Genes and epigenetic processes as prospective pain targets

Megan Crow, Franziska Denk, Stephen B McMahon, Megan Crow, Franziska Denk, Stephen B McMahon

Abstract

Chronic pain affects approximately one in five adults, resulting in a greatly reduced quality of life and a higher risk of developing co-morbidities such as depression. Available treatments often provide inadequate pain relief, but it is hoped that through deeper understanding of the molecular mechanisms underlying chronic pain states we can discover new and improved therapies. Although genetic research has flourished over the past decade and has identified many key genes in pain processing, the budding field of epigenetics promises to provide new insights and a more dynamic view of pain regulation. This review gives an overview of basic mechanisms and current therapies to treat pain, and discusses the clinical and preclinical evidence for the contribution of genetic and epigenetic factors, with a focus on how this knowledge can affect drug development.

Figures

Figure 1
Figure 1
How polymorphisms can confer risk to pain. Single nucleotide polymorphisms (SNPs) can confer increased risk to pain by causing missense mutations that alter protein function. One of the most dramatic examples of this phenomenon is SNPs in the voltage-gated sodium channel Nav1.7. In this case, a SNP causing a change from an isoleucine to threonine residue in the loop domain leads to loss of channel inactivation, which is responsible for inherited paroxysmal pain disorder [27]. (a) Structure of Nav1.7. Arrow indicates the mutation in the loop domain. (b) Human embryonic kidney (HEK) cells transfected with wild-type Nav1.7 show normal channel inactivation. (c) HEK cells transfected with mutant Nav1.7 are unable to inactivate. Adapted with permission from [27].
Figure 2
Figure 2
Evidence for epigenetic modulation in pain. Evidence has been obtained for such modulation at four different levels, numbered here in order from peripheral to central. 1, Pain-associated hyperacetylation of MIP2 and CXCR5 in the nerve after partial sciatic nerve ligation (PSL) [86] (shown in yellow). 2, Decreased expression of MeCP2 target genes after CFA [91]; miRNA expression changes [104,106]; intrathecal HDAC inhibitor treatment reduces acute pain after CFA [83] (shown in green). 3, GAD2 hypoacetylation after CFA leads to loss of descending inhibition [84] (shown in pink). 4, Carrageenan-associated miRNA dysregulation in the prefrontal cortex [105] (shown in purple).
Figure 3
Figure 3
How epigenetic mechanisms can influence pain processing. (a) Under normal conditions, histone tails are acetylated at the GAD2 promoter in the nucleus raphe magnus (NRM). (b) After application of complete Freund's adjuvant (CFA), Gad65 expression is suppressed through hypoacetylation of the GAD2 promoter, leading to loss of descending inhibition from the NRM [84]. GABA, γ-aminobutyric acid.

References

    1. Breivik H, Collett B, Ventafridda V, Cohen R, Gallacher D. Survey of chronic pain in Europe: Prevalence, impact on daily life, and treatment. Eur J Pain. 2006;10:287–333. doi: 10.1016/j.ejpain.2005.06.009.
    1. Gureje O. VKMSGEGR. Persistent pain and well-being: A world health organization study in primary care. JAMA. 1998;280:147–151. doi: 10.1001/jama.280.2.147.
    1. The British Pain Society - FAQs.
    1. Kissin I. The development of new analgesics over the past 50 years: a lack of real breakthrough drugs. Anesth Analg. 2010;110:780–789. doi: 10.1213/ANE.0b013e3181cde882.
    1. Elliott MJ, Maini RN, Feldmann M, Kalden JR, Antoni C, Smolen JS, Leeb B, Breedveld FC, Macfarlane JD, Bijl JA, Woody JN. Randomised double-blind comparison of chimeric monoclonal antibody to tumour necrosis factor α (cA2) versus placebo in rheumatoid arthritis. Lancet. 1994;344:1105–1110. doi: 10.1016/S0140-6736(94)90628-9.
    1. Tobinick E, Davoodifar S. Efficacy of etanercept delivered by perispinal administration for chronic back and/or neck disc-related pain: a study of clinical observations in 143 patients. Curr Med Res Opin. 2004;20:1075–1085. doi: 10.1185/030079903125004286.
    1. Lane NE, Schnitzer TJ, Birbara CA, Mokhtarani M, Shelton DL, Smith MD, Brown MT. Tanezumab for the treatment of pain from osteoarthritis of the knee. N Engl J Med. 2010;363:1521–1531. doi: 10.1056/NEJMoa0901510.
