Browse > Article
http://dx.doi.org/10.18325/jkmr.2020.30.1.63

Epigenetic Modification in Chronic Pain: A Literature Review  

Song, Eun-Mo (Department of Clinical Korean Medicine, Graduate School, Kyung Hee University)
Cho, Hong-Seok (Department of Clinical Korean Medicine, Graduate School, Kyung Hee University)
Kim, Koh-Woon (Department of Korean Medicine Rehabilitation, College of Korean Medicine, Kyung Hee University)
Cho, Jae-Heung (Department of Korean Medicine Rehabilitation, College of Korean Medicine, Kyung Hee University)
Park, Hi-Joon (Acupuncture and Meridian Science Research Center, College of Korean Medicine, Kyung Hee University)
Song, Mi-Yeon (Department of Korean Medicine Rehabilitation, College of Korean Medicine, Kyung Hee University)
Publication Information
Journal of Korean Medicine Rehabilitation / v.30, no.1, 2020 , pp. 63-78 More about this Journal
Abstract
Objectives To review the epigenetic modifications involved in chronic pain and to improve individualized intervention for the chronic pain. Methods Focused literature review. Results Significant laboratory and clinical data support that epigenetic modifications have a potential role for development of chronic pain. Conclusions Epigenetic approach may identify mechanisms critical to the development of chronic pain after injury, and may provide new pathways and target mechanisms for future treatment and individualized medicine.
Keywords
Chronic pain; Epigenomics; Histones; MicroRNAs; DNA methylation;
Citations & Related Records
연도 인용수 순위
  • Reference
1 Zhang Z, Cai YQ, Zou F, Bie B, Pan ZZ. Epigenetic suppression of GAD65 expression mediates persistent pain. Nat Med. 2011;17(11):1448-55.   DOI
2 Burgess SE, Gardell LR, Ossipov MH, Malan TP Jr, Vanderah TW, Lai J, Porreca F. Time-dependent descending facilitation from the rostral ventromedial medulla maintains, but does not initiate, neuropathic pain. The Journal of Neuroscience: the Official Journal of the Society for Neuroscience. 2002;22(12):5129-36.   DOI
3 Nasca C, Xenos D, Barone Y, Caruso A, Scaccianoce S, Matrisciano F, Battaglia G, Mathe AA, Pittaluga A, Lionetto L, Simmaco M, Nicoletti F. L-acetylcarnitine causes rapid antidepressant effects through the epigenetic induction of mGlu2 receptors. Proceedings of the National Academy of Sciences of the United States of America. 2013;110(12):4804-9.   DOI
4 Tajerian M, Alvarado S, Millecamps M, Vachon P, Crosby C, Bushnell MC, Szyf M, Stone LS. Peripheral nerve injury is associated with chronic, reversible changes in global DNA methylation in the mouse prefrontal cortex. PloS One. 2013;8(1):e55259.   DOI
5 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. The Journal of Neuroscience: the Official Journal of the Society for Neuroscience. 2011;31(43):15294-9.   DOI
6 Poh KW, Yeo JF, Ong WY. MicroRNA changes in the mouse prefrontal cortex after inflammatory pain. Eur J Pain. 2011;15(8):801 e1-12.
7 Bushnell MC, Ceko M, Low LA. Cognitive and emotional control of pain and its disruption in chronic pain. Nature reviews Neuroscience. 2013;14(7):502-11.   DOI
8 Loeser JD, Melzack R. Pain: an overview. Lancet. 1999;353(9164):1607-9.   DOI
9 Petrenko AB, Yamakura T, Baba H, Shimoji K. The role of N-methyl-D-aspartate (NMDA) receptors in pain: a review. Anesth Analg. 2003;97(4):1108-16.
