We use cookies to improve your experience. By continuing to browse this site, you accept our cookie policy.×
Skip main navigation
Aging Health
Bioelectronics in Medicine
Biomarkers in Medicine
Breast Cancer Management
CNS Oncology
Colorectal Cancer
Concussion
Epigenomics
Future Cardiology
Future Medicine AI
Future Microbiology
Future Neurology
Future Oncology
Future Rare Diseases
Future Virology
Hepatic Oncology
HIV Therapy
Immunotherapy
International Journal of Endocrine Oncology
International Journal of Hematologic Oncology
Journal of 3D Printing in Medicine
Lung Cancer Management
Melanoma Management
Nanomedicine
Neurodegenerative Disease Management
Pain Management
Pediatric Health
Personalized Medicine
Pharmacogenomics
Regenerative Medicine

Epigenetic regulation of chronic pain

    Lingli Liang

    Department of Anesthesiology, New Jersey Medical School, Rutgers, The State University of New Jersey, 185 S. Orange Avenue, MSB F-548, Newark, NJ 07103, USA

    ,
    Brianna Marie Lutz

    Rutgers Graduate School of Biomedical Sciences, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ 07103, USA

    ,
    Alex Bekker

    Department of Anesthesiology, New Jersey Medical School, Rutgers, The State University of New Jersey, 185 S. Orange Avenue, MSB F-548, Newark, NJ 07103, USA

    &
    Yuan-Xiang Tao

    *Author for correspondence:

    E-mail Address: yt211@njms.rutgers.edu

    Department of Anesthesiology, New Jersey Medical School, Rutgers, The State University of New Jersey, 185 S. Orange Avenue, MSB F-548, Newark, NJ 07103, USA

    Department of Cell Biology & Molecular Medicine, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ 07103, USA

    Department of Neurology & Neuroscience, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ 07103, USA

    Department of Physiology & Pharmacology, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ 07103, USA

    Published Online:https://doi.org/10.2217/epi.14.75

    Chronic pain arising from peripheral inflammation and tissue or nerve injury is a common clinical symptom. Although intensive research on the neurobiological mechanisms of chronic pain has been carried out during previous decades, this disorder is still poorly managed by current drugs such as opioids and nonsteroidal anti-inflammatory drugs. Inflammation, tissue injury and/or nerve injury-induced changes in gene expression in sensory neurons of the dorsal root ganglion, spinal cord dorsal horn and pain-associated brain regions are thought to participate in chronic pain genesis; however, how these changes occur is still elusive. Epigenetic modifications including DNA methylation and covalent histone modifications control gene expression. Recent studies have shown that peripheral noxious stimulation changes DNA methylation and histone modifications and that these changes may be related to the induction of pain hypersensitivity under chronic pain conditions. This review summarizes the current knowledge and progress in epigenetic research in chronic pain and discusses the potential role of epigenetic modifications as therapeutic antinociceptive targets in this disorder.

    Papers of special note have been highlighted as: • of interest; •• of considerable interest