    1. Tanezumab: Arthritis Advisory Committee Briefing Document.
    1. Costigan M, Scholz J, Woolf CJ. Neuropathic pain: a maladaptive response of the nervous system to damage. Annu Rev Neurosci. 2009;32:1–32. doi: 10.1146/annurev.neuro.051508.135531.
    1. Norbury TA, MacGregor AJ, Urwin J, Spector TD, McMahon SB. Heritability of responses to painful stimuli in women: a classical twin study. Brain. 2007;130:3041–3049. doi: 10.1093/brain/awm233.
    1. Nielsen CS, Stubhaug A, Price DD, Vassend O, Czajkowski N, Harris JR. Individual differences in pain sensitivity: genetic and environmental contributions. Pain. 2008;136:21–29. doi: 10.1016/j.pain.2007.06.008.
    1. Hartvigsen J, Nielsen J, Kyvik KO, Fejer R, Vach W, Iachine I, Leboeuf-Yde C. Heritability of spinal pain and consequences of spinal pain: a comprehensive genetic epidemiologic analysis using a population-based sample of 15,328 twins ages 20-71 years. Arthritis Rheum. 2009;61:1343–1351. doi: 10.1002/art.24607.
    1. Williams FM, Scollen S, Cao D, Memari Y, Hyde CL, Zhang B, Sidders B, Ziemek D, Shi Y, Harris J, Harrow I, Dougherty B, Malarstig A, McEwen R, Stephens JC, Patel K, Menni C, Shin SY, Hodgkiss D, Surdulescu G, He W, Jin X, McMahon SB, Soranzo N, John S, Wang J, Spector TD. Genes contributing to pain sensitivity in the normal population: an exome sequencing study. PLoS Genet. 2012;8:e1003095. doi: 10.1371/journal.pgen.1003095.
    1. Hocking LJ, Morris AD, Dominiczak AF, Porteous DJ, Smith BH. Heritability of chronic pain in 2195 extended families. Eur J Pain. 2012;16:1053–1063. doi: 10.1002/j.1532-2149.2011.00095.x.
    1. Livshits G, Popham M, Malkin I, Sambrook PN, Macgregor AJ, Spector T, Williams FM. Lumbar disc degeneration and genetic factors are the main risk factors for low back pain in women: the UK Twin Spine Study. Ann Rheum Dis. 2011;70:1740–1745. doi: 10.1136/ard.2010.137836.
    1. MacGregor AJ, Andrew T, Sambrook PN, Spector TD. Structural, psychological, and genetic influences on low back and neck pain: a study of adult female twins. Arthritis Rheum. 2004;51:160–167. doi: 10.1002/art.20236.
    1. Mogil JS. Pain genetics: past, present and future. Trends Genet. 2012;28:258–266. doi: 10.1016/j.tig.2012.02.004.
    1. Foulkes T, Wood JN. Pain genes. PLoS Genet. 2008;4:e1000086. doi: 10.1371/journal.pgen.1000086.
    1. Indo Y, Tsuruta M, Hayashida Y, Karim MA, Ohta K, Kawano T, Mitsubuchi H, Tonoki H, Awaya Y, Matsuda I. Mutations in the TRKA/NGF receptor gene in patients with congenital insensitivity to pain with anhidrosis. Nat Genet. 1996;13:485–488. doi: 10.1038/ng0896-485.
    1. Cox JJ, Reimann F, Nicholas AK, Thornton G, Roberts E, Springell K, Karbani G, Jafri H, Mannan J, Raashid Y, Al-Gazali L, Hamamy H, Valente EM, Gorman S, Williams R, McHale DP, Wood JN, Gribble FM, Woods CG. An SCN9A channelopathy causes congenital inability to experience pain. Nature. 2006;444:894–898. doi: 10.1038/nature05413.
    1. Weiss J, Pyrski M, Jacobi E, Bufe B, Willnecker V, Schick B, Zizzari P, Gossage SJ, Greer CA, Leinders-Zufall T, Woods CG, Wood JN, Zufall F. Loss-of-function mutations in sodium channel Nav1.7 cause anosmia. Nature. 2011;472:186–190. doi: 10.1038/nature09975.
    1. Nilsen KB, Nicholas AK, Woods CG, Mellgren SI, Nebuchennykh M, Aasly J. Two novel SCN9A mutations causing insensitivity to pain. Pain. 2009;143:155–158. doi: 10.1016/j.pain.2009.02.016.
    1. Nassar MA, Stirling LC, Forlani G, Baker MD, Matthews EA, Dickenson AH, Wood JN. Nociceptor-specific gene deletion reveals a major role for Nav1.7 (PN1) in acute and inflammatory pain. Proc Natl Acad Sci USA. 2004;101:12706–12711. doi: 10.1073/pnas.0404915101.