10 Sherman SM, Guillery RW. Functional organization of thalamocortical relays. J Neurophysiol. 1996;76(3):1367-95.   DOI
11 Voisin DL, Guy N, Chalus M, Dallel R. Nociceptive stimulation activates locus coeruleus neurones projecting to the somatosensory thalamus in the rat. J Physiol. 2005;566(Pt 3):929-37.   DOI
12 Apkarian AV, Bushnell MC, Treede RD, Zubieta JK. Human brain mechanisms of pain perception and regulation in health and disease. Eur J Pain. 2005;9(4):463-84.   DOI
13 Heinricher MM, Tavares I, Leith JL, Lumb BM. Descending control of nociception: Specificity, recruitment and plasticity. Brain Res Rev. 2009;60(1):214-25.   DOI
14 Tracey I, Mantyh PW. The cerebral signature for pain perception and its modulation. Neuron. 2007;55(3):377-91.   DOI
15 Zhuo M, Gebhart GF. Biphasic modulation of spinal nociceptive transmission from the medullary raphe nuclei in the rat. J Neurophysiol. 1997;78(2):746-58.   DOI
16 Carlson JD, Maire JJ, Martenson ME, Heinricher MM. Sensitization of pain-modulating neurons in the rostral ventromedial medulla after peripheral nerve injury. J Neurosci. 2007;27(48):13222-31.   DOI
17 Buchheit T, Van de Ven T, Shaw A. Epigenetics and the transition from acute to chronic pain. Pain Med. 2012;13(11):1474-90.   DOI
18 Cherng CH, Lee KC, Chien CC, Chou KY, Cheng YC, Hsin ST, Lee SO, Shen CH, Tsai RY, Wong CS. Baicalin ameliorates neuropathic pain by suppressing HDAC1 expression in the spinal cord of spinal nerve ligation rats. J Formos Med Assoc. 2014;113(8):513-20.   DOI
19 Bai G, Wei D, Zou S, Ren K, Dubner R. Inhibition of class II histone deacetylases in the spinal cord attenuates inflammatory hyperalgesia. Molecular Pain. 2010;6:51.   DOI
20 Ni J, Gao Y, Gong S, Guo S, Hisamitsu T, Jiang X. Regulation of mu-opioid type 1 receptors by microRNA134 in dorsal root ganglion neurons following peripheral inflammation. Eur J Pain. 2013;17(3):313-23.   DOI
21 Geranton SM. Targeting epigenetic mechanisms for pain relief. Current Opinion in Pharmacology. 2012;12(1):35-41.   DOI
22 Fu SP, He SY, Xu B, Hu CJ, Lu SF, Shen WX, Huang Y, Hong H, Li Q, Wang N, Liu XL, Liang F, Zhu BM. Acupuncture promotes angiogenesis after myocardial ischemia through H3K9 acetylation regulation at VEGF gene. PloS One. 2014;9(4):e94604.   DOI
23 Wang JY, Li H, Ma CM, Wang JL, Lai XS, Zhou SF. MicroRNA profiling response to acupuncture therapy in spontaneously hypertensive rats. Evid Based Complement Alternat Med. 2015;2015:204367.
24 Wang JY, Li H, Ma CM, Wang JL, Lai XS, Zhou SF. Acupuncture may exert its therapeutic effect through microRNA-339/Sirt2/NFkappaB/FOXO1 axis. BioMed Research International. 2015;2015:249013.   DOI
25 Wang N, Lu SF, Chen H, Wang JF, Fu SP, Hu CJ, Yang Y, Liang FR, Zhu BM. A protocol of histone modification-based mechanistic study of acupuncture in patients with stable angina pectoris. BMC Complement Altern Med. 2015;15:139.   DOI
26 Zhao HL, Sui Y, Qiao CF, Yip KY, Leung RK, Tsui SK, Lee HM, Wong HK, Zhu X, Siu JJ, He L, Guan J, Liu LZ, Xu HX, Tong PC, Chan JC. Sustained antidiabetic effects of a berberine-containing Chinese herbal medicine through regulation of hepatic gene expression. Diabetes. 2012;61(4):933-43.   DOI
27 Hsieh HY, Chiu PH, Wang SC. Epigenetics in traditional chinese pharmacy: a bioinformatic study at pharmacopoeia scale. Evid Based Complement Alternat Med. 2011;2011:816714.
