Article Data

  • Views 406
  • Dowloads 57

Original Research

Open Access

Exercise-Induced Hypoalgesia Profile in a Rat Neuropathic Pain Model Predicts Pain Severity Following Infraorbital Nerve Injury and Is Associated with Local Cytokine Levels, Systemic Endocannabinoids, and Endogenous Opioids

  • Junad Khan1,*,
  • Qian Wang1
  • Olga A. Korczeniewska2
  • Rotem Eliav3
  • Yanfang Ren1
  • Eli Eliav1

1Orofacial Pain and TMJ Disorders, Eastman Institute for Oral Health, University of Rochester Medical Center, Rochester, New York, USA

2Rutgers School of Dental Medicine, Department of Diagnostic Sciences Rutgers, The State University of New Jersey, Newark, New Jersey, USA

3Rabin Medical Center, Petah Tikva, Israel

DOI: 10.11607/ofph.3003 Vol.35,Issue 3,September 2021 pp.230-240

Submitted: 04 May 2021 Accepted: 25 May 2021

Published: 30 September 2021

*Corresponding Author(s): Junad Khan E-mail: junad_khan@URMC.rochester.edu

Abstract

Aims: To investigate the role of exercise-induced hypoalgesia (EIH) in the development of neuropathic pain (NP) following infraorbital nerve (ION) injury and to explore possible underlying mechanisms defining the differences between rats with high and low EIH. Methods: EIH was evaluated by measuring the percentage of withdrawal responses to a series of 30 mechanical stimuli applied to the hind paw before and after 180 seconds of exercise on a rotating rod. The rats were assigned to low- and high-EIH groups based on reduction in the percent of withdrawal responses following exercise. NP was induced in high- and low-EIH rats via ION constriction injury. Rats were tested with graded nylon monofilaments to establish the withdrawal threshold. Increasingly stiff monofilaments were applied to the ION territory until there was a clear withdrawal by the rat. This was repeated a total of three times. A decreased withdrawal threshold indicates allodynia. Testing was performed at baseline and at 3, 10, and 17 days following the injury. On day 17 postinjury, IONs were harvested for the assessment of interleukin (IL)-6, IL-1β, and IL-10 levels. Samples from high-EIH and low-EIH surgically naïve rats served as control for the cytokines study. In this second part of the study, the effects of cannabinoid 1 (CB1) and cannabinoid 2 (CB2) antagonists and naltrexone on EIH profiles and on the withdrawal thresholds to mechanical stimulation were measured. EIH and withdrawal thresholds in high- and low-EIH rats were measured before and after administration of antagonists. Results: Low-EIH rats developed significantly more pronounced allodynia in the ION territory following injury compared to high-EIH rats. At 17 days postinjury, ION IL-1β levels were higher in low-EIH rats, and IL-10 levels were higher in high-EIH rats. CB1 antagonist blocked the analgesic effect induced by exercise in high- but not in low-EIH rats. The CB2 antagonist had no significant effect on high- or low-EIH rats. Naltrexone blocked the effects of EIH in both high- and low-EIH rats. Exercise induced a significant analgesic effect in high-EIH but not in low-EIH rats. CB1 or CB2 antagonist administration had no effect on pre-exercise responses to mechanical stimulation, while naltrexone administration resulted in significant allodynia in both low- and high-EIH rats. Conclusion: This study demonstrated substantial differences between rats with high and low EIH. The results suggest that following ION injury, high-EIH rats may have a more prominent or activated endocannabinoids system and that their inflammatory response is moderated, with higher levels of IL-10 and lower levels of IL-1β.

Keywords

exercise-induced hypoalgesia; nerve injury; orofacial neuropathic pain; pain modulation; trigeminal

Cite and Share

Junad Khan,Qian Wang,Olga A. Korczeniewska,Rotem Eliav,Yanfang Ren,Eli Eliav. Exercise-Induced Hypoalgesia Profile in a Rat Neuropathic Pain Model Predicts Pain Severity Following Infraorbital Nerve Injury and Is Associated with Local Cytokine Levels, Systemic Endocannabinoids, and Endogenous Opioids. Journal of Oral & Facial Pain and Headache. 2021. 35(3);230-240.