    1. Raouf R, Quick K, Wood JN. Pain as a channelopathy. J Clin Invest. 2010;120:3745–3752. doi: 10.1172/JCI43158.
    1. Smith LA, Allen EV. Erythermalgia (erythromelalgia) of the extremities: a syndrome characterized by Redness, heat, and pain. Am Heart J. 1938;16:175–188. doi: 10.1016/S0002-8703(38)90693-3.
    1. Yang Y, Wang Y, Li S, Xu Z, Li H, Ma L, Fan J, Bu D, Liu B, Fan Z, Wu G, Jin J, Ding B, Zhu X, Shen Y. Mutations in SCN9A, encoding a sodium channel alpha subunit, in patients with primary erythermalgia. J Med Genet. 2004;41:171–174. doi: 10.1136/jmg.2003.012153.
    1. Segerdahl AR, Xie J, Paterson K, Ramirez JD, Tracey I, Bennett DL. Imaging the neural correlates of neuropathic pain and pleasurable relief associated with inherited erythromelalgia in a single subject with quantitative arterial spin labelling. Pain. 2012;153:1122–1127. doi: 10.1016/j.pain.2011.12.012.
    1. Fertleman CR, Baker MD, Parker KA, Moffatt S, Elmslie FV, Abrahamsen B, Ostman J, Klugbauer N, Wood JN, Gardiner RM, Rees M. SCN9A mutations in paroxysmal extreme pain disorder: allelic variants underlie distinct channel defects and phenotypes. Neuron. 2006;52:767–774. doi: 10.1016/j.neuron.2006.10.006.
    1. Wang T, Yu D, Lamb ML. Trk kinase inhibitors as new treatments for cancer and pain. Expert Opin Ther Pat. 2009;19:305–319. doi: 10.1517/13543770902721261.
    1. Pezet S, McMahon SB. Neurotrophins: mediators and modulators of pain. Annu Rev Neurosci. 2006;29:507–538. doi: 10.1146/annurev.neuro.29.051605.112929.
    1. England S. Voltage-gated sodium channels: the search for subtype-selective analgesics. Expert Opin Investig Drugs. 2008;17:1849–1864. doi: 10.1517/13543780802514559.
    1. Chowdhury S, Chafeev M, Liu S, Sun J, Raina V, Chui R, Young W, Kwan R, Fu J, Cadieux JA. Discovery of XEN907, a spirooxindole blocker of NaV1.7 for the treatment of pain. Bioorg Med Chem Lett. 2011;21:3676–3681. doi: 10.1016/j.bmcl.2011.04.088.
    1. Hoyt SB, London C, Ok H, Gonzalez E, Duffy JL, Abbadie C, Dean B, Felix JP, Garcia ML, Jochnowitz N, Karanam BV, Li X, Lyons KA, McGowan E, Macintyre DE, Martin WJ, Priest BT, Smith MM, Tschirret-Guth R, Warren VA, Williams BS, Kaczorowski GJ, Parsons WH. Benzazepinone Nav1.7 blockers: potential treatments for neuropathic pain. Bioorg Med Chem Lett. 2007;17:6172–6177. doi: 10.1016/j.bmcl.2007.09.032.
    1. McGowan E, Hoyt SB, Li X, Lyons KA, Abbadie C. A peripherally acting Nav1.7 sodium channel blocker reverses hyperalgesia and allodynia on rat models of inflammatory and neuropathic pain. Anesth Analg. 2009;109:951–958. doi: 10.1213/ane.0b013e3181b01b02.
    1. Nguyen HN, Bregman H, Buchanan JL, Du B, Feric E, Huang L, Li X, Ligutti J, Liu D, Malmberg AB, Matson DJ, McDermott JS, Patel VF, Wilenkin B, Zou A, McDonough SI, Dimauro EF. Discovery and optimization of aminopyrimidinones as potent and state-dependent Nav1.7 antagonists. Bioorg Med Chem Lett. 2012;22:1055–1060. doi: 10.1016/j.bmcl.2011.11.111.
    1. Schmalhofer WA, Calhoun J, Burrows R, Bailey T, Kohler MG, Weinglass AB, Kaczorowski GJ, Garcia ML, Koltzenburg M, Priest BT. ProTx-II, a selective inhibitor of NaV1.7 sodium channels, blocks action potential propagation in nociceptors. Mol Pharmacol. 2008;74:1476–1484. doi: 10.1124/mol.108.047670.