28 Hsieh HY, Chiu PH, Wang SC. Histone modifications and traditional Chinese medicinals. BMC Complement Altern Med. 2013;13:115.   DOI
29 Hong M, Wang N, Tan HY, Tsao SW, Feng Y. MicroRNAs and Chinese medicinal herbs: new possibilities in cancer therapy. Cancers (Basel). 2015;7(3):1643-57.   DOI
30 Zhang Y, Wang YH, Zhang XH, Ge HY, Arendt-Nielsen L, Shao JM, Yue SW. Proteomic analysis of differential proteins related to the neuropathic pain and neuroprotection in the dorsal root ganglion following its chronic compression in rats. Exp Brain Res. 2008;189(2):199-209.   DOI
31 Bai L, Tian J, Zhong C, Xue T, You Y, Liu Z, Chen P, G ong Q, A i L, Q in W, Dai J , Liu Y. A cupuncture modulates temporal neural responses in wide brain networks: evidence from fMRI study. Molecular Pain. 2010;6:73.   DOI
32 Huang W, Pach D, Napadow V, Park K, Long X, Neumann J, Maeda Y, Nierhaus T, Liang F, Witt CM. Characterizing acupuncture stimuli using brain imaging with FMRI--a systematic review and meta-analysis of the literature. PloS One. 2012;7(4):e32960.   DOI
33 Gao YH, Chen SP, Wang JY, Qiao LN, Meng FY, Xu QL, Liu JL. Differential proteomics analysis of the analgesic effect of electroacupuncture intervention in the hippocampus following neuropathic pain in rats. BMC Complement Altern Med. 2012;12:241.
34 Woolf CJ, Salter MW. Neuronal plasticity: increasing the gain in pain. Science. 2000;288(5472):1765-9.   DOI
35 Sun J , Shao XM, F ang F, S hen Z, Wu YY, Fang J Q. Electroacupuncture alleviates retrieval of pain memory and its effect on phosphorylation of cAMP response element-binding protein in anterior cingulate cortex in rats. Behav Brain Funct. 2015;11:9.   DOI
36 Gao Y, Chen S , Xu Q, Yu K, Wang J , Qiao L , Meng F, Liu J. Proteomic analysis of differential proteins related to anti-nociceptive effect of electroacupuncture in the hypothalamus following neuropathic pain in rats. Neurochemical Research. 2013;38(7):1467-78.   DOI
37 Millan MJ. Descending control of pain. Progress in Neurobiology. 2002;66(6):355-474.   DOI
38 Jasmin L, Rabkin SD, Granato A, Boudah A, Ohara PT. Analgesia and hyperalgesia from GABA-mediated modulation of the cerebral cortex. Nature. 2003;424(6946):316-20.   DOI
39 Yaksh TL. Opioid receptor systems and the endorphins: a review of their spinal organization. J Neurosurg. 1987;67(2):157-76.   DOI
40 Gureje O, Von Korff M, Simon GE, Gater R. Persistent pain and well-being: a World Health Organization Study in Primary Care. JAMA. 1998;280(2):147-51.   DOI
41 Muralidharan A, Smith MT. Pain, analgesia and genetics. The Journal of Pharmacy and Pharmacology. 2011;63(11):1387-400.   DOI
42 Smith MT, Muralidharan A. Pharmacogenetics of pain and analgesia. Clinical genetics. 2012;82(4):321-30.   DOI