References

1. Benoliel R, Eliav E, Tal M. No sympathetic nerve sprouting in rat trigeminal ganglion following painful and non-painful infraorbital nerve neuropathy. Neurosci Lett 2001;297:151–154.

2. Tal M, Devor M. Ectopic discharge in injured nerves: Comparison of trigeminal and somatic afferents. Brain Res 1992;579:148–151.

3. Korczeniewska, OA, Katzmann Rider G, Gajra S, et al. Differential gene expression changes in the dorsal root versus trigeminal ganglia following peripheral nerve injury in rats. Eur J Pain 2020;24:967–982.

4. Graven-Nielsen T, Kendall SA, Henriksson KG. Ketamine reduces muscle pain, temporal summation, and referred pain in fibromyalgia patients. Pain 2000;85:483–491.

5. Staud R, Vierck CJ, Cannon RL, Mauderli AP, Price DD. Abnormal sensitization and temporal summation of second pain (wind-up) in patients with fibromyalgia syndrome. Pain 2001;91:165–175.

6. Price DD, Staud R, Robinson ME, Mauderli AP, Cannon R, Vierck CJ. Enhanced temporal summation of second pain and its central modulation in fibromyalgia patients. Pain 2002;99:49–59.

7. Staud R, Robinson ME, Vierck CJ Jr, Price DD. Diffuse noxious inhibitory controls (DNIC) attenuate temporal summation of second pain in normal males but not in normal females or fibromyalgia pa-tients. Pain 2003;101:167–174.

8. Kleinböhl D, Hölzl R, Möltner A, Rommel C, Weber C, Osswald PM. Psychophysical measures of sensitization to tonic heat discriminate chronic pain patients. Pain 1999;81:35–43.

9. Ashina S, Bendtsen L, Ashina M, Magerl W, Jensen R. Generalized hyperalgesia in patients with chronic tensiontype headache. Cephalalgia 2006;26:940–948.

10. Weissman-Fogel I, Sprecher E, Granovsky Y, Yarnitsky D. Repeated noxious stimulation of the skin enhances cutaneous pain perception of migraine patients inbetween attacks: Clinical evidence for continuous subthreshold increase in membrane excitability of central trigeminovascular neurons. Pain 2003;104:693–700.

11. Kleinböhl D, Görtelmeyer R, Bender HJ, Hölzl R. Amantadine sulfate reduces experimental sensitization and pain in chronic back pain patients. Anesth Analg 2006;102:840–847.

12. Piché M, Bouin M, Arsenault M, Poitras P, Rainville P. Decreased pain inhibition in irritable bowel syndrome depends on altered descending modulation and higher-order brain processes. Neuroscience 2011;195:166–175.

13. Nasri-Heir C, Khan J, Benoliel R. Altered pain modulation in patients with persistent postendodontic pain. Pain 2015;156:2032–2041.

14. Nasri-Heir C, Shigdar D, Alnaas D, Korczeniewska OA, Eliav R, Heir GM. Primary burning mouth syndrome: Literature review and preliminary findings suggesting possible association with pain modulation. Quintessence Int 2017;49:49–60.

15. Nishihara C, Watanabe K, Ozasa K, et al. Altered pain modulation to noxious heat thermal stimuli in burning mouth syndrome. Oral Dis 2020;26:1777–1782.

16. Black J, Chesher GB, Starmer GA, Egger G. The painlessness of the long distance runner. Med J Aust 1979;1:522–523.

17. Stagg NJ, Mata HP, Ibrahim MM, et al. Regular exercise reverses sensory hypersensitivity in a rat neuropathic pain model: Role of endogenous opioids. Anesthesiology 2011;114:940–948.