    References

    • 1 Morley S. Psychology of pain. Br. J. Anaesth. 101(1), 25–31 (2008).
    • 2 Latremoliere A, Woolf CJ. Central sensitization: a generator of pain hypersensitivity by central neural plasticity. J. Pain 10(9), 895–926 (2009).
    • 3 Campbell JN, Meyer RA. Mechanisms of neuropathic pain. Neuron 52(1), 77–92 (2006).
    • 4 Wang W, Gu J, Li YQ, Tao YX. Are voltage-gated sodium channels on the dorsal root ganglion involved in the development of neuropathic pain? Mol. Pain 7, 16 (2011).
    • 5 Bali P, Im HI, Kenny PJ. Methylation, memory and addiction. Epigenetics 6(6), 671–674 (2011).
    • 6 Feng J, Zhou Y, Campbell SL et al. Dnmt1 and Dnmt3a maintain DNA methylation and regulate synaptic function in adult forebrain neurons. Nat. Neurosci. 13(4), 423–430 (2010).
    • 7 Kramer JM, Kochinke K, Oortveld MA et al. Epigenetic regulation of learning and memory by Drosophila EHMT/G9a. PLoS Biol. 9(1), e1000569 (2011).
    • 8 Lutz BM, Bekker A, Tao YX. Noncoding RNAs: new players in chronic pain. Anesthesiology 121(2), 409–417 (2014).•• More updated review regarding the role of noncoding RNAs in chronic pain.
    • 9 Wang F, Stefano GB, Kream RM. Epigenetic modification of DRG neuronal gene expression subsequent to nerve injury: etiological contribution to complex regional pain syndromes (Part II). Med. Sci. Monit. 20, 1188–1200 (2014).
    • 10 Zovkic IB, Guzman-Karlsson MC, Sweatt JD. Epigenetic regulation of memory formation and maintenance. Learn. Mem. 20(2), 61–74 (2013).
    • 11 Mauck M, Van de Ven T, Shaw AD. Epigenetics of chronic pain after thoracic surgery. Curr. Opin. Anaesthesiol. 27(1), 1–5 (2014).
    • 12 Rahn EJ, Guzman-Karlsson MC, David SJ. Cellular, molecular, and epigenetic mechanisms in non-associative conditioning: implications for pain and memory. Neurobiol. Learn. Mem. 105, 133–150 (2013).
    • 13 Seo S, Grzenda A, Lomberk G, Ou XM, Cruciani RA, Urrutia R. Epigenetics: a promising paradigm for better understanding and managing pain. J. Pain 14(6), 549–557 (2013).
    • 14 Stone LS, Szyf M. The emerging field of pain epigenetics. Pain 154(1), 1–2 (2013).
    • 15 Kouzarides T. Chromatin modifications and their function. Cell 128(4), 693–705 (2007).
    • 16 Kuo MH, Allis CD. Roles of histone acetyltransferases and deacetylases in gene regulation. Bioessays 20(8), 615–626 (1998).
    • 17 Bannister AJ, Kouzarides T. Regulation of chromatin by histone modifications. Cell Res. 21(3), 381–395 (2011).
    • 18 Contestabile A, Sintoni S. Histone acetylation in neurodevelopment. Curr. Pharm. Des. 19(28), 5043–5050 (2013).
    • 19 Gong F, Miller KM. Mammalian DNA repair: HATs and HDACs make their mark through histone acetylation. Mutat. Res. 750(1–2), 23–30 (2013).
    • 20 Bai G, Wei D, Zou S, Ren K, Dubner R. Inhibition of class II histone deacetylases in the spinal cord attenuates inflammatory hyperalgesia. Mol. Pain 6, 51 (2010).
    • 21 Chiechio S, Zammataro M, Morales ME et al. Epigenetic modulation of mGlu2 receptors by histone deacetylase inhibitors in the treatment of inflammatory pain. Mol. Pharmacol. 75(5), 1014–1020 (2009).
    • 22 Zhang Z, Cai YQ, Zou F, Bie B, Pan ZZ. Epigenetic suppression of GAD65 expression mediates persistent pain. Nat. Med. 17(11), 1448–1455 (2011).• Recognized the significant role of histone modefication in inflammatory pain
