Korean J Pain.  2022 Oct;35(4):423-432. 10.3344/kjp.2022.35.4.423.

Perioperative stress prolong post-surgical pain via miR-339-5p targeting oprm1 in the amygdala

Affiliations
  • 1Department of Anesthesiology, General Hospital of The Southern Theater Command of PLA, Guangzhou, China
  • 2Department of Burns and Plastic Surgery, General Hospital of The Southern Theater Command of PLA, Guangzhou, China
  • 3Department of Anesthesiology and Peri-Operative Medicine, New Jersey Medical School, Newark, NJ, USA

Abstract

Background
The decreased expression of mu-opioid receptors (MOR) in the amygdala may be a key molecular in chronic post-surgical pain (CPSP). It is known that miR-339-5p expression in the amygdala of a stressed rat model was increased. Analyzed by RNAhybrid, miR-339-5p could target opioid receptor mu 1 (oprm1) which codes MOR directly. So, the authors hypothesized that miR-339-5p could regulate the expression of MOR via targeting oprm1 and cause the effects to CPSP.
Methods
To simulate perioperative short-term stress, a perioperative stress prolongs incision-induced pain hypersensitivity without changing basal pain perception rat model was built. A pmiR-RB-REPORT™ dual luciferase assay was taken to verify whether miR-339-5p could act on oprm1 as a target. The serum glucocorticoid level of rats was test. Differential expressions of MOR, GFAP, and pERK1/2 in each group of the rats’ amygdala were tested, and the expressions of miR-339-5p in each group of rats’ amygdalas were also measured.
Results
Perioperative stress prolonged the recovery time of incision pain. The expression of MOR was down-regulated in the amygdala of rats in stress + incision (S + IN) group significantly compared with other groups (P < 0.050). miR-339-5p was up-regulated in the amygdala of rats in group S + IN significantly compared with other groups (P < 0.050). miR-339-5p acts on oprm1 3’UTR and take MOR mRNA as a target.
Conclusions
Perioperative stress could increase the expression of miR-339-5p, and miR-339-5p could cause the expression of MOR to decrease via targeting oprm1. This regulatory pathway maybe an important molecular mechanism of CPSP.

Keyword

Amygdala; Glucocorticoids; MicroRNAs; Models; Animal; Pain Perception; Pain; Postoperative; Physiopathology; Receptors; Opioid; mu; Stress; Physiological

Figure

  • Fig. 1 (A–C) The mechanical pain threshold, thermal withdrawal latency, and cold withdrawal latency of the rats did not change within 3 days of stress (sleep deprived and movements restriction) compared with the control group (*P > 0.050). The mechanical pain and cold withdrawal latency significantly declined from the 4th day (*P < 0.050), and the thermal withdrawal latency is significantly declined from the 5th day (**P < 0.050). (D–F) The recovery time of postoperative incision pain is significantly delayed in group stress + incision (S + IN). The mechanical pain threshold, and cold withdrawal latency are significantly lower than that in group IN from the 5th day after operation (***P < 0.050). The thermal withdrawal latency of the rats in group S + IN is significantly lower than that in group IN from the 5th day after operation (***P < 0.050). At the end of the experiment (9th day after operation), the mechanical pain threshold, cold withdrawal latency, and thermal withdrawal latency are significantly lower than that in other groups. The error bars indicate mean ± standard deviation.

  • Fig. 2 (A, C) Western blotting assays of GFAP, p-ERK1/2, and MOR were performed in the amygdala tissue of rats. The results show that the expression of GFAP and p-ERK1/2 are up-regulated in group S + IN. The expression of MOR is down-regulated in group S + IN. (B) Both stress and operation could increase the level of serum cortisol in rats. The level of serum cortisol in the rats of group S + IN is significantly higher than that in other groups (*P < 0.050), but there was no significant difference between group IN and group S (**P < 0.050). The error bars indicate mean ± standard deviation. GFAP: glial fibrillary acidic protein, p-ERK1/2: phosphorylated extracellular regulatory protein kinase, MOR: mu-opioid receptors, S: stress, IN: incision.