18. Bement MK, Sluka KA. Low-intensity exercise reverses chronic muscle pain in the rat in a naloxone-dependent manner. Arch Phys Med Rehabil 2005;86:1736–1740.

19. Koltyn KF, Brellenthin AG, Cook DB, Sehgal N, Hillard C. Mechanisms of exercise-induced hypoalgesia. J Pain 2014;15: 1294–1304.

20. Dietrich A, McDaniel WF. Endocannabinoids and exercise. Br J Sports Med 2004;38:536–541.

21. Bobinski F, Ferreira TAA, Córdova MM, et al. Role of brainstem serotonin in analgesia produced by low-intensity exercise on neuropathic pain after sciatic nerve injury in mice. Pain 2015;156:2595–2606.

22. Martins DF, Mazzardo-Martins L, Soldi F, Stramosk J, Piovezan AP, Santos ARS. High-intensity swimming exercise reduces neuropathic pain in an animal model of complex regional pain syndrome type I: Evidence for a role of the adenosinergic system. Neuroscience 2013;234:69–76.

23. Lovick TA. Integrated activity of cardiovascular and pain regulatory systems: Role in adaptive behavioural responses. Prog Neurobiol 1993;40:631–644.

24. Khan J, Benavent V, Korczeniewska OA, Benoliel R, Eliav E. Exercise-induced hypoalgesia profile in rats predicts neuropathic pain intensity induced by sciatic nerve constriction injury. J Pain 2014;15:1179–1189.

25. Zimmermann M. Ethical guidelines for investigations of experimental pain in conscious animals. Pain 1983;16:109–110.

26. Imamura Y, Kawamoto H, Nakanishi O. Characterization of heat-hyperalgesia in an experimental trigeminal neuropathy in rats. Exp Brain Res 1997;116:97–103.

27. Morgan RW, Nicholson KL. Characterization of the antinoci-ceptive effects of the individual isomers of methadone after acute and chronic administrations. Behav Pharmacol 2011;22: 548–557.

28. Galdino G, Romero TR, Silva JF, et al. The endocannabinoid system mediates aerobic exercise-induced antinociception in rats. Neuropharmacology 2014;77:313–324.

29. Pharmacology of the cannabinoid system. In: Beaulieu P, Lussier D, Porreca F, Dickenson A (eds). Pharmacology of Pain. Seattle: IASP, 2010:111–138.

30. Kuphal KE, Fibuch EE, Taylor BK. Extended swimming exercise reduces inflammatory and peripheral neuropathic pain in rodents. J Pain 2007;8:989–997.

31. Chen YW, Li YT, Chen YC, Li ZY, Hung CH. Exercise training attenuates neuropathic pain and cytokine expression after chronic constriction injury of rat sciatic nerve. Anesth Analg 2012;114:1330–1337.

32. Shen J, Foxw LE, Cheng J. Swim therapy reduces mechanical allodynia and thermal hyperalgesia induced by chronic constriction nerve injury in rats. Pain Med 2013;14:516–525.

33. Bobinski F, Ferreira TAA, Córdova MM, et al. Role of brain-stem serotonin in analgesia produced by low-intensity exercise on neuropathic pain after sciatic nerve injury in mice. Pain 2015;156:2595–2606.

34. Detloff MR, Quiros-Molina D, Javia AS, et al. Delayed exercise is ineffective at reversing aberrant nociceptive afferent plasticity or neuropathic pain after spinal cord injury in rats. Neurorehabil Neural Repair 2016;30:685–700.

35. Anheyer D, Klose P, Koch AK, Haller H, Dobos G, Cramer H. Comparative efficacy of different exercise interventions in chronic non-specific low back pain: Protocol of a systematic review and network meta-analysis. BMJ Open 2020;10:e036050.