    • 23 Tran L, Chaloner A, Sawalha AH, Greenwood Van-Meerveld B. Importance of epigenetic mechanisms in visceral pain induced by chronic water avoidance stress. Psychoneuroendocrinology 38(6), 898–906 (2013).
    • 24 Kukkar A, Singh N, Jaggi AS. Attenuation of neuropathic pain by sodium butyrate in an experimental model of chronic constriction injury in rats. J. Formos. Med. Assoc. S0929–S6646(13), 00180 (2013).
    • 25 Denk F, Huang W, Sidders B et al. HDAC inhibitors attenuate the development of hypersensitivity in models of neuropathic pain. Pain 154(9), 1668–1679 (2013).
    • 26 Matsushita Y, Araki K, Omotuyi O, Mukae T, Ueda H. HDAC inhibitors restore C-fibre sensitivity in experimental neuropathic pain model. Br. J. Pharmacol. 170(5), 991–998 (2013).
    • 27 Uchida H, Ma L, Ueda H. Epigenetic gene silencing underlies C-fiber dysfunctions in neuropathic pain. J. Neurosci. 30(13), 4806–4814 (2010).•• The first study to identify the role of histone modefication in neuropathic pain.
    • 28 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), 1–4 (2010).
    • 29 Uchida H, Matsushita Y, Ueda H. Epigenetic regulation of BDNF expression in the primary sensory neurons after peripheral nerve injury: implications in the development of neuropathic pain. Neuroscience 240, 147–154 (2013).
    • 30 Li K, Zhao GQ, Li LY, Wu GZ, Cui SS. Epigenetic upregulation of Cdk5 in the dorsal horn contributes to neuropathic pain in rats. Neuroreport 25(14), 1116–1121 (2014).
    • 31 Kiguchi N, Kobayashi Y, Maeda T et al. Epigenetic augmentation of the macrophage inflammatory protein 2/C-X-C chemokine receptor type 2 axis through histone H3 acetylation in injured peripheral nerves elicits neuropathic pain. J. Pharmacol. Exp. Ther. 340(3), 577–587 (2012).
    • 32 Kiguchi N, Kobayashi Y, Saika F, Kishioka S. Epigenetic upregulation of CCL2 and CCL3 via histone modifications in infiltrating macrophages after peripheral nerve injury. Cytokine 64(3), 666–672 (2013).
    • 33 Zhu XY, Huang CS, Li Q et al. p300 exerts an epigenetic role in chronic neuropathic pain through its acetyltransferase activity in rats following chronic constriction injury (CCI). Mol. Pain 8, 84 (2012).
    • 34 Zhu XY, Huang CS, Li Q et al. Temporal distribution of p300/CBP immunoreactivity in the adult rat spinal dorsal horn following chronic constriction injury (CCI). Cell Mol. Neurobiol. 33(2), 197–204 (2013).
    • 35 Yin Q, Lu FF, Zhao Y et al. Resveratrol facilitates pain attenuation in a rat model of neuropathic pain through the activation of spinal Sirt1. Reg. Anesth. Pain Med. 38(2), 93–99 (2013).
    • 36 Sun Y, Liang D, Sahbaie P, Clark JD. Effects of methyl donor diets on incisional pain in mice. PLoS One 8(10), e77881 (2013).
    • 37 Sun Y, Sahbaie P, Liang DY et al. Epigenetic regulation of spinal CXCR2 signaling in incisional hypersensitivity in mice. Anesthesiology 119(5), 1198–1208 (2013).
    • 38 Mayer DJ, Mao J, Holt J, Price DD. Cellular mechanisms of neuropathic pain, morphine tolerance, and their interactions. Proc. Natl Acad. Sci. USA 96(14), 7731–7736 (1999).
    • 39 Liang DY, Li X, Clark JD. Epigenetic regulation of opioid-induced hyperalgesia, dependence, and tolerance in mice. J. Pain 14(1), 36–47 (2013).
    • 40 He X, Ou P, Wu K et al. Resveratrol attenuates morphine antinociceptive tolerance via SIRT1 regulation in the rat spinal cord. Neurosci. Lett. 566, 55–60 (2014).
    • 41 Cherng CH, Lee KC, Chien CC et al. Baicalin ameliorates neuropathic pain by suppressing HDAC1 expression in the spinal cord of spinal nerve ligation rats. J. Formos. Med. Assoc. 113(8), 513–520 (2014).