43 Seo S, Grzenda A, Lomberk G, Ou XM, Cruciani RA, Urrutia R. Epigenetics: a promising paradigm for better understanding and managing pain. The Journal of Pain: Official Journal of the American Pain Society. 2013;14(6):549-57.   DOI
44 Kuner R. Central mechanisms of pathological pain. Nat Med. 2010;16(11):1258-66.   DOI
45 Su RC, Becker AB, Kozyrskyj AL, Hayglass KT. Epigenetic regulation of established human type 1 versus type 2 cytokine responses. J Allergy Clin Immunol. 2008;121(1):57-63 e3.   DOI
46 Besson JM. The neurobiology of pain. Lancet. 1999;353(9164):1610-5.   DOI
47 Riccio A. Dynamic epigenetic regulation in neurons: enzymes, stimuli and signaling pathways. Nature Neuroscience. 2010;13(11):1330-7.   DOI
48 Hammer P, Banck MS, Amberg R, Wang C, Petznick G, Luo S, Khrebtukova I, Schroth GP, Beyerlein P, Beutler AS. mRNA-seq with agnostic splice site discovery for nervous system transcriptomics tested in chronic pain. Genome Res. 2010;20(6):847-60.   DOI
49 Gangadharan V, Kuner R. Pain hypersensitivity mechanisms at a glance. Dis Model Mech. 2013;6(4):889-95.   DOI
50 Denk F, McMahon SB. Chronic pain: emerging evidence for the involvement of epigenetics. Neuron. 2012;73(3):435-44.   DOI
51 McCall CE, El Gazzar M, Liu T, Vachharajani V, Yoza B. Epigenetics, bioenergetics, and microRNA coordinate gene-specific reprogramming during acute systemic inflammation. Journal of Leukocyte Biology. 2011;90(3):439-46.   DOI
52 Fukuoka T, Kondo E, Dai Y, Hashimoto N, Noguchi K. Brain-derived neurotrophic factor increases in the uninjured dorsal root ganglion neurons in selective spinal nerve ligation model. The Journal of Neuroscience: the Official Journal of the Society for Neuroscience. 2001;21(13):4891-900.   DOI
53 Shakespear MR, Halili MA, Irvine KM, Fairlie DP, Sweet MJ. Histone deacetylases as regulators of inflammation and immunity. Trends Immunol. 2011;32(7):335-43.   DOI
54 Bruce AW, Donaldson IJ, Wood IC, Yerbury SA, Sadowski MI, Chapman M, Gottgens B, Buckley NJ. Genome-wide analysis of repressor element 1 silencing transcription factor/neuron-restrictive silencing factor (REST/NRSF) target genes. Proc Natl Acad Sci U S A. 2004;101(28):10458-63.   DOI
55 Uchida H, Ma L, Ueda H. Epigenetic gene silencing underlies C-fiber dysfunctions in neuropathic pain. The Journal of Neuroscience: the Official Journal of the Society for Neuroscience. 2010;30(13):4806-14.   DOI
56 Uchida H, Sasaki K, Ma L, Ueda H. Neuron-restrictive silencer factor causes epigenetic silencing of Kv4.3 gene after peripheral nerve injury. Neuroscience. 2010;166(1):1-4.   DOI
57 Crofford LJ. Chronic pain: where the body meets the brain. Transactions of the American Clinical and Climatological Association. 2015;126:167-83.