    1. A Phase 2a, Randomized, Double-Blind, Placebo-Controlled, Two-Period Crossover Study to Evaluate the Safety, Tolerability, Preliminary Efficacy, and Systemic Exposure of Topical XPF-002 in Subjects With Postherpetic Neuralgia.
    1. Phase 2a, Exploratory, Double-blind, Placebo-controlled Two-part Study to Evaluate the Safety, Efficacy, Tolerability and Pharmacokinetics of Topically Applied XPF-002 (XEN402 8% w/w Ointment) in Patients With Primary/Inherited Erythromelalgia.
    1. A Randomized, Double Blind, Cross-over Study to Evaluate the Safety and Efficacy of CNV1014802 in Subjects With Neuropathic Pain From Lumbosacral Radiculopathy.
    1. Goldberg YP, Price N, Namdari R, Cohen CJ, Lamers MH, Winters C, Price J, Young CE, Verschoof H, Sherrington R, Pimstone SN, Hayden MR. Treatment of Na(v)1.7-mediated pain in inherited erythromelalgia using a novel sodium channel blocker. Pain. 2012;153:8085.
    1. Diatchenko L, Nackley AG, Tchivileva IE, Shabalina SA, Maixner W. Genetic architecture of human pain perception. Trends Genet. 2007;23:605–613. doi: 10.1016/j.tig.2007.09.004.
    1. Tegeder I, Costigan M, Griffin RS, Abele A, Belfer I, Schmidt H, Ehnert C, Nejim J, Marian C, Scholz J, Wu T, Allchorne A, Diatchenko L, Binshtok AM, Goldman D, Adolph J, Sama S, Atlas SJ, Carlezon WA, Parsegian A, Lötsch J, Fillingim RB, Maixner W, Geisslinger G, Max MB, Woolf CJ. GTP cyclohydrolase and tetrahydrobiopterin regulate pain sensitivity and persistence. Nat Med. 2006;12:1269–1277. doi: 10.1038/nm1490.
    1. Campbell CM, Edwards RR, Carmona C, Uhart M, Wand G, Carteret A, Kim YK, Frost J, Campbell JN. Polymorphisms in the GTP cyclohydrolase gene (GCH1) are associated with ratings of capsaicin pain. Pain. 2009;141:114–118. doi: 10.1016/j.pain.2008.10.023.
    1. Kim H, Dionne RA. Lack of influence of GTP cyclohydrolase gene (GCH1) variations on pain sensitivity in humans. Mol Pain. 2007;3:6. doi: 10.1186/1744-8069-3-6.
    1. Wadley AL, Lombard Z, Cherry CL, Price P, Kamerman PR. Analysis of a previously identified 'pain protective' haplotype and individual polymorphisms in the GCH1 gene in Africans with HIV-associated sensory neuropathy: a genetic association study. J Acquir Immune Defic Syndr. 2012;60:20–23. doi: 10.1097/QAI.0b013e31824bcc17.
    1. Zubieta JK, Heitzeg MM, Smith YR, Bueller JA, Xu K, Xu Y, Koeppe RA, Stohler CS, Goldman D. COMT val158met genotype affects mu-opioid neurotransmitter responses to a pain stressor. Science. 2003;299:1240–1243. doi: 10.1126/science.1078546.
    1. Mukherjee N, Kidd KK, Pakstis AJ, Speed WC, Li H, Tarnok Z, Barta C, Kajuna SL, Kidd JR. The complex global pattern of genetic variation and linkage disequilibrium at catechol-O-methyltransferase. Mol Psychiatry. 2010;15:216–225. doi: 10.1038/mp.2008.64.
    1. Diatchenko L, Nackley AG, Slade GD, Bhalang K, Belfer I, Max MB, Goldman D, Maixner W. Catechol-O-methyltransferase gene polymorphisms are associated with multiple pain-evoking stimuli. Pain. 2006;125:216–224. doi: 10.1016/j.pain.2006.05.024.
    1. Segall SK, Nackley AG, Diatchenko L, Lariviere WR, Lu X, Marron JS, Grabowski-Boase L, Walker JR, Slade G, Gauthier J, Bailey JS, Steffy BM, Maynard TM, Tarantino LM, Wiltshire T. Comt1 genotype and expression predicts anxiety and nociceptive sensitivity in inbred strains of mice. Genes Brain Behav. 2010;9:933–946. doi: 10.1111/j.1601-183X.2010.00633.x.
    1. Kim H, Mittal DP, Iadarola MJ, Dionne RA. Genetic predictors for acute experimental cold and heat pain sensitivity in humans. J Med Genet. 2006;43:e40. doi: 10.1136/jmg.2005.036079.
