathecal injections of SAHA, UNC0638 or GSK503 on mechanical thresholds of rats treated with SNL or sham surgery.

(a-f) Quantitative data show the effects of intrathecal injection of 1-50 µg SAHA (a,b, n = 6 rats in each group), 0.1-10 µg UNC0638 (c,d, n = 6 rats in each group) or 0.5-25 µg GSK503 (e,f, n = 6 rats in each group) for 8 days on the Randall-Selitto and von Frey thresholds of SNL rats. (g,h) Quantitative data show the lack of effects of intrathecal daily injections of 5 µg GSK503, 50 µg SAHA or 10 µg UNC0638 for 8 days on the Randall-Selitto and von Frey thresholds in sham rats (n = 6 rats in each group). Data are presented as mean ± s.e.m. Statistical analysis was performed using repeated measures ANOVA followed by Dunnett’s post hoc test. * P < 0.05, ** P < 0.01, *** P < 0.001, compared with the baseline control (time 0).

Supplementary Figure 5 Effects of UNC0638, SAHA and GSK503 treatment on substrate levels of G9a, HDACs and EZH2 in the injured DRG.

(a,b) Western blot images and quantitative data showing the effects of intrathecal injections of vehicle, SAHA (50 µg), UNC0638 (10 µg), or GSK503 (5 µg) for 8 days on the protein levels of histone H3 in the DRG obtained from SNL rats (n = 6 in each group). GAPDH was used as a loading control. (c,d) Gel images and quantification data showing that intrathecal SAHA treatment increased the level of acetyl-H3, whereas treatment with UNC0638 and GSK503 decreased the level of H3K9me2 and H3K27me3, respectively, in the DRG obtained from SNL rats 28 days after surgery (n = 6 in each group). (e,f) Original gel images and quantification data showing the effects of intrathecal injections of SAHA, UNC0638, and GSK503 on the protein levels of H3K27me3, H3K9me2, H3K9ac and acetyl-H3 in the DRG obtained from SNL rats 28 days after surgery (n = 6 in each group). Histone H3 was used as a loading control. Data are shown as mean ± s.e.m. Statistical analysis was performed using the Mann-Whitney test or one-way ANOVA test. * P < 0.05, ** P < 0.01, *** P < 0.001, compared with the vehicle group. Supplementary Figure S12 contains the uncropped version of gel images.

Supplementary Figure 6 Nerve injury and UNC0638 treatment do not change the distribution of IB4-positive neurons in the DRG.

Representative confocal images show the distribution of IB4-positive and NeuN-immunoreactive neurons in the DRG. Double immunocytochemical labeling was performed on the left L5 DRG removed from sham-operated and SNL rats treated with vehicle or UNC0638 four weeks after surgery. The percentage of IB4-positive neurons over the total of NeuN-immunoreactive neurons in sham-operated rats, SNL rats treated with vehicle, and SNL rats treated with UNC0638 was 69.7 ± 6.5, 38.3 ± 5.7 and 38.5 ± 5.9, respectively (n = 12 DRG sections from 3 separate rats in each group).

Supplementary Figure 7 Nerve injury changes expression levels of multiple genes in the DRGs.

(a) Principal component analysis representation of the correlation among the expression values for all mapped genes (n = 16,876) for the different samples in each of the two experimental conditions (SNL plus vehicle, or SNL plus UNC0638) and the control (Sham). (b) Heat map generated with the expression values of the 2,035 genes with a change in expression level of at least two fold after nerve injury. The small dendrogram at top shows the clustering of the different samples based on the expression values (4 rats in the sham group, 4 rats in the SNL group, and 3 SNL rats treated with UNC0638). (c) Ingenuity Pathway Analysis showing the principal canonical pathways for the genes with changes in expression after nerve injury. The raw RNA sequence data have been deposited in the database repository of Gene Expression Omnibus (GEO accession #GSE59043).

Supplementary Figure 8 Uncropped gel images for Figure 1.

Original full-length western blotting gel images used for Figure 1b,d.

Supplementary Figure 9 Uncropped gel images for Figure 4.

Original full-length western blotting gel images used for Figure 4e,g.

Supplementary Figure 10 Uncropped gel images for Figure 7.

Original full-length western blotting gel images used for Figure 7b.

Supplementary Figure 11 Uncropped gel images for Supplementary Figure 2.

Original full-length western blotting gel images used for Supplementary Figure 2b.

Supplementary Figure 12 Uncropped gel images for Supplementary Figure 5.

Original full-length western blotting gel images used for Supplementary Figure 5a,c,e.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–12 and Supplementary Table 4 (PDF 3513 kb)

Supplementary Methods Checklist (PDF 567 kb)

Supplementary Table 1

List of DNA methylation levels of 80 CpG sites within 1 kb of TSSs in 30 K+ channel genes of DRGs obtained from sham-treated and SNL rats. (XLSX 11 kb)

Supplementary Table 2

List of 105 K+ channel-related genes and their expression levels in the DRGs from sham control, vehicle-treated SNL and UNC0638-treated SNL rats. (XLSX 22 kb)

Supplementary Table 3

List of 638 genes in the DRG that are altered by nerve injury and normalized by treatment with UNC06838. (XLSX 86 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Laumet, G., Garriga, J., Chen, SR. et al. G9a is essential for epigenetic silencing of K+ channel genes in acute-to-chronic pain transition. Nat Neurosci 18, 1746–1755 (2015). https://doi.org/10.1038/nn.4165

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nn.4165

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