Cargando…

DNA methylation at the mu-1 opioid receptor gene (OPRM1) promoter predicts preoperative, acute, and chronic postsurgical pain after spine fusion

INTRODUCTION: The perioperative pain experience shows great interindividual variability and is difficult to predict. The mu-1 opioid receptor gene (OPRM1) is known to play an important role in opioid-pain pathways. Since deoxyribonucleic acid (DNA) methylation is a potent repressor of gene expressio...

Descripción completa

Detalles Bibliográficos
Autores principales: Chidambaran, Vidya, Zhang, Xue, Martin, Lisa J, Ding, Lili, Weirauch, Matthew T, Geisler, Kristie, Stubbeman, Bobbie L, Sadhasivam, Senthilkumar, Ji, Hong
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Dove Medical Press 2017
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5432115/
https://www.ncbi.nlm.nih.gov/pubmed/28533693
http://dx.doi.org/10.2147/PGPM.S132691
_version_ 1783236574161403904
author Chidambaran, Vidya
Zhang, Xue
Martin, Lisa J
Ding, Lili
Weirauch, Matthew T
Geisler, Kristie
Stubbeman, Bobbie L
Sadhasivam, Senthilkumar
Ji, Hong
author_facet Chidambaran, Vidya
Zhang, Xue
Martin, Lisa J
Ding, Lili
Weirauch, Matthew T
Geisler, Kristie
Stubbeman, Bobbie L
Sadhasivam, Senthilkumar
Ji, Hong
author_sort Chidambaran, Vidya
collection PubMed
description INTRODUCTION: The perioperative pain experience shows great interindividual variability and is difficult to predict. The mu-1 opioid receptor gene (OPRM1) is known to play an important role in opioid-pain pathways. Since deoxyribonucleic acid (DNA) methylation is a potent repressor of gene expression, DNA methylation was evaluated at the OPRM1 promoter, as a predictor of preoperative, acute, and chronic postsurgical pain (CPSP). METHODS: A prospective observational cohort study was conducted in 133 adolescents with idiopathic scoliosis undergoing spine fusion under standard protocols. Data regarding pain, opioid consumption, anxiety, and catastrophizing (using validated questionnaires) were collected before and 2–3 months postsurgery. Outcomes evaluated were preoperative pain, acute postoperative pain (area under curve [AUC] for pain scores over 48 hours), and CPSP (numerical rating scale >3/10 at 2–3 months postsurgery). Blood samples collected preoperatively were analyzed for DNA methylation by pyrosequencing of 22 CpG sites at the OPRM1 gene promoter. The association of each pain outcome with the methylation percentage of each CpG site was assessed using multivariable regression, adjusting for significant (P<0.05) nongenetic variables. RESULTS: Majority (83%) of the patients reported no pain preoperatively, while CPSP occurred in 36% of the subjects (44/121). Regression on dichotomized preoperative pain outcome showed association with methylation at six CpG sites (1, 3, 4, 9, 11, and 17) (P<0.05). Methylation at CpG sites 4, 17, and 18 was associated with higher AUC after adjusting for opioid consumption and preoperative pain score (P<0.05). After adjusting for postoperative opioid consumption and preoperative pain score, methylation at CpG sites 13 and 22 was associated with CPSP (P<0.05). DISCUSSION: Novel CPSP biomarkers were identified in an active regulatory region of the OPRM1 gene that binds multiple transcription factors. Inhibition of binding by DNA methylation potentially decreases the OPRM1 gene expression, leading to a decreased response to endogenous and exogenous opioids, and an increased pain experience.
format Online
Article
Text
id pubmed-5432115
institution National Center for Biotechnology Information
language English
publishDate 2017
publisher Dove Medical Press
record_format MEDLINE/PubMed
spelling pubmed-54321152017-05-22 DNA methylation at the mu-1 opioid receptor gene (OPRM1) promoter predicts preoperative, acute, and chronic postsurgical pain after spine fusion Chidambaran, Vidya Zhang, Xue Martin, Lisa J Ding, Lili Weirauch, Matthew T Geisler, Kristie Stubbeman, Bobbie L Sadhasivam, Senthilkumar Ji, Hong Pharmgenomics Pers Med Original Research INTRODUCTION: The perioperative pain experience shows great interindividual variability and is difficult to predict. The mu-1 opioid receptor gene (OPRM1) is known to play an important role in opioid-pain pathways. Since deoxyribonucleic acid (DNA) methylation is a potent repressor of gene expression, DNA methylation was evaluated at the OPRM1 promoter, as a predictor of preoperative, acute, and chronic postsurgical pain (CPSP). METHODS: A prospective observational cohort study was conducted in 133 adolescents with idiopathic scoliosis undergoing spine fusion under standard protocols. Data regarding pain, opioid consumption, anxiety, and catastrophizing (using validated questionnaires) were collected before and 2–3 months