DOI QR코드

DOI QR Code

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

  • Zhu, Yi (Department of Anesthesiology, General Hospital of The Southern Theater Command of PLA) ;
  • Sun, Mei (Department of Anesthesiology, General Hospital of The Southern Theater Command of PLA) ;
  • Liu, Peng (Department of Burns and Plastic Surgery, General Hospital of The Southern Theater Command of PLA) ;
  • Shao, Weidong (Department of Anesthesiology, General Hospital of The Southern Theater Command of PLA) ;
  • Xiong, Ming (Department of Anesthesiology and Peri-Operative Medicine, New Jersey Medical School) ;
  • Xu, Bo (Department of Anesthesiology, General Hospital of The Southern Theater Command of PLA)
  • Received : 2022.05.26
  • Accepted : 2022.08.02
  • Published : 2022.10.01

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-REPORTTM 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.

Keywords

Acknowledgement

We thank Dr. Ji Jia for providing advice in experimental design, and a thank-you to Dr. Xingan Zhang for the guidance on the design of the rat model.

References

  1. Glare P, Aubrey KR, Myles PS. Transition from acute to chronic pain after surgery. Lancet 2019; 393: 1537-46. https://doi.org/10.1016/S0140-6736(19)30352-6
  2. Chapman CR, Vierck CJ. The transition of acute postoperative pain to chronic pain: an integrative overview of research on mechanisms. J Pain 2017; 18: 359.e1-38. https://doi.org/10.1093/pm/pnx033
  3. Grolimund J. [Chronic postoperative pain and psychological factors]. Ther Umsch 2020; 77: 252-7. German. https://doi.org/10.1024/0040-5930/a001187
  4. Thapa P, Euasobhon P. Chronic postsurgical pain: current evidence for prevention and management. Korean J Pain 2018; 31: 155-73. https://doi.org/10.3344/kjp.2018.31.3.155
  5. Connor M, Christie MD. Opioid receptor signalling mechanisms. Clin Exp Pharmacol Physiol 1999; 26: 493-9. https://doi.org/10.1046/j.1440-1681.1999.03049.x
  6. Gu D, Zhou M, Han C, Lei D, Xie S, Yuan Y, et al. Preoperative anxiety induces chronic postoperative pain by activating astrocytes in the anterior cingulate cortex region. Rev Assoc Med Bras (1992) 2019; 65: 1174-80. https://doi.org/10.1590/1806-9282.65.9.1174
  7. Valentino RJ, Volkow ND. Untangling the complexity of opioid receptor function. Neuropsychopharmacology 2018; 43: 2514-20. https://doi.org/10.1038/s41386-018-0225-3
  8. Neugebauer V, Mazzitelli M, Cragg B, Ji G, Navratilova E, Porreca F. Amygdala, neuropeptides, and chronic painrelated affective behaviors. Neuropharmacology 2020; 170: 108052. https://doi.org/10.1016/j.neuropharm.2020.108052
  9. Wilson TD, Valdivia S, Khan A, Ahn HS, Adke AP, Martinez Gonzalez S, et al. Dual and opposing functions of the central amygdala in the modulation of pain. Cell Rep 2019; 29: 332-46.e5. https://doi.org/10.1016/j.celrep.2019.09.011
  10. Navratilova E, Nation K, Remeniuk B, Neugebauer V, Bannister K, Dickenson AH, et al. 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 2020; 161: 609-18. https://doi.org/10.1097/j.pain.0000000000001748
  11. Shin MS, Helmstetter FJ. 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 2005; 1064: 56-65. https://doi.org/10.1016/j.brainres.2005.09.065
  12. Kissiwaa SA, Patel SD, Winters BL, Bagley EE. Opioids differentially modulate two synapses important for pain processing in the amygdala. Br J Pharmacol 2020; 177: 420-31. https://doi.org/10.1111/bph.14877
  13. Goedecke L, Bengoetxea X, Blaesse P, Pape HC, Jungling K. μ-opioid receptor-mediated downregulation of midline thalamic pathways to basal and central amygdala. Sci Rep 2019; 9: 17837. https://doi.org/10.1038/s41598-019-54128-8
  14. Sanchez-Fernandez C, Montilla-Garcia A, Gonzalez-Cano R, Nieto FR, Romero L, Artacho-Cordon A, et al. Modulation of peripheral μ-opioid analgesia by σ1 receptors. J Pharmacol Exp Ther 2014; 348: 32-45. https://doi.org/10.1124/jpet.113.208272