  • Fig. 3 miR-339-5p directly targets the oprm1 3’UTR. (A) pmiR-RB-REPORTTM Double luciferase reporter vector. (B) The predicted sequences for target site of r-oprm1-WT and r-oprm1-MUT (indicated by red letters). (C) Relative luciferase activity. After the transfection of rno-mir-339-5p, the reported fluorescence expression of r-oprm1-MUT increased significantly compared with that of r-oprm1-WT (*P < 0.050). This significant difference does not exist in the NC (**P > 0.050). The error bars indicate mean ± standard deviation. 3’UTR: 3’untranslated region, NC: negative control.

  • Fig. 4 (A) The expression of GFAP, p-ERK1/2, and MOR by immunohistochemistry staining in amygdala tissue of rats (magnification: 200×). The positive expressing proteins appeared brown in color. (B) The results showed that GFAP and p-ERK1/2 are significantly up-regulated in group S + IN (*P < 0.050), the expression of MOR is significantly down-regulated in group S + IN (*P < 0.050). There were no significant differences between group IN and group S (**P > 0.050). The expression of GFAP and p-ERK1/2 are significantly up-regulated in group S and group IN (*P < 0.050) compared with the sham group. The expression of MOR is significantly down-regulated in group S and group IN (*P < 0.050) compared with the sham group. The error bars indicate mean ± standard deviation. GFAP: glial fibrillary acidic protein, p-ERK1/2: phosphorylated extracellular regulatory protein kinase, MOR: mu-opioid receptors, S: stress, IN: incision, IOD: integrated optical density.

  • Fig. 5 (A, B) Immunofluoresence assays indicate that the expression of MOR is significantly down-regulated in group S + IN (*P < 0.050). Under the laser-scanning confocal microscope the MOR appeared with a green fluorescence (magnification: 400×). (C) The results of qRT-PCR show that the expression of miR-339-5p is significantly up-regulated in group S + IN (*P < 0.050). There were no significant differences between group IN and group S (**P > 0.050). The error bars indicate mean ± standard deviation. MOR: mu-opioid receptors, S: stress, IN: incision, qRT-PCR: quantitative real-time polymerase chain reaction.