    • 42 Klein K, Ospelt C, Gay S. Epigenetic contributions in the development of rheumatoid arthritis. Arthritis Res. Ther. 14(6), 227 (2012).
    • 43 Covington HE III, Maze I, Sun H et al. A role for repressive histone methylation in cocaine-induced vulnerability to stress. Neuron 71(4), 656–670 (2011).
    • 44 Gupta S, Kim SY, Artis S et al. Histone methylation regulates memory formation. J. Neurosci. 30(10), 3589–3599 (2010).
    • 45 Maze I, Covington HE III, Dietz DM et al. Essential role of the histone methyltransferase G9a in cocaine-induced plasticity. Science 327(5962), 213–216 (2010).
    • 46 Imai S, Ikegami D, Yamashita A et al. Epigenetic transcriptional activation of monocyte chemotactic protein 3 contributes to long-lasting neuropathic pain. Brain 136(Pt 3), 828–843 (2013).
    • 47 Tsai RY, Shen CH, Feng YP et al. Ultra-low-dose naloxone enhances the antinociceptive effect of morphine in PTX-treated rats: regulation on global histone methylation. Acta Anaesthesiol. Taiwan. 50(3), 106–111 (2012).
    • 48 Zhang Z, Tao W, Hou YY, Wang W, Kenny PJ, Pan ZZ. MeCP2 repression of G9a in regulation of pain and morphine reward. J. Neurosci 34(27), 9076–9087 (2014).
    • 49 Chedin F, Lieber MR, Hsieh CL. The DNA methyltransferase-like protein DNMT3L stimulates de novo methylation by Dnmt3a. Proc. Natl Acad. Sci. USA 99(26), 16916–16921 (2002).
    • 50 Jeltsch A. Molecular enzymology of mammalian DNA methyltransferases. Curr. Top. Microbiol. Immunol. 301, 203–225 (2006).
    • 51 Siedlecki P, Zielenkiewicz P. Mammalian DNA methyltransferases. Acta Biochim. Pol. 53(2), 245–256 (2006).
    • 52 Fatemi M, Hermann A, Gowher H, Jeltsch A. Dnmt3a and Dnmt1 functionally cooperate during de novo methylation of DNA. Eur. J. Biochem. 269(20), 4981–4984 (2002).
    • 53 Gowher H, Stockdale CJ, Goyal R, Ferreira H, Owen-Hughes T, Jeltsch A. De novo methylation of nucleosomal DNA by the mammalian Dnmt1 and Dnmt3A DNA methyltransferases. Biochemistry 44(29), 9899–9904 (2005).
    • 54 Liang G, Chan MF, Tomigahara Y et al. Cooperativity between DNA methyltransferases in the maintenance methylation of repetitive elements. Mol. Cell Biol. 22(2), 480–491 (2002).
    • 55 Lubin FD, Gupta S, Parrish RR, Grissom NM, Davis RL. Epigenetic mechanisms: critical contributors to long-term memory formation. Neuroscientist 17(6), 616–632 (2011).
    • 56 Mifsud KR, Gutierrez-Mecinas M, Trollope AF, Collins A, Saunderson EA, Reul JM. Epigenetic mechanisms in stress and adaptation. Brain Behav. Immun. 25(7), 1305–1315 (2011).
    • 57 Turek-Plewa J, Jagodzinski PP. The role of mammalian DNA methyltransferases in the regulation of gene expression. Cell Mol. Biol. Lett. 10(4), 631–647 (2005).
    • 58 Qi F, Zhou Y, Xiao Y et al. Promoter demethylation of cystathionine-beta-synthetase gene contributes to inflammatory pain in rats. Pain 154(1), 34–45 (2013).
    • 59 Pan Z, Zhu LJ, Li YQ et al. Epigenetic modification of spinal miR-219 expression regulates chronic inflammation pain by targeting CaMKIIgamma. J. Neurosci 34(29), 9476–9483 (2014).
    • 60 Wang Y, Liu C, Guo QL et al. Intrathecal 5-azacytidine inhibits global DNA methylation and methyl-CpG-binding protein 2 expression and alleviates neuropathic pain in rats following chronic constriction injury. Brain Res. 1418, 64–69 (2011).• The first study to demonstrate the role of DNA methylation in neuropathic pain.