58 Woolf CJ. Central sensitization: implications for the diagnosis and treatment of pain. Pain. 2011;152(3 Suppl):S2-15.   DOI
59 Kawasaki Y, Kohno T, Zhuang ZY, Brenner GJ, Wang H, Van Der Meer C, Befort K, Woolf CJ, Ji RR. Ionotropic and metabotropic receptors, protein kinase A, protein kinase C, and Src contribute to C-fiber-induced ERK activation and cAMP response element-binding protein phosphorylation in dorsal horn neurons, leading to central sensitization. The Journal of Neuroscience: the Official Journal of the Society for Neuroscience. 2004;24(38):8310-21.   DOI
60 Geranton 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. The Journal of Neuroscience: the Official Journal of the Society for Neuroscience. 2007;27(23):6163-73.   DOI
61 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(2):71-9.   DOI
62 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(3):577-87.   DOI
63 Chiechio S, Caricasole A, Barletta E, Storto M, Catania MV, Copani A, Vertechy M, Nicolai R, Calvani M, Melchiorri D, Nicoletti F. L-Acetylcarnitine induces analgesia by selectively up-regulating mGlu2 metabotropic glutamate receptors. Molecular Pharmacology. 2002;61(5):989-96.   DOI
64 Imai S, Ikegami D, Yamashita A, Shimizu T, Narita M, Niikura K, Furuya M, Kobayashi Y, Miyashita K, Okutsu D, Kato A, Nakamura A, Araki A, Omi K, Nakamura M, James Okano H, Okano H, Ando T, Takeshima H, Ushijima T, Kuzumaki N, Suzuki T. Epigenetic transcriptional activation of monocyte chemotactic protein 3 contributes to long-lasting neuropathic pain. Brain: a Journal of Neurology. 2013;136(Pt 3):828-43.   DOI
65 Woolf CJ. Pain: moving from symptom control toward mechanism-specific pharmacologic management. Annals of Internal Medicine. 2004;140(6):441-51.   DOI
66 Bali KK, Kuner R. Noncoding RNAs: key molecules in understanding and treating pain. Trends in Molecular Medicine. 2014;20(8):437-48.   DOI
67 Woolf CJ, Decosterd I. Implications of recent advances in the understanding of pain pathophysiology for the assessment of pain in patients. Pain. 1999;Suppl 6:S141-7.
68 Kissin I. The development of new analgesics over the past 50 years: a lack of real breakthrough drugs. Anesthesia and Analgesia. 2010;110(3):780-9.   DOI
69 Ito K, Barnes PJ, Adcock IM. Glucocorticoid receptor recruitment of histone deacetylase 2 inhibits interleukin-1beta-induced histone H4 acetylation on lysines 8 and 12. Mol Cell Biol. 2000;20(18):6891-903.   DOI
70 Leoni F, Zaliani A, Bertolini G, Porro G, Pagani P, Pozzi P, Dona G, Fossati G, Sozzani S, Azam T, Bufler P, Fantuzzi G, Goncharov I, Kim SH, Pomerantz BJ, Reznikov LL, Siegmund B, Dinarello CA, Mascagni P. The antitumor histone deacetylase inhibitor suberoylanilide hydroxamic acid exhibits antiinflammatory properties via suppression of cytokines. Proc Natl Acad Sci U S A. 2002;99(5):2995-3000.   DOI
71 Chiechio S, Copani A, Zammataro M, Battaglia G, Gereau RWt, Nicoletti F. Transcriptional regulation of type-2 metabotropic glutamate receptors: an epigenetic path to novel treatments for chronic pain. Trends Pharmacol Sci. 2010;31(4):153-60.   DOI
72 Kim CS, Hwang CK, Choi HS, Song KY, Law PY, Wei LN, Loh HH. Neuron-restrictive silencer factor (NRSF) functions as a repressor in neuronal cells to regulate the mu opioid receptor gene. The Journal of Biological Chemistry. 2004;279(45):46464-73.   DOI
73 Crow M, Denk F, McMahon SB. Genes and epigenetic processes as prospective pain targets. Genome Med. 2013;5(2):12.   DOI
74 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. Molecular Pain. 2008;4:35.   DOI
75 Kouzarides T. Chromatin modifications and their function. Cell. 2007;128(4):693-705.   DOI
76 Javierre BM, Fernandez AF, Richter J, Al-Shahrour F, Martin-Subero JI, Rodriguez-Ubreva J, Berdasco M, Fraga MF, O'Hanlon TP, Rider LG, Jacinto FV, Lopez-Longo FJ, Dopazo J, Forn M, Peinado MA, Carreno L, Sawalha AH, Harley JB, Siebert R, Esteller M, Miller FW, Ballestar E. Changes in the pattern of DNA methylation associate with twin discordance in systemic lupus erythematosus. Genome Research. 2010;20(2):170-9.   DOI
77 Descalzi G, Ikegami D, Ushijima T, Nestler EJ, Zachariou V, Narita M. Epigenetic mechanisms of chronic pain. Trends Neurosci. 2015;38(4):237-46.   DOI
78 Wu C, Morris JR. Genes, genetics, and epigenetics: a correspondence. Science. 2001;293(5532):1103-5.   DOI
79 Guan Z, Giustetto M, Lomvardas S, Kim JH, Miniaci MC, Schwartz JH, Thanos D, Kandel ER. Integration of long-term-memory-related synaptic plasticity involves bidirectional regulation of gene expression and chromatin structure. Cell. 2002;111(4):483-93.   DOI
80 Gregory PD, Wagner K, Horz W. Histone acetylation and chromatin remodeling. Experimental Cell Research. 2001;265(2):195-202.   DOI
81 LaCroix-Fralish ML, Austin JS, Zheng FY, Levitin DJ, Mogil JS. Patterns of pain: meta-analysis of microarray studies of pain. Pain. 2011;152(8):1888-98.   DOI
82 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.