Reference

1. Glare P, Aubrey KR, Myles PS. 2019; Transition from acute to chronic pain after surgery. Lancet. 393:1537–46. DOI: 10.1016/S0140-6736(19)30352-6. PMID: 30983589. PMID: https://www.scopus.com/inward/record.uri?partnerID=HzOxMe3b&scp=85064048141&origin=inward.
Article
2. Chapman CR, Vierck CJ. 2017; The transition of acute postoperative pain to chronic pain: an integrative overview of research on mechanisms. J Pain. 18:359.e1–38. DOI: 10.1016/j.jpain.2016.11.004. PMID: 27908839. PMID: https://www.scopus.com/inward/record.uri?partnerID=HzOxMe3b&scp=85009756610&origin=inward.
Article
3. Grolimund J. 2020; Chronic postoperative pain and psychological factors. Ther Umsch. 77:252–7. German. DOI: 10.1024/0040-5930/a001187. PMID: 32930078. PMID: https://www.scopus.com/inward/record.uri?partnerID=HzOxMe3b&scp=85092717287&origin=inward.
4. Thapa P, Euasobhon P. 2018; Chronic postsurgical pain: current evidence for prevention and management. Korean J Pain. 31:155–73. DOI: 10.3344/kjp.2018.31.3.155. PMID: 30013730. PMCID: PMC6037807. PMID: https://www.scopus.com/inward/record.uri?partnerID=HzOxMe3b&scp=85049382882&origin=inward.
Article
5. Connor M, Christie MD. 1999; Opioid receptor signalling mechanisms. Clin Exp Pharmacol Physiol. 26:493–9. DOI: 10.1046/j.1440-1681.1999.03049.x. PMID: 10405772. PMID: https://www.scopus.com/inward/record.uri?partnerID=HzOxMe3b&scp=0033057206&origin=inward.
Article
6. Gu D, Zhou M, Han C, Lei D, Xie S, Yuan Y, et al. 2019; Preoperative anxiety induces chronic postoperative pain by activating astrocytes in the anterior cingulate cortex region. Rev Assoc Med Bras (1992). 65:1174–80. DOI: 10.1590/1806-9282.65.9.1174. PMID: 31618333. PMID: https://www.scopus.com/inward/record.uri?partnerID=HzOxMe3b&scp=85073468335&origin=inward.
Article
7. Valentino RJ, Volkow ND. 2018; Untangling the complexity of opioid receptor function. Neuropsychopharmacology. 43:2514–20. DOI: 10.1038/s41386-018-0225-3. PMID: 30250308. PMCID: PMC6224460. PMID: https://www.scopus.com/inward/record.uri?partnerID=HzOxMe3b&scp=85054485115&origin=inward.
Article
8. Neugebauer V, Mazzitelli M, Cragg B, Ji G, Navratilova E, Porreca F. 2020; Amygdala, neuropeptides, and chronic pain-related affective behaviors. Neuropharmacology. 170:108052. DOI: 10.1016/j.neuropharm.2020.108052. PMID: 32188569. PMCID: PMC7214122. PMID: https://www.scopus.com/inward/record.uri?partnerID=HzOxMe3b&scp=85081887242&origin=inward.
Article
9. Wilson TD, Valdivia S, Khan A, Ahn HS, Adke AP, Martinez Gonzalez S, et al. 2019; Dual and opposing functions of the central amygdala in the modulation of pain. Cell Rep. 29:332–46.e5. DOI: 10.1016/j.celrep.2019.09.011. PMID: 31597095. PMCID: PMC6816228. PMID: https://www.scopus.com/inward/record.uri?partnerID=HzOxMe3b&scp=85072781729&origin=inward.
Article
10. Navratilova E, Nation K, Remeniuk B, Neugebauer V, Bannister K, Dickenson AH, et al. 2020; Selective modulation of tonic aversive qualities of neuropathic pain by morphine in the central nucleus of the amygdala requires endogenous opioid signaling in the anterior cingulate cortex. Pain. 161:609–18. DOI: 10.1097/j.pain.0000000000001748. PMID: 31725062. PMCID: PMC7124010. PMID: https://www.scopus.com/inward/record.uri?partnerID=HzOxMe3b&scp=85079358399&origin=inward.
Article
11. Shin MS, Helmstetter FJ. 2005; Antinociception following application of DAMGO to the basolateral amygdala results from a direct interaction of DAMGO with Mu opioid receptors in the amygdala. Brain Res. 1064:56–65. DOI: 10.1016/j.brainres.2005.09.065. PMID: 16289487. PMID: https://www.scopus.com/inward/record.uri?partnerID=HzOxMe3b&scp=28844506690&origin=inward.