postsurgery. Outcomes evaluated were preoperative pain, acute postoperative pain (area under curve [AUC] for pain scores over 48 hours), and CPSP (numerical rating scale >3/10 at 2–3 months postsurgery). Blood samples collected preoperatively were analyzed for DNA methylation by pyrosequencing of 22 CpG sites at the OPRM1 gene promoter. The association of each pain outcome with the methylation percentage of each CpG site was assessed using multivariable regression, adjusting for significant (P<0.05) nongenetic variables. RESULTS: Majority (83%) of the patients reported no pain preoperatively, while CPSP occurred in 36% of the subjects (44/121). Regression on dichotomized preoperative pain outcome showed association with methylation at six CpG sites (1, 3, 4, 9, 11, and 17) (P<0.05). Methylation at CpG sites 4, 17, and 18 was associated with higher AUC after adjusting for opioid consumption and preoperative pain score (P<0.05). After adjusting for postoperative opioid consumption and preoperative pain score, methylation at CpG sites 13 and 22 was associated with CPSP (P<0.05). DISCUSSION: Novel CPSP biomarkers were identified in an active regulatory region of the OPRM1 gene that binds multiple transcription factors. Inhibition of binding by DNA methylation potentially decreases the OPRM1 gene expression, leading to a decreased response to endogenous and exogenous opioids, and an increased pain experience. Dove Medical Press 2017-05-09 /pmc/articles/PMC5432115/ /pubmed/28533693 http://dx.doi.org/10.2147/PGPM.S132691 Text en © 2017 Chidambaran et al. This work is published and licensed by Dove Medical Press Limited The full terms of this license are available at https://www.dovepress.com/terms.php and incorporate the Creative Commons Attribution – Non Commercial (unported, v3.0) License (http://creativecommons.org/licenses/by-nc/3.0/). By accessing the work you hereby accept the Terms. Non-commercial uses of the work are permitted without any further permission from Dove Medical Press Limited, provided the work is properly attributed.
spellingShingle Original Research
Chidambaran, Vidya
Zhang, Xue
Martin, Lisa J
Ding, Lili
Weirauch, Matthew T
Geisler, Kristie
Stubbeman, Bobbie L
Sadhasivam, Senthilkumar
Ji, Hong
DNA methylation at the mu-1 opioid receptor gene (OPRM1) promoter predicts preoperative, acute, and chronic postsurgical pain after spine fusion
title DNA methylation at the mu-1 opioid receptor gene (OPRM1) promoter predicts preoperative, acute, and chronic postsurgical pain after spine fusion
title_full DNA methylation at the mu-1 opioid receptor gene (OPRM1) promoter predicts preoperative, acute, and chronic postsurgical pain after spine fusion
title_fullStr DNA methylation at the mu-1 opioid receptor gene (OPRM1) promoter predicts preoperative, acute, and chronic postsurgical pain after spine fusion
title_full_unstemmed DNA methylation at the mu-1 opioid receptor gene (OPRM1) promoter predicts preoperative, acute, and chronic postsurgical pain after spine fusion
title_short DNA methylation at the mu-1 opioid receptor gene (OPRM1) promoter predicts preoperative, acute, and chronic postsurgical pain after spine fusion
title_sort dna methylation at the mu-1 opioid receptor gene (oprm1) promoter predicts preoperative, acute, and chronic postsurgical pain after spine fusion
topic Original Research
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5432115/
https://www.ncbi.nlm.nih.gov/pubmed/28533693
http://dx.doi.org/10.2147/PGPM.S132691
work_keys_str_mv AT chidambaranvidya dnamethylationatthemu1opioidreceptorgeneoprm1promoterpredictspreoperativeacuteandchronicpostsurgicalpainafterspinefusion
AT zhangxue dnamethylationatthemu1opioidreceptorgeneoprm1promoterpredictspreoperativeacuteandchronicpostsurgicalpainafterspinefusion
AT martinlisaj dnamethylationatthemu1opioidreceptorgeneoprm1promoterpredictspreoperativeacuteandchronicpostsurgicalpainafterspinefusion
AT dinglili dnamethylationatthemu1opioidreceptorgeneoprm1promoterpredictspreoperativeacuteandchronicpostsurgicalpainafterspinefusion
AT weirauchmatthewt dnamethylationatthemu1opioidreceptorgeneoprm1promoterpredictspreoperativeacuteandchronicpostsurgicalpainafterspinefusion
AT geislerkristie dnamethylationatthemu1opioidreceptorgeneoprm1promoterpredictspreoperativeacuteandchronicpostsurgicalpainafterspinefusion
AT stubbemanbobbiel dnamethylationatthemu1opioidreceptorgeneoprm1promoterpredictspreoperativeacuteandchronicpostsurgicalpainafterspinefusion
AT sadhasivamsenthilkumar dnamethylationatthemu1opioidreceptorgeneoprm1promoterpredictspreoperativeacuteandchronicpostsurgicalpainafterspinefusion
AT jihong dnamethylationatthemu1opioidreceptorgeneoprm1promoterpredictspreoperativeacuteandchronicpostsurgicalpainafterspinefusion