83 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.
84 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(10):2323-32.   DOI
85 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(32):10860-71.   DOI
86 Kynast KL, Russe OQ, Moser CV, Geisslinger G, Niederberger E. Modulation of central nervous system-specific microRNA-124a alters the inflammatory response in the formalin test in mice. Pain. 2013;154(3):368-76.   DOI
87 Sakai A, Suzuki H. Nerve injury-induced upregulation of miR-21 in the primary sensory neurons contributes to neuropathic pain in rats. Biochemical and Biophysical Research Communications. 2013;435(2):176-81.   DOI
88 Willemen HL, Huo XJ, Mao-Ying QL, Zijlstra J, Heijnen CJ, Kavelaars A. MicroRNA-124 as a novel treatment for persistent hyperalgesia. Journal of Neuroinflammation. 2012;9:143.   DOI
89 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. The EMBO Journal. 2011;30(18):3830-41.   DOI
90 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(3):1073-85.   DOI
91 Sakai A, Saitow F, Miyake N, Miyake K, Shimada T, Suzuki H. miR-7a alleviates the maintenance of neuropathic pain through regulation of neuronal excitability. Brain: A Journal of Neurology. 2013;136(Pt 9):2738-50.   DOI
92 Chen HP, Zhou W, Kang LM, Yan H, Zhang L, Xu BH, Cai WH. Intrathecal miR-96 inhibits Nav1.3 expression and alleviates neuropathic pain in rat following chronic construction injury. Neurochemical Research. 2014;39(1):76-83.   DOI
93 Baliki MN, Petre B, Torbey S, Herrmann KM, Huang L, Schnitzer TJ, Fields HL, Apkarian AV. Corticostriatal functional connectivity predicts transition to chronic back pain. Nature Neuroscience. 2012;15(8):1117-9.   DOI
94 Rainville P, Bushnell MC, Duncan GH. Representation of acute and persistent pain in the human CNS: potential implications for chemical intolerance. Annals of the New York Academy of Sciences. 2001;933:130-41.   DOI
95 Apkarian AV, Sosa Y, Sonty S, Levy RM, Harden RN, Parrish TB, Gitelman DR. Chronic back pain is associated with decreased prefrontal and thalamic gray matter density. J Neurosci. 2004;24(46):10410-5.   DOI
96 Porreca F, Ossipov MH, Gebhart GF. Chronic pain and medullary descending facilitation. Trends in Neurosciences. 2002;25(6):319-25.   DOI
97 Bartel DP. MicroRNAs: genomics, biogenesis, mechanism, and function. Cell. 2004;116(2):281-97.   DOI
98 Fan S, Zhang X. CpG island methylation pattern in different human tissues and its correlation with gene expression. Biochemical and Biophysical Research Communications. 2009;383(4):421-5.   DOI