Article
12. Kissiwaa SA, Patel SD, Winters BL, Bagley EE. 2020; Opioids differentially modulate two synapses important for pain processing in the amygdala. Br J Pharmacol. 177:420–31. DOI: 10.1111/bph.14877. PMID: 31596498. PMCID: PMC6989950. PMID: https://www.scopus.com/inward/record.uri?partnerID=HzOxMe3b&scp=85077889866&origin=inward.
Article
13. Goedecke L, Bengoetxea X, Blaesse P, Pape HC, Jüngling K. 2019; µ-opioid receptor-mediated downregulation of midline thalamic pathways to basal and central amygdala. Sci Rep. 9:17837. DOI: 10.1038/s41598-019-54128-8. PMID: 31780740. PMCID: PMC6882837. PMID: https://www.scopus.com/inward/record.uri?partnerID=HzOxMe3b&scp=85075668441&origin=inward.
Article
14. Sánchez-Fernández C, Montilla-García Á, González-Cano R, Nieto FR, Romero L, Artacho-Cordón A, et al. 2014; Modulation of peripheral μ-opioid analgesia by σ1 receptors. J Pharmacol Exp Ther. 348:32–45. DOI: 10.1124/jpet.113.208272. PMID: 24155346. PMID: https://www.scopus.com/inward/record.uri?partnerID=HzOxMe3b&scp=84890502351&origin=inward.
Article
15. Mohr AM, Mott JL. 2015; Overview of microRNA biology. Semin Liver Dis. 35:3–11. DOI: 10.1055/s-0034-1397344. PMID: 25632930. PMCID: PMC4797991. PMID: https://www.scopus.com/inward/record.uri?partnerID=HzOxMe3b&scp=84928553627&origin=inward.
Article
16. Saliminejad K, Khorram Khorshid HR, Soleymani Fard S, Ghaffari SH. 2019; An overview of microRNAs: biology, functions, therapeutics, and analysis methods. J Cell Physiol. 234:5451–65. DOI: 10.1002/jcp.27486. PMID: 30471116. PMID: https://www.scopus.com/inward/record.uri?partnerID=HzOxMe3b&scp=85057204100&origin=inward.
Article
17. Bartel DP. 2004; MicroRNAs: genomics, biogenesis, mechanism, and function. Cell. 116:281–97. DOI: 10.1016/S0092-8674(04)00045-5. PMID: 14744438. PMID: https://www.scopus.com/inward/record.uri?partnerID=HzOxMe3b&scp=0347444723&origin=inward.
18. Lu J, Getz G, Miska EA, Alvarez-Saavedra E, Lamb J, Peck D, et al. 2005; MicroRNA expression profiles classify human cancers. Nature. 435:834–8. DOI: 10.1038/nature03702. PMID: 15944708. PMID: https://www.scopus.com/inward/record.uri?partnerID=HzOxMe3b&scp=20444460289&origin=inward.
Article
19. Ali Syeda Z, Langden SSS, Munkhzul C, Lee M, Song SJ. 2020; Regulatory mechanism of microRNA expression in cancer. Int J Mol Sci. 21:1723. DOI: 10.3390/ijms21051723. PMID: 32138313. PMCID: PMC7084905. PMID: 6cb81bbbff3c4e34b90b9254ca2b9885. PMID: https://www.scopus.com/inward/record.uri?partnerID=HzOxMe3b&scp=85080979790&origin=inward.
Article
20. Giordano R, Petersen KK, Andersen HH, Lichota J, Valeriani M, Simonsen O, et al. 2020; Preoperative serum circulating microRNAs as potential biomarkers for chronic postoperative pain after total knee replacement. Mol Pain. 16:1744806920962925. DOI: 10.1177/1744806920962925. PMID: 33021154. PMCID: PMC7543153. PMID: https://www.scopus.com/inward/record.uri?partnerID=HzOxMe3b&scp=85092391389&origin=inward.
Article
21. Cata JP, Gorur A, Yuan X, Berg NK, Sood AK, Eltzschig HK. 2020; Role of micro-RNA for pain after surgery: narrative review of animal and human studies. Anesth Analg. 130:1638–52. DOI: 10.1213/ANE.0000000000004767. PMID: 32384350. PMCID: PMC8543451. PMID: https://www.scopus.com/inward/record.uri?partnerID=HzOxMe3b&scp=85084721851&origin=inward.
22. Wylde V, Dennis J, Beswick AD, Bruce J, Eccleston C, Howells N, et al. 2017; Systematic review of management of chronic pain after surgery. Br J Surg. 104:1293–306. DOI: 10.1002/bjs.10601. PMID: 28681962. PMCID: PMC5599964. PMID: https://www.scopus.com/inward/record.uri?partnerID=HzOxMe3b&scp=85021831926&origin=inward.
Article