36. de Zoete RM, Armfield NR, McAuley JH, Chen K, Sterling M. Comparative effectiveness of physical exercise interventions for chronic non-specific neck pain: A systematic review with network meta-analysis of 40 randomised controlled trials. Br J Sports Med 2020;bjsports-2020-102664.

37. Bakdach WMM, Hadad R. Effectiveness of supplemental vibrational force in reducing pain associated with orthodontic treatment: A systematic review. Quintessence Int 2020;51:742–752.

38. Sluka KA, O’Donnell JM, Danielson J, Rasmussen LA. Regular physical activity prevents development of chronic pain and activation of central neurons. J Appl Physiol (1985) 2013;114:725–733.

39. Brosseau L, Wells GA, Tugwell P, et al. Ottawa Panel evidence-based clinical practice guidelines for aerobic fitness exercises in the management of fibromyalgia: Part 1. Phys Ther 2008;88:857–871.

40. Jansen MJ, Viechtbauer W, Lenssen AF, Hendriks EJM, de Bie RA. Strength training alone, exercise therapy alone, and exercise therapy with passive manual mobilisation each reduce pain and disability in people with knee osteoarthritis: A systematic review. J Physiother 2011;57:11–20.

41. Stewart MJ, Maher CG, Refshauge KM, Herbert RD, Bogduk N, Nicholas M. Randomized controlled trial of exercise for chronic whiplash-associated disorders. Pain 2007;128:59–68.

42. van Middelkoop M, Rubinstein SM, Verhagen AP, Ostelo RW, Koes BW, van Tulder MW. Exercise therapy for chronic nonspecific low-back pain. Best Pract Res Clin Rheumatol 2010;24:193–204.

43. Fransen M, McConnell S, Hernandez-Molina G, Reichenbach S. Exercise for osteoarthritis of the hip. Cochrane Database Syst Rev 2014;(4):CD007912.

44. Busch AJ, Webber SC, Richards RS, et al. Resistance exercise training for fibromyalgia. Cochrane Database Syst Rev 2013;2013:CD010884.

45. Nasri-Heir C, Patil AG, Korczeniewska OA, et al. The effect of nonstrenuous aerobic exercise in patients with chronic masticatory myalgia. J Oral Facial Pain Headache 2019;33:143–152.

46. Benoliel R, Birenboim R, Regev E, Eliav E. Neurosensory changes in the infraorbital nerve following zygomatic fractures. Oral Surg Oral Med Oral Pathol Oral Radiol Endod 2005;99: 657–665.

47. Bongenhielm U, Boissonade FM, Westermark A, Robinson PP, Fried K. Sympathetic nerve sprouting fails to occur in the trigeminal ganglion after peripheral nerve injury in the rat. Pain 1999;82: 283–288.

48. Moana-Filho EJ, Herrero Babiloni A, Theis-Mahon NR. Endogenous pain modulation in chronic orofacial pain: A systematic review and meta-analysis. Pain 2018;159:1441–1455.

49. Khan J, Zusman T, Wang Q, Eliav E. Acute and chronic pain in orofacial trauma patients. J Endod 2019;45:S28–S38.

50. Baad-Hansen L, List T, Kaube H, Jensen TS, Svensson P. Blink reflexes in patients with atypical odontalgia and matched healthy controls. Exp Brain Res 2006;172:498–506.

51. Lannersten L, Kosek E. Dysfunction of endogenous pain inhibition during exercise with painful muscles in patients with shoulder myalgia and fibromyalgia. Pain 2010;151:77–86.

52. Bement MK, Sluka KA. Low-intensity exercise reverses chronic muscle pain in the rat in a naloxone-dependent manner. Arch Phys Med Rehabil 2005;86:1736–1740.

53. Crombie KM, Brellenthin AG, Hillard CJ, Koltyn KF. Endocannabinoid and opioid system interactions in exercise-induced hypoalgesia. Pain Med 2018;19:118–123.

