Chronic pain after lower abdominal surgery: do catechol-O-methyl transferase/opioid receptor μ-1 polymorphisms contribute?

Yuri Kolesnikov, Boris Gabovits, Ariel Levin, Andres Veske, Li Qin, Feng Dai, Inna Belfer, Yuri Kolesnikov, Boris Gabovits, Ariel Levin, Andres Veske, Li Qin, Feng Dai, Inna Belfer

Abstract

Background: Preoperative pain, type of operation and anesthesia, severity of acute postoperative pain, and psychosocial factors have been identified as risk factors for chronic postsurgical pain (CPP). Recently, it has been suggested that genetic factors also contribute to CPP. In this study, we aimed to determine whether the catechol-O-methyl transferase (COMT) and opioid receptor μ-1 (OPRM1) common functional polymorphisms rs4680 and rs1799971 were associated with the incidence, intensity, or duration of CPP in patients after lower abdominal surgery.

Methods: One hundred and two patients with American Society of Anesthesiologists (ASA) physical status I/II underwent either abdominal radical prostatectomy (n = 45) or hysterectomy (n = 57). The incidences of CPP in the pelvic and scar areas were evaluated in all patients three months after surgery.

Results: Thirty-five (34.3%) patients experienced CPP after lower abdominal surgery. Within this group, six (17.1%) patients demonstrated symptoms of neuropathic pain. For COMT rs4680, 22 (21.6%) patients had Met158Met, 55 (53.9%) patients had Val158Met, and 25 (24.5%) patients had Val158Val. No association was found between CPP phenotypes (incidence, intensity, and duration) and different rs4680 genotypes. For OPRM1 rs1799971, only CPP patients carrying at least one copy of the G allele had higher pain intensity than A118A carriers (p=0.02). No associations with other phenotypes were found. No combined effect of COMT/OPRM1 polymorphisms on CPP phenotypes was observed.

Conclusions: OPRM1 genotype influences CPP following lower abdominal surgery. COMT didn't affect CPP, suggesting its potential modality-specific effects on human pain.