  15. Mohr AM, Mott JL. Overview of microRNA biology. Semin Liver Dis 2015; 35: 3-11. https://doi.org/10.1055/s-0034-1397344
  16. Saliminejad K, Khorram Khorshid HR, Soleymani Fard S, Ghaffari SH. An overview of microRNAs: biology, functions, therapeutics, and analysis methods. J Cell Physiol 2019; 234: 5451-65. https://doi.org/10.1002/jcp.27486
  17. Bartel DP. MicroRNAs: genomics, biogenesis, mechanism, and function. Cell 2004; 116: 281-97. https://doi.org/10.1016/S0092-8674(04)00045-5
  18. Lu J, Getz G, Miska EA, Alvarez-Saavedra E, Lamb J, Peck D, et al. MicroRNA expression profiles classify human cancers. Nature 2005; 435: 834-8. https://doi.org/10.1038/nature03702
  19. Ali Syeda Z, Langden SSS, Munkhzul C, Lee M, Song SJ. Regulatory mechanism of microRNA expression in cancer. Int J Mol Sci 2020; 21: 1723. https://doi.org/10.3390/ijms21051723
  20. Giordano R, Petersen KK, Andersen HH, Lichota J, Valeriani M, Simonsen O, et al. Preoperative serum circulating microRNAs as potential biomarkers for chronic postoperative pain after total knee replacement. Mol Pain 2020; 16: 1744806920962925.
  21. Cata JP, Gorur A, Yuan X, Berg NK, Sood AK, Eltzschig HK. Role of micro-RNA for pain after surgery: narrative review of animal and human studies. Anesth Analg 2020; 130: 1638-52. https://doi.org/10.1213/ANE.0000000000004767
  22. Wylde V, Dennis J, Beswick AD, Bruce J, Eccleston C, Howells N, et al. Systematic review of management of chronic pain after surgery. Br J Surg 2017; 104: 1293-306. https://doi.org/10.1002/bjs.10601
  23. Tian Y, Liu X, Jia M, Yu H, Lichtner P, Shi Y, et al. Targeted genotyping identifies susceptibility locus in brain-derived neurotrophic factor gene for chronic postsurgical pain. Anesthesiology 2018; 128: 587-97. Erratum in: Anesthesiology 2018; 128: 1049.
  24. James SK. Chronic postsurgical pain: is there a possible genetic link? Br J Pain 2017; 11: 178-85. https://doi.org/10.1177/2049463717723222
  25. Chidambaran V, Zhang X, Martin LJ, Ding L, Weirauch MT, Geisler K, et al. DNA methylation at the mu-1 opioid receptor gene (OPRM1) promoter predicts preoperative, acute, and chronic postsurgical pain after spine fusion. Pharmgenomics Pers Med 2017; 10: 157-68.
  26. Rabbitts JA, Palermo TM, Lang EA. A conceptual model of biopsychosocial mechanisms of transition from acute to chronic postsurgical pain in children and adolescents. J Pain Res 2020; 13: 3071-80. https://doi.org/10.2147/JPR.S239320
  27. Luo A, Zhou X, Shi X, Zhao Y, Men Y, Chang X, et al. Exosome-derived miR-339-5p mediates radiosensitivity by targeting Cdc25A in locally advanced esophageal squamous cell carcinoma. Oncogene 2019; 38: 4990-5006. https://doi.org/10.1038/s41388-019-0771-0
  28. Li Y, Zhang X, Yang Z, Li Y, Han B, Chen LA. miR-339-5p inhibits metastasis of non-small cell lung cancer by regulating the epithelial-to-mesenchymal transition. Oncol Lett 2018; 15: 2508-14.
  29. Shan W, Li J, Bai Y, Lu X. miR-339-5p inhibits migration and invasion in ovarian cancer cell lines by targeting NACC1 and BCL6. Tumour Biol 2016; 37: 5203-11. https://doi.org/10.1007/s13277-015-4390-2
  30. Yu Z, Zhao S, Wang L, Wang J, Zhou J. miRNA-339-5p plays an important role in invasion and migration of pancreatic cancer cells. Med Sci Monit 2019; 25: 7509-17. https://doi.org/10.12659/msm.917038
  31. Liu B, Li J, Cairns MJ. Identifying miRNAs, targets and functions. Brief Bioinform 2014; 15: 1-19. https://doi.org/10.1093/bib/bbs075
  32. Fabian MR, Sonenberg N, Filipowicz W. Regulation of mRNA translation and stability by microRNAs. Annu Rev Biochem 2010; 79: 351-79. https://doi.org/10.1146/annurev-biochem-060308-103103