    • 61 Zhou XL, Yu LN, Wang Y et al. Increased methylation of the MOR gene proximal promoter in primary sensory neurons plays a crucial role in the decreased analgesic effect of opioids in neuropathic pain. Mol. Pain 10, 51 (2014).
    • 62 Pollema-Mays SL, Centeno MV, Apkarian AV, Martina M. Expression of DNA methyltransferases in adult dorsal root ganglia is cell-type specific and up regulated in a rodent model of neuropathic pain. Front Cell Neurosci. 8, 217 (2014).
    • 63 Tajerian M, Alvarado S, Millecamps M et al. Peripheral nerve injury is associated with chronic, reversible changes in global DNA methylation in the mouse prefrontal cortex. PLoS One 8(1), e55259 (2013).
    • 64 Menzies V, Lyon DE, Archer KJ et al. Epigenetic alterations and an increased frequency of micronuclei in women with fibromyalgia. Nurs. Res. Pract. 2013, 795784 (2013).
    • 65 Tajerian M, Alvarado S, Millecamps M et al. DNA methylation of SPARC and chronic low back pain. Mol. Pain 7, 65 (2011).
    • 66 Viet CT, Ye Y, Dang D et al. Re-expression of the methylated EDNRB gene in oral squamous cell carcinoma attenuates cancer-induced pain. Pain 152(10), 2323–2332 (2011).
    • 67 Bell JT, Loomis AK, Butcher LM et al. Differential methylation of the TRPA1 promoter in pain sensitivity. Nat. Commun. 5, 2978 (2014).
    • 68 Karouzakis E, Gay RE, Michel BA, Gay S, Neidhart M. DNA hypomethylation in rheumatoid arthritis synovial fibroblasts. Arthritis Rheum. 60(12), 3613–3622 (2009).
    • 69 Liu CC, Fang TJ, Ou TT et al. Global DNA methylation, DNMT1, and MBD2 in patients with rheumatoid arthritis. Immunol. Lett. 135(1–2), 96–99 (2011).
    • 70 Nakano K, Whitaker JW, Boyle DL, Wang W, Firestein GS. DNA methylome signature in rheumatoid arthritis. Ann. Rheum. Dis. 72(1), 110–117 (2013).
    • 71 Karouzakis E, Gay RE, Gay S, Neidhart M. Increased recycling of polyamines is associated with global DNA hypomethylation in rheumatoid arthritis synovial fibroblasts. Arthritis Rheum. 64(6), 1809–1817 (2012).
    • 72 Klein K, Gay S. Epigenetic modifications in rheumatoid arthritis, a review. Curr. Opin. Pharmacol. 13(3), 420–425 (2013).
    • 73 Sansom OJ, Maddison K, Clarke AR. Mechanisms of disease: methyl-binding domain proteins as potential therapeutic targets in cancer. Nat. Clin Pract. Oncol. 4(5), 305–315 (2007).
    • 74 Cuddapah VA, Pillai RB, Shekar KV et al. Methyl-CpG-binding protein 2 (MECP2) mutation type is associated with disease severity in Rett syndrome. J. Med. Genet. 51(3), 152–158 (2014).
    • 75 Downs J, Geranton SM, Bebbington A et al. Linking MECP2 and pain sensitivity: the example of Rett syndrome. Am. J. Med. Genet. A 152A(5), 1197–1205 (2010).
    • 76 Geranton SM, Fratto V, Tochiki KK, Hunt SP. Descending serotonergic controls regulate inflammation-induced mechanical sensitivity and methyl-CpG-binding protein 2 phosphorylation in the rat superficial dorsal horn. Mol. Pain 4, 350 (2008).• First study that demonstrated the involvement of MeCP2 in inflammatory pain.
    • 77 Tochiki KK, Cunningham J, Hunt SP, Geranton SM. The expression of spinal methyl-CpG-binding protein 2, DNA methyltransferases and histone deacetylases is modulated in persistent pain states. Mol. Pain 8, 14 (2012).
    • 78 Chen WG, Chang Q, Lin Y et al. Derepression of BDNF transcription involves calcium-dependent phosphorylation of MeCP2. Science 302(5646), 885–889 (2003).