    1. Hocking LJ, Smith BH, Jones GT, Reid DM, Strachan DP, Macfarlane GJ. Genetic variation in the beta2-adrenergic receptor but not catecholamine-O-methyltransferase predisposes to chronic pain: results from the 1958 British Birth Cohort Study. Pain. 2010;149:143–151. doi: 10.1016/j.pain.2010.01.023.
    1. Bond C, LaForge KS, Tian M, Melia D, Zhang S, Borg L, Gong J, Schluger J, Strong JA, Leal SM, Tischfield JA, Kreek MJ, Yu L. Single-nucleotide polymorphism in the human mu opioid receptor gene alters beta-endorphin binding and activity: possible implications for opiate addiction. Proc Natl Acad Sci USA. 1998;95:9608–9613. doi: 10.1073/pnas.95.16.9608.
    1. Wendel B, Hoehe MR. The human mu opioid receptor gene: 5' regulatory and intronic sequences. J Mol Med (Berl) 1998;76:525–532. doi: 10.1007/s001090050246.
    1. Walter C, Lotsch J. Meta-analysis of the relevance of the OPRM1 118A>G genetic variant for pain treatment. Pain. 2009;146:270–275. doi: 10.1016/j.pain.2009.07.013.
    1. Maier C, Baron R, Tölle TR, Binder A, Birbaumer N, Birklein F, Gierthmühlen J, Flor H, Geber C, Huge V, Krumova EK, Landwehrmeyer GB, Magerl W, Maihöfner C, Richter H, Rolke R, Scherens A, Schwarz A, Sommer C, Tronnier V, Uçeyler N, Valet M, Wasner G, Treede RD. Quantitative sensory testing in the German Research Network on Neuropathic Pain (DFNS): somatosensory abnormalities in 1236 patients with different neuropathic pain syndromes. Pain. 2010;150:439–450. doi: 10.1016/j.pain.2010.05.002.
    1. Frazer KA, Murray SS, Schork NJ, Topol EJ. Human genetic variation and its contribution to complex traits. Nat Rev Genet. 2009;10:241–251.
    1. Hardy J, Singleton A. Genomewide association studies and human disease. New Engl J Med. 2009;360:1759–1768. doi: 10.1056/NEJMra0808700.
    1. Consortium WTCC. Genome-wide association study of CNVs in 16,000 cases of eight common diseases and 3,000 shared controls. Nature. 2010;464:713–720. doi: 10.1038/nature08979.
    1. Gibson G. Rare and common variants: twenty arguments. Nat Rev Genet. 2011;13:135–145.
    1. Nelson MR, Wegmann D, Ehm MG, Kessner D, St Jean P, Verzilli C, Shen J, Tang Z, Bacanu SA, Fraser D, Warren L, Aponte J, Zawistowski M, Liu X, Zhang H, Zhang Y, Li J, Li Y, Li L, Woollard P, Topp S, Hall MD, Nangle K, Wang J, Abecasis G, Cardon LR, Zöllner S, Whittaker JC, Chissoe SL, Novembre J, Mooser V. An abundance of rare functional variants in 202 drug target genes sequenced in 14,002 people. Science. 2012;337:100–104. doi: 10.1126/science.1217876.
    1. Tennessen JA, Bigham AW, O'Connor TD, Fu W, Kenny EE, Gravel S, McGee S, Do R, Liu X, Jun G, Kang HM, Jordan D, Leal SM, Gabriel S, Rieder MJ, Abecasis G, Altshuler D, Nickerson DA, Boerwinkle E, Sunyaev S, Bustamante CD, Bamshad MJ, Akey JM. Evolution and functional impact of rare coding variation from deep sequencing of human exomes. Science. 2012;337:64–69. doi: 10.1126/science.1219240.
    1. Chesler EJ, Wilson SG, Lariviere WR, Rodriguez-Zas SL, Mogil JS. Influences of laboratory environment on behavior. Nat Neurosci. 2002;5:1101–1102. doi: 10.1038/nn1102-1101.
    1. Devor M, Gilad A, Arbilly M, Nissenbaum J, Yakir B, Raber P, Minert A, Pisante A, Darvasi A. Sex-specific variability and a 'cage effect' independently mask a neuropathic pain quantitative trait locus detected in a whole genome scan. Eur J Neurosci. 2007;26:681–688. doi: 10.1111/j.1460-9568.2007.05704.x.