23. Tian Y, Liu X, Jia M, Yu H, Lichtner P, Shi Y, et al. 2018; Targeted genotyping identifies susceptibility locus in brain-derived neurotrophic factor gene for chronic postsurgical pain. Anesthesiology. 128:587–97. Erratum in: Anesthesiology 2018; 128: 1049. DOI: 10.1097/ALN.0000000000001977. PMID: 29120944. PMID: https://www.scopus.com/inward/record.uri?partnerID=HzOxMe3b&scp=85046787514&origin=inward.
Article
24. James SK. 2017; Chronic postsurgical pain: is there a possible genetic link? Br J Pain. 11:178–85. DOI: 10.1177/2049463717723222. PMID: 29123662. PMCID: PMC5661692. PMID: https://www.scopus.com/inward/record.uri?partnerID=HzOxMe3b&scp=85032485309&origin=inward.
Article
25. Chidambaran V, Zhang X, Martin LJ, Ding L, Weirauch MT, Geisler K, et al. 2017; DNA methylation at the mu-1 opioid receptor gene (OPRM1) promoter predicts preoperative, acute, and chronic postsurgical pain after spine fusion. Pharmgenomics Pers Med. 10:157–68. DOI: 10.2147/PGPM.S132691. PMID: 28533693. PMCID: PMC5432115. PMID: https://www.scopus.com/inward/record.uri?partnerID=HzOxMe3b&scp=85019981845&origin=inward.
26. Rabbitts JA, Palermo TM, Lang EA. 2020; A conceptual model of biopsychosocial mechanisms of transition from acute to chronic postsurgical pain in children and adolescents. J Pain Res. 13:3071–80. DOI: 10.2147/JPR.S239320. PMID: 33262642. PMCID: PMC7699440. PMID: https://www.scopus.com/inward/record.uri?partnerID=HzOxMe3b&scp=85096636811&origin=inward.
27. Luo A, Zhou X, Shi X, Zhao Y, Men Y, Chang X, et al. 2019; Exosome-derived miR-339-5p mediates radiosensitivity by targeting Cdc25A in locally advanced esophageal squamous cell carcinoma. Oncogene. 38:4990–5006. DOI: 10.1038/s41388-019-0771-0. PMID: 30858545. PMID: https://www.scopus.com/inward/record.uri?partnerID=HzOxMe3b&scp=85062825958&origin=inward.
Article
28. Li Y, Zhang X, Yang Z, Li Y, Han B, Chen LA. 2018; miR-339-5p inhibits metastasis of non-small cell lung cancer by regulating the epithelial-to-mesenchymal transition. Oncol Lett. 15:2508–14. DOI: 10.3892/ol.2017.7608. PMID: 29434966. PMCID: PMC5777278. PMID: https://www.scopus.com/inward/record.uri?partnerID=HzOxMe3b&scp=85038429902&origin=inward.
Article
29. Shan W, Li J, Bai Y, Lu X. 2016; miR-339-5p inhibits migration and invasion in ovarian cancer cell lines by targeting NACC1 and BCL6. Tumour Biol. 37:5203–11. DOI: 10.1007/s13277-015-4390-2. PMID: 26553360. PMID: https://www.scopus.com/inward/record.uri?partnerID=HzOxMe3b&scp=84946779914&origin=inward.
Article
30. Yu Z, Zhao S, Wang L, Wang J, Zhou J. 2019; miRNA-339-5p plays an important role in invasion and migration of pancreatic cancer cells. Med Sci Monit. 25:7509–17. DOI: 10.12659/MSM.917038. PMID: 31588120. PMCID: PMC6792519. PMID: https://www.scopus.com/inward/record.uri?partnerID=HzOxMe3b&scp=85072929417&origin=inward.
Article
31. Liu B, Li J, Cairns MJ. 2014; Identifying miRNAs, targets and functions. Brief Bioinform. 15:1–19. DOI: 10.1093/bib/bbs075. PMID: 23175680. PMCID: PMC3896928. PMID: https://www.scopus.com/inward/record.uri?partnerID=HzOxMe3b&scp=84893025641&origin=inward.
Article
32. Fabian MR, Sonenberg N, Filipowicz W. 2010; Regulation of mRNA translation and stability by microRNAs. Annu Rev Biochem. 79:351–79. DOI: 10.1146/annurev-biochem-060308-103103. PMID: 20533884. PMID: https://www.scopus.com/inward/record.uri?partnerID=HzOxMe3b&scp=77953629046&origin=inward.
Article
Full Text Links
  • KJP
Actions
Cited
CITED
export Copy
Close
Share
  • Twitter
  • Facebook
Similar articles
Copyright © 2024 by Korean Association of Medical Journal Editors. All rights reserved.     E-mail: koreamed@kamje.or.kr