References

    1. Kehlet H, Jensen T, Woolf C. Persistent postsurgical pain: risk factors and prevention. Lancet. 2006;367:1618–1625. doi: 10.1016/S0140-6736(06)68700-X.
    1. Schreiber KL, Martel MO, Shnol H, Shaffer JR, Greco C, Viray N, Taylor LN, McLaughlin M, Brufsky A, Ahrendt G, Bovbjerg D, Edwards RR, Belfer I. Persistent pain in postmastectomy patients: comparison of psychophysical, medical, surgical, and psychosocial characteristics between patients with and without pain. Pain. 2012. Epud ahead of print.
    1. Perkins FM, Kehlet H. Chronic pain as an outcome of surgery: review of predictive factors. Anesthesiology. 2000;93:1123–1133. doi: 10.1097/00000542-200010000-00038.
    1. Hartvigsen J, Christensen K, Frederiksen H, Pedersen HC. Genetic and environmental contributions to back pain in old age: a study of 2,108 danish twins aged 70 and older. Spine. 2004;29:897–901. doi: 10.1097/00007632-200404150-00015.
    1. Young E, Lariviere W, Belfer I. Genetic basis of pain variability: recent advances. J Med Genet. 2012;49:1–9. doi: 10.1136/jmedgenet-2011-100386.
    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. Costigan M, Belfer I, Griffin RS, Dai F, Barrett LB, Coppola G, Wu T, Kiselycznyk C, Poddar M, Lu Y, Diatchenko L, Smith S, Cobos EJ, Zaykin D, Allchorne A, Gershon E, Livneh J, Shen PH, Nikolajsen L, Karppinen J, Männikkö M, Kelempisioti A, Goldman D, Maixner W, Geschwind DH, Max MB, Seltzer Z, Woolf CJ. Multiple chronic pain states are associated with a common amino acid-changing allele in KCNS1. Brain. 2010;133(9):2519–2527. doi: 10.1093/brain/awq195.
    1. Belfer I, Segall S. COMT genetic variants and pain. Drugs Today (Barc) 2011;47:457–467. doi: 10.1358/dot.2011.47.6.1611895.
    1. Diatchenko L, Slade GD, Nackley AG, Bhalang K, Sigurdsson A, Belfer I, Goldman D, Xu K, Shabalina SA, Shagin D, Max MB, Makarov SS, Maixner W. Genetic basis for individual variations in pain perception and the development of a chronic pain condition. Hum Mol Genet. 2005;14:135–143.
    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(5610):1240–1243. doi: 10.1126/science.1078546.
    1. Fillingim RB, Kaplan L, Staud R, Ness TJ, Glover TL, Campbell CM, Mogil JS, Wallace MR. The A118G single nucleotide polymorphism of the mu-opioid receptor gene (OPRM1) is associated with pressure pain sensitivity in humans. J Pain. 2005;6:159–167. doi: 10.1016/j.jpain.2004.11.008.
    1. Kolesnikov Y, Gabovits B, Levin A, Voiko E, Veske A. Combined catechol-O-methyltransferase and mu-opioid receptor gene polymorphisms affect morphine postoperative analgesia and central side effects. Anesth Analg. 2011;112(2):448–453. doi: 10.1213/ANE.0b013e318202cc8d.
    1. Macrae WA. Chronic pain after surgery. Br J Anaesth. 2001;87:88–98. doi: 10.1093/bja/87.1.88.
    1. Brandsborg B, Nikolajsen L, Hansen CT, Kehlet H, Jensen TS. Risk factors for chronic pain after hysterectomy: a nationwide questionnaire and database study. Anesthesiology. 2007;106(5):1003–1012. doi: 10.1097/01.anes.0000265161.39932.e8.
    1. Mongardon N, Pinton-Gonnet C, Szekely B, Michel-Cherqui M, Dreyfus JF, Fischler M. Assessment of chronic pain after thoracotomy: a 1-year prevalence study. Clin J Pain. 2011;27(8):677–681. doi: 10.1097/AJP.0b013e31821981a3.
    1. Wylde V, Hewlett S, Learmonth ID, Dieppe P. Persistent pain after joint replacement: prevalence, sensory qualities, and postoperative determinants. Pain. 2011;152(3):566–572. doi: 10.1016/j.pain.2010.11.023.
    1. Voscopoulos C, Lema M. When does acute pain become chronic? Br J Anaesth. 2010;105:69–85. doi: 10.1093/bja/aeq133.
    1. Fayad JN, Linthicum FH Jr. Symptomatic postsurgical traumatic neuromas. Otol Neurotol. 2009;30(7):981–984. doi: 10.1097/MAO.0b013e3181b4ef04.
    1. Flatters SJ. Effect of analgesic standards on persistent postoperative pain evoked by skin/muscle incision and retraction (SMIR) Neurosci Lett. 2010;14(477(1)):43–47.
    1. Wildgaard K, Ravn J, Kehlet H. Chronic post-thoracotomy pain: a critical review of pathogenic mechanisms and strategies for prevention. Eur J Cardiothorac Surg. 2009;36(1):170–180. doi: 10.1016/j.ejcts.2009.02.005.
    1. Linderoth G, Kehlet H, Aasvang EK, Werner MU. Neurophysiological characterization of persistent pain after laparoscopic inguinal hernia repair. Hernia. 2011;15(5):521–529. doi: 10.1007/s10029-011-0815-z.
    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. Nackley AG, Tan KS, Fecho K, Flood P, Diatchenko L, Maixner W. Catechol-O-methyltransferase inhibition increases pain sensitivity through activation of both beta2- and beta3-adrenergic receptors. Pain. 2007;128(3):199–208. doi: 10.1016/j.pain.2006.09.022.
    1. Dai F, Belfer I, Schwartz CE, Banco R, Martha JF, Tighioughart H, Tromanhauser SG, Jenis LG, Kim DH. Association of catechol-O-methyltransferase genetic variants with outcome in patients undergoing surgical treatment for lumbar degenerative disc disease. Spine J. 2010;10(11):949–957. doi: 10.1016/j.spinee.2010.07.387.
    1. Segall SK, Maixner W, Belfer I, Wiltshire T, Seltzer Z, Diatchenko L. Janus molecule I: dichotomous effects of COMT in neuropathic vs nociceptive pain modalities. CNS Neurol Disord Drug Targets. 2012;11(3):222–235. doi: 10.2174/187152712800672490.
    1. Tchivileva IE, Lim PF, Smith SB, Slade GD, Diatchenko L, McLean SA, Maixner W. Effect of catechol-O-methyltransferase polymorphism on response to propranolol therapy in chronic musculoskeletal pain: a randomized, double-blind, placebo-controlled, crossover pilot study. Pharmacogenet Genomics. 2010;20(4):239–248.
    1. Chou WY, Yang LC, Lu HF, Ko JY, Wang CH, Lin SH, Lee TH, Concejero A, Hsu CJ. Association of mu-opioid receptor gene polymorphism (A118G) with variations in morphine consumption for analgesia after total knee arthroplasty. Acta Anaesthesiologica Scand. 2006;50:787–792. doi: 10.1111/j.1399-6576.2006.01058.x.
    1. Chou WY, Wang CH, Liu PH, Liu CC, Tseng CC, Jawan B. Human opioid receptor A118G polymorphism affects intravenous patient-controlled analgesia morphine consumption after total abdominal hysterectomy. Anesthesiology. 2006;105(2):334–337. doi: 10.1097/00000542-200608000-00016.
    1. International Association for the Study of Pain. Pain. 1986. pp. 1–225.
    1. Belfer I, Dai F. Phenotyping and genotyping neuropathic pain. Curr Pain Headache Rep. 2010;4(3):203–212.
    1. Attal N, Perrot S, Fermanian J, Bouhassira D. The neuropathic components of chronic low back pain: a prospective multicenter study using the DN4 Questionnaire. J Pain. 2011;12(10):1080–1087. doi: 10.1016/j.jpain.2011.05.006.
    1. Spallone V, Morganti R, D'Amato C, Greco C, Cacciotti L, Marfia GA. Validation of DN4 as a screening tool for neuropathic pain in painful diabetic polyneuropathy. Diabet Med. 2012;29(5):578–585. doi: 10.1111/j.1464-5491.2011.03500.x.
    1. Purcell S, Neale B, Todd-Brown K, Thomas L, Ferreira MAR, Bender D, Maller J, Sklar P, de Bakker PIW, Daly MJ, Sham PC. PLINK: a toolset for whole-genome association and population-based linkage analysis. Am J Hum Genet. 2007;81(3):559–575. doi: 10.1086/519795.

Source: PubMed

3
Abonneren