    1. Mogil JS, Sorge RE, LaCroix-Fralish ML, Smith SB, Fortin A, Sotocinal SG, Ritchie J, Austin JS, Schorscher-Petcu A, Melmed K, Czerminski J, Bittong RA, Mokris JB, Neubert JK, Campbell CM, Edwards RR, Campbell JN, Crawley JN, Lariviere WR, Wallace MR, Sternberg WF, Balaban CD, Belfer I, Fillingim RB. Pain sensitivity and vasopressin analgesia are mediated by a gene-sex-environment interaction. Nat Neurosci. 2011;14:1569–1573. doi: 10.1038/nn.2941.
    1. Jones GT, Power C, Macfarlane GJ. Adverse events in childhood and chronic widespread pain in adult life: Results from the 1958 British Birth Cohort Study. Pain. 2009;143:92–96. doi: 10.1016/j.pain.2009.02.003.
    1. Maneyapanda SB, Venkatasubramanian A. Relationship between significant perinatal events and migraine severity. Pediatrics. 2005;116:e555–558. doi: 10.1542/peds.2005-0454.
    1. LaCroix-Fralish ML, Austin JS, Zheng FY, Levitin DJ, Mogil JS. Patterns of pain: meta-analysis of microarray studies of pain. Pain. 2011;152:1888–1898. doi: 10.1016/j.pain.2011.04.014.
    1. Denk F, McMahon SB. Chronic pain: emerging evidence for the involvement of epigenetics. Neuron. 2012;73:435–444. doi: 10.1016/j.neuron.2012.01.012.
    1. Kouzarides T. Chromatin modifications and their function. Cell. 2007;128:693–705. doi: 10.1016/j.cell.2007.02.005.
    1. Shakespear MR, Halili MA, Irvine KM, Fairlie DP, Sweet MJ. Histone deacetylases as regulators of inflammation and immunity. Trends Immunol. 2011;32:335–343. doi: 10.1016/j.it.2011.04.001.
    1. Francis YI, Fà M, Ashraf H, Zhang H, Staniszewski A, Latchman DS, Arancio O. Dysregulation of histone acetylation in the APP/PS1 mouse model of Alzheimer's disease. J Alzheimers Dis. 2009;18:131–139.
    1. Monsey MS, Ota KT, Akingbade IF, Hong ES, Schafe GE. Epigenetic alterations are critical for fear memory consolidation and synaptic plasticity in the lateral amygdala. PLoS One. 2011;6:e19958. doi: 10.1371/journal.pone.0019958.
    1. Haettig J, Stefanko DP, Multani ML, Figueroa DX, McQuown SC, Wood MA. HDAC inhibition modulates hippocampus-dependent long-term memory for object location in a CBP-dependent manner. Learn Mem. 2011;18:71–79. doi: 10.1101/lm.1986911.
    1. Haberland M, Montgomery RL, Olson EN. The many roles of histone deacetylases in development and physiology: implications for disease and therapy. Nat Rev Genet. 2009;10:32–42. doi: 10.1038/nrg2485.
    1. Bradner JE, West N, Grachan ML, Greenberg EF, Haggarty SJ, Warnow T, Mazitschek R. Chemical phylogenetics of histone deacetylases. Nat Chem Biol. 2010;6:238–243. doi: 10.1038/nchembio.313.
    1. Kilgore M, Miller CA, Fass DM, Hennig KM, Haggarty SJ, Sweatt JD, Rumbaugh G. Inhibitors of class 1 histone deacetylases reverse contextual memory deficits in a mouse model of Alzheimer's disease. Neuropsychopharmacology. 2010;35:870–880. doi: 10.1038/npp.2009.197.
    1. Rai M, Soragni E, Chou CJ, Barnes G, Jones S, Rusche JR, Gottesfeld JM, Pandolfo M. Two new pimelic diphenylamide HDAC inhibitors induce sustained frataxin upregulation in cells from Friedreich's ataxia patients and in a mouse model. PloS One. 2010;5:e8825. doi: 10.1371/journal.pone.0008825.
    1. Inks ES, Josey BJ, Jesinkey SR, Chou CJ. A novel class of small molecule inhibitors of HDAC6. ACS Chem Biol. 2012;7:331–339. doi: 10.1021/cb200134p.
    1. Kim MS, Akhtar MW, Adachi M, Mahgoub M, Bassel-Duby R, Kavalali ET, Olson EN, Monteggia LM. An essential role for histone deacetylase 4 in synaptic plasticity and memory formation. J Neurosci. 2012;32:10879–10886. doi: 10.1523/JNEUROSCI.2089-12.2012.