54. Dietrich A, McDaniel WF. Endocannabinoids and exercise. Br J Sports Med 2004;38:536–541.

55. Droste C, Meyer-Blankenburg H, Greenlee MW, Roskamm H. Effect of physical exercise on pain thresholds and plasma beta-endorphins in patients with silent and symptomatic myocardial isch-aemia. Eur Heart J 1988;9(suppl N):s25–s33.

56. Lima LV, Abner TSS, Sluka KA. Does exercise increase or decrease pain? Central mechanisms underlying these two phenomena. J Physiol 2017;595:4141–4150.

57. Vaegter HB, Handberg G, Graven-Nielsen T. Similarities between exercise-induced hypoalgesia and conditioned pain modulation in humans. Pain 2014;155:158–167.

58. Vaegter HB, Handberg G, Graven-Nielsen T. Similarities between exercise-induced hypoalgesia and conditioned pain modulation in humans. Pain 2014;155:158–167.

59. Yarnitsky D, Crispel Y, Eisenberg E, et al. Prediction of chronic post-operative pain: Preoperative DNIC testing identifies patients at risk. Pain 2008;138:22–28.

60. Lazaridou A, Paschali M, Schreiber K, et al. The association between daily physical exercise and pain among women with fibromyalgia: The moderating role of pain catastrophizing. Pain Rep 2020;5:e832.

61. Gomolka S, Vaegter HB, Nijs J, et al. Assessing endogenous pain inhibition: Test-retest reliability of exercise-induced hypoalgesia in local and remote body parts after aerobic cycling. Pain Med 2019;20:2272–2282.

62. Hviid JT, Thorlund JB, Vaegter HB. Walking increases pain tolerance in humans: An experimental cross-over study. Scand J Pain 2019;19:813–822.

63. Vaegter HB, Bjerregaard LK, Redin MM, Rasmussen SH, Graven-Nielsen T. Hypoalgesia after bicycling at lactate threshold is reliable between sessions. Eur J Appl Physiol 2019;119:91–102.

64. Vaegter HB, Dørge DB, Schmidt KS, Jensen AH, Graven-Nielsen T. Test-retest reliabilty of exercise-induced hypoalgesia after aerobic exercise. Pain Med 2018;19:2212–2222.

65. Vaegter HB, Lyng KD, Yttereng FW, Christensen MH, Sørensen MB, Graven-Nielsen T. Exercise-induced hypoalgesia after iso-metric wall squat exercise: A test-retest reliabilty study. Pain Med 2019;20:129–137.

66. Van Oosterwijck J, Nijs J, Meeus M, Van Loo M, Paul L. Lack of endogenous pain inhibition during exercise in people with chron-ic whiplash associated disorders: An experimental study. J Pain 2012;13:242–254.

67. Fingleton C, Smart KM, Doody CM. Exercise-induced hypoalgesia in people with knee osteoarthritis with normal and abnormal conditioned pain modulation. Clin J Pain 2017;33:395–404.

68. Vaegter HB, Handberg G, Graven-Nielsen T. Hypoalgesia after exercise and the cold pressor test is reduced in chronic musculoskeletal pain patients with high pain sensitivity. Clin J Pain 2016;32:58–69.

69. Dailey DL, Keffala VJ, Sluka KA. Do cognitive and physical fatigue tasks enhance pain, cognitive fatigue, and physical fatigue in people with fibromyalgia? Arthritis Care Res (Hoboken) 2015;67:288–296.

70. Staud R, Robinson ME, Price DD. Isometric exercise has opposite effects on central pain mechanisms in fibromyalgia patients compared to normal controls. Pain 2005;118:176–184.

71. Bote ME, García JJ, Hinchado MD, Ortega E. An exploratory study of the effect of regular aquatic exercise on the function of neutrophils from women with fibromyalgia: Role of IL-8 and noradrena-line. Brain Behav Immun 2014;39:107–112.

72. Pilat C, Frech T, Wagner A, et al. Exploring effects of a natural combination medicine on exercise-induced inflammatory im-mune response: A double-blind RCT. Scand J Med Sci Sports 2015;25:534–542.

73. Zheng Q, Cui G, Chen J, et al. Regular exercise enhances the immune response against microbial antigens through up-regulation of toll-like receptor signaling pathways. Cell Physiol Biochem 2015;37:735–746.

74. Amaral LSB, Souza CS, Volpini RA, et al. Previous exercise training reduces markers of renal oxidative stress and inflammation in streptozotocin-induced diabetic female rats. J Diabetes Res 2018;2018:6170352.

75. Rezende RM, Gouveia Pelúzio MdCG, de Jesus Silva F, et al. Does aerobic exercise associated with tryptophan supplementation attenuates hyperalgesia and inflammation in female rats with experimental fibromyalgia? PLoS One 2019;14:e0211824.

76. Clark AK, Old EA, Malcangio M. Neuropathic pain and cytokines: Current perspectives. J Pain Res 2013;6:803–814.

77. Noma N, Khan J, Chen IF, et al. Interleukin-17 levels in rat models of nerve damage and neuropathic pain. Neurosci Lett 2011;493:86–91.

78. Khan J, Ramadan K, Korczeniewska O, Anwer MM, Benoliel R, Eliav E. Interleukin-10 levels in rat models of nerve damage and neuropathic pain. Neurosci Lett 2015;592:99–106.

79. Khan J, Wang Q, Ren Y, et al. Exercise induced hypoalgesia profile in rats is associated with IL-10 and IL-1 beta levels and pain sever-ity following nerve injury. Cytokine 2021;143:155540.

80. Khan J, Hassun H, Zusman T, Korczeniewska O, Eliav E. Interleukin-8 levels in rat models of nerve damage and neuropathic pain. Neurosci Lett 2017;657:106–112.

81. Eliav E, Benoliel R, Herzberg U, Kalladka M, Tal M. The role of IL-6 and IL-1beta in painful perineural inflammatory neuritis. Brain Behav Immun 2009;23:474–484.

82. da Silva MD, Bobinski F, Sato KL, Kolker SJ, Sluka KA, Santos AR. IL-10 cytokine released from M2 macrophages is crucial for analgesic and anti-inflammatory effects of acupuncture in a model of inflammatory muscle pain. Mol Neurobiol 2015;51:19–31.

83. Shao Q, Li Y, Wang Q, Zhao J. IL-10 and IL-1beta mediate neuropathic-pain like behavior in the ventrolateral orbital cortex. Neurochem Res 2015;40:733–739.

84. Soderquist RG, Sloane EM, Loram LC, et al. Release of plasmid DNA-encoding IL-10 from PLGA microparticles facilitates long-term reversal of neuropathic pain following a single intrathecal administration. Pharm Res 2010;27:841–854.

85. Koltyn KF. Analgesia following exercise: A review. Sports Med 2000;29:85–98.

86. Welch SP. Interaction of the cannabinoid and opioid systems in the modulation of nociception. Int Rev Psychiatry 2009;21: 143–151.

87. Tesarz J, Gerhardt A, Schommer K, Treede RD, Eich W. Alterations in endogenous pain modulation in endurance athletes: An experimental study using quantitative sensory testing and the cold-pres-sor task. Pain 2013;154:1022–1029.

88. Janal MN, Glusman M, Kuhl JP, Clark CW. Are runners stoical? An examination of pain sensitivity in habitual runners and normally active controls. Pain 1994;58:109–116.

89. McDougall J, Jutzeler CR, Scott A, Crocker PRE, Kramer JLK. Conditioned pain modulation in elite athletes: A systematic review and meta-analysis. Scand J Pain 2020;20:429–438.


Abstracted / indexed in

Science Citation Index (SCI)

Science Citation Index Expanded (SCIE)

BIOSIS Previews

Scopus

Cumulative Index to Nursing and Allied Health Literature (CINAHL)

Submission Turnaround Time

Conferences

Top