    1. Renthal W, Maze I, Krishnan V, Covington HE, Xiao G, Kumar A, Russo SJ, Graham A, Tsankova N, Kippin TE, Kerstetter KA, Neve RL, Haggarty SJ, McKinsey TA, Bassel-Duby R, Olson EN, Nestler EJ. Histone deacetylase 5 epigenetically controls behavioral adaptations to chronic emotional stimuli. Neuron. 2007;56:517–529. doi: 10.1016/j.neuron.2007.09.032.
    1. Wang L, Lv Z, Hu Z, Sheng J, Hui B, Sun J, Ma L. Chronic cocaine-induced H3 acetylation and transcriptional activation of CaMKIIalpha in the nucleus accumbens is critical for motivation for drug reinforcement. Neuropsychopharmacology. 2010;35:913–928. doi: 10.1038/npp.2009.193.
    1. Chiechio S, Zammataro M, Morales ME, Busceti CL, Drago F, Gereau RWt, Copani A, Nicoletti F. Epigenetic modulation of mGlu2 receptors by histone deacetylase inhibitors in the treatment of inflammatory pain. Mol Pharmacol. 2009;75:1014–1020. doi: 10.1124/mol.108.054346.
    1. 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. 2010;6:51. doi: 10.1186/1744-8069-6-51.
    1. Zhang Z, Cai YQ, Zou F, Bie B, Pan ZZ. Epigenetic suppression of GAD65 expression mediates persistent pain. Nat Med. 2011;17:1448–1455. doi: 10.1038/nm.2442.
    1. Tran L, Chaloner A, Sawalha AH, Greenwood Van-Meerveld B. Importance of epigenetic mechanisms in visceral pain induced by chronic water avoidance stress. Psychoneuroendocrinology. 2012. doi: 10.1016/j.psyneuen.2012.09.016.
    1. Kiguchi N, Kobayashi Y, Maeda T, Fukazawa Y, Tohya K, Kimura M, Kishioka S. 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. 2012;340:577–587. doi: 10.1124/jpet.111.187724.
    1. Zhu XY, Huang CS, Li Q, Chang RM, Song ZB, Zou WY, Guo QL. p300 exerts an epigenetic role in chronic neuropathic pain through its acetyltransferase activity in rats following chronic constriction injury (CCI). Mol Pain. 2012;8:84. doi: 10.1186/1744-8069-8-84.
    1. Guo JU, Ma DK, Mo H, Ball MP, Jang MH, Bonaguidi MA, Balazer JA, Eaves HL, Xie B, Ford E, Zhang K, Ming GL, Gao Y, Song H. Neuronal activity modifies the DNA methylation landscape in the adult brain. Nat Neurosci. 2011;14:1345–1351. doi: 10.1038/nn.2900.
    1. Hark AT, Schoenherr CJ, Katz DJ, Ingram RS, Levorse JM, Tilghman SM. CTCF mediates methylation-sensitive enhancer-blocking activity at the H19/Igf2 locus. Nature. 2000;405:486–489. doi: 10.1038/35013106.
    1. Bird A. DNA methylation patterns and epigenetic memory. Genes Dev. 2002;16:6–21. doi: 10.1101/gad.947102.
    1. Géranton SM, Morenilla-Palao C, Hunt SP. A role for transcriptional repressor methyl-CpG-binding protein 2 and plasticity-related gene serum- and glucocorticoid-inducible kinase 1 in the induction of inflammatory pain states. J Neurosci. 2007;27:6163–6173. doi: 10.1523/JNEUROSCI.1306-07.2007.
    1. 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. 2012;8:14. doi: 10.1186/1744-8069-8-14.
    1. Tajerian M, Alvarado S, Millecamps M, Dashwood T, Anderson KM, Haglund L, Ouellet J, Szyf M, Stone LS. DNA methylation of SPARC and chronic low back pain. Mol Pain. 2011;7:65. doi: 10.1186/1744-8069-7-65.
    1. Williams FM, Bansal AT, van Meurs JB, Bell JT, Meulenbelt I, Suri P, Rivadeneira F, Sambrook PN, Hofman A, Bierma-Zeinstra S, Menni C, Kloppenburg M, Slagboom PE, Hunter DJ, Macgregor AJ, Uitterlinden AG, Spector TD. Novel genetic variants associated with lumbar disc degeneration in northern Europeans: a meta-analysis of 4600 subjects. Ann Rheum Dis. 2012. doi:10.1136/annrheumdis-2012-201551.
    1. Viet CT, Ye Y, Dang D, Lam DK, Achdjian S, Zhang J, Schmidt BL. Re-expression of the methylated EDNRB gene in oral squamous cell carcinoma attenuates cancer-induced pain. Pain. 2011;152:2323–2332. doi: 10.1016/j.pain.2011.06.025.
    1. Wang Y, Liu C, Guo Q-L, Yan J-Q, Zhu X-Y, Huang C-S, Zou W-Y. 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. 2011;1418:64–69.
    1. Skene PJ, Illingworth RS, Webb S, Kerr AR, James KD, Turner DJ, Andrews R, Bird AP. Neuronal MeCP2 is expressed at near histone-octamer levels and globally alters the chromatin state. Mol Cell. 2010;37:457–468. doi: 10.1016/j.molcel.2010.01.030.
    1. Szyf M. Epigenetics, DNA methylation, and chromatin modifying drugs. Annu Rev Pharmacol Toxicol. 2009;49:243–263. doi: 10.1146/annurev-pharmtox-061008-103102.
    1. Garzon R, Marcucci G, Croce CM. Targeting microRNAs in cancer: rationale, strategies and challenges. Nat Rev Drug Discov. 2010;9:775–789. doi: 10.1038/nrd3179.
    1. Bartel DP. MicroRNAs: genomics, biogenesis, mechanism, and function. Cell. 2004;116:281–297. doi: 10.1016/S0092-8674(04)00045-5.
    1. Aldrich BT, Frakes EP, Kasuya J, Hammond DL, Kitamoto T. Changes in expression of sensory organ-specific microRNAs in rat dorsal root ganglia in association with mechanical hypersensitivity induced by spinal nerve ligation. Neuroscience. 2009;164:711–723. doi: 10.1016/j.neuroscience.2009.08.033.
    1. Bai G, Ambalavanar R, Wei D, Dessem D. Downregulation of selective microRNAs in trigeminal ganglion neurons following inflammatory muscle pain. Mol Pain. 2007;3:15. doi: 10.1186/1744-8069-3-15.
    1. Imai S, Saeki M, Yanase M, Horiuchi H, Abe M, Narita M, Kuzumaki N, Suzuki T. Change in microRNAs associated with neuronal adaptive responses in the nucleus accumbens under neuropathic pain. J Neurosci. 2011;31:15294–15299. doi: 10.1523/JNEUROSCI.0921-11.2011.
    1. Kusuda R, Cadetti F, Ravanelli MI, Sousa TA, Zanon S, De Lucca FL, Lucas G. Differential expression of microRNAs in mouse pain models. Mol Pain. 2011;7:17. doi: 10.1186/1744-8069-7-17.
    1. Poh KW, Yeo JF, Ong WY. MicroRNA changes in the mouse prefrontal cortex after inflammatory pain. Eur J Pain. 2011;15:801 e801–812.
    1. von Schack D, Agostino MJ, Murray BS, Li Y, Reddy PS, Chen J, Choe SE, Strassle BW, Li C, Bates B, Zhang L, Hu H, Kotnis S, Bingham B, Liu W, Whiteside GT, Samad TA, Kennedy JD, Ajit SK. Dynamic changes in the microRNA expression profile reveal multiple regulatory mechanisms in the spinal nerve ligation model of neuropathic pain. PLoS One. 2011;6:e17670. doi: 10.1371/journal.pone.0017670.
    1. Zhao J, Lee MC, Momin A, Cendan CM, Shepherd ST, Baker MD, Asante C, Bee L, Bethry A, Perkins JR, Nassar MA, Abrahamsen B, Dickenson A, Cobb BS, Merkenschlager M, Wood JN. Small RNAs control sodium channel expression, nociceptor excitability, and pain thresholds. J Neurosci. 2010;30:10860–10871. doi: 10.1523/JNEUROSCI.1980-10.2010.
    1. Favereaux A, Thoumine O, Bouali-Benazzouz R, Roques V, Papon MA, Salam SA, Drutel G, Leger C, Calas A, Nagy F, Landry M. Bidirectional integrative regulation of Cav1.2 calcium channel by microRNA miR-103: role in pain. EMBO J. 2011;30:3830–3841. doi: 10.1038/emboj.2011.249.
    1. Fossat P, Dobremez E, Bouali-Benazzouz R, Favereaux A, Bertrand SS, Kilk K, Leger C, Cazalets JR, Langel U, Landry M, Nagy F. Knockdown of L calcium channel subtypes: differential effects in neuropathic pain. J Neurosci. 2010;30:1073–1085. doi: 10.1523/JNEUROSCI.3145-09.2010.
    1. Betel D, Wilson M, Gabow A, Marks DS, Sander C. The resource: targets and expression. Nucleic Acids Res. 2008;36:D149–D153.

Source: PubMed

3
Abonneren