Endogenous Opioid-Masked Latent Pain Sensitization: Studies from Mouse to Human

Manuel P Pereira, Renee R Donahue, Jørgen B Dahl, Marianne Werner, Bradley K Taylor, Mads U Werner, Manuel P Pereira, Renee R Donahue, Jørgen B Dahl, Marianne Werner, Bradley K Taylor, Mads U Werner

Abstract

Following the resolution of a severe inflammatory injury in rodents, administration of mu-opioid receptor inverse agonists leads to reinstatement of pain hypersensitivity. The mechanisms underlying this form of latent pain sensitization (LS) likely contribute to the development of chronic pain, but LS has not yet been demonstrated in humans. Using a C57BL/6 mouse model of cutaneous mild heat injury (MHI) we demonstrated a dose-dependent reinstatement of pain sensitization, assessed as primary (P < 0.001) and secondary hyperalgesia (P < 0.001) by naloxone (0.3–10 mg/kg), 168 hrs after the induction of MHI. Forward-translating the dose data to a human MHI model (n = 12) we could show that LS does indeed occur after naloxone 2 mg/kg, 168 hrs after a MHI. Our previous unsuccessful efforts to demonstrate unmasking of LS in humans are thus likely explained by an insufficient naloxone dose (0.021 mg/kg). However, while LS was consistently demonstrated in 21/24 mice, LS was only seen in 4/12 subjects. This difference is likely due to selection bias since the C57BL/6 mouse strain exhibits markedly enhanced pain sensitivity in assays of acute thermal nociception. Future exploratory studies in humans should prioritize inclusion of “high-sensitizers” prone to develop LS and use post-surgical models to elucidate markers of vulnerability to chronic postsurgical pain.

Trial registration: EudraCT 2012-005663-27.

Conflict of interest statement

Competing Interests: The authors have declared that no competing interests exist.

Figures

Fig 1. CONSORT flow-algorithm for human subjects.
Fig 1. CONSORT flow-algorithm for human subjects.
Four subjects were allocated to a feasibility study. One subject was excluded from this part due to an unsuspected finding of an abnormal ultrasound cardiography. Fifteen subjects were included and randomized. Three subjects were excluded for reasons not related to the study. Twelve subjects completed the study and data were examined in an interim analysis by an independent statistician.
Fig 2. Study timeline in the human…
Fig 2. Study timeline in the human MHI-model.
Day 1/Day 3 include induction of mild heat injury (MHI) with baseline assessments (green rectangle), heat injury (red) and post-injury assessments (blue). Day 2/Day 4 include a pre-drug assessment (blue; post-injury 165 hrs), drug-infusions (naloxone or placebo; grey) and post-infusion assessments (magenta). Stars indicate assessments of mechanical pain thresholds (red star), thermal thresholds (yellow) and secondary hyperalgesia areas (green).
Fig 3. Hyperalgesia was assessed by a…
Fig 3. Hyperalgesia was assessed by a decrease in paw withdrawal threshold (PWT50%) in a model of mild heat injury (MHI).
Line graphs illustrate response to von Frey (vF) stimulation at baseline (BL) and at 1, 2, 3, 72, and 168 hrs after MHI. Naloxone reinstated primary hyperalgesia (A-B) at a dose of 0.3 mg/kg (60 min, P = 0.0015), 3 mg/kg (30 min, P = 0.0011; 60 min, P = 0.0002), and 10 mg/kg (30 min, P < 0.0001; 60 min, P < 0.0001; 120 min, P < 0.0001) and secondary hyperalgesia (C-D) at a dose of 0.3 mg/kg (60 min, P = 0.0148), 3 mg/kg (30 min, P = 0.0004; 60 min, P = 0.0002), and 10 mg/kg (30 min, P < 0.0001; 60 min, P < 0.0001; 120 min, P = 0.0013). A,C: Line drawings. B,D: Histograms of the post-injection time points (plotted as the mean of the 30–120 min time points). Values represent mean ± 95% CI. * P < 0.05.
Fig 4. Heat pain thresholds and secondary…
Fig 4. Heat pain thresholds and secondary hyperalgesia areas before and after a mild heat injury.
Heat pain thresholds (white circles; left y-axis) and secondary hyperalgesia areas (red circles; right y-axis) before and, 60, 120 and 180 min, after a mild heat injury (MHI). Values represent mean of Day 1 and Day 3 values (mean ± 95% CI). * P < 0.01; ** P < 0.005; **** P < 0.0005.
Fig 5. Individual trajectories of secondary hyperalgesia…
Fig 5. Individual trajectories of secondary hyperalgesia areas during the early and late phase after a mild heat injury.
Individual trajectories of secondary hyperalgesia areas (n = 12) during the early phase (0–3 hrs) and the late phase (165–169 hrs) after induction of the mild heat injury (MHI). Data are from naloxone (A) and placebo (B) sessions. The trajectories of the four naloxone responders, indicating presence of endogenous opioid masked sensitization, are delineated by red circles and the trajectories from the eight non-responders by blue filled circles. MHI and drug administration are indicated by vertical arrows. One (#11) of the responders demonstrated residual SHA at 165 hrs after the induction of MHI (indicated by oblique arrow; B), but not during placebo. Secondary hyperalgesia areas developed in 4/12 subjects (#2,6,7,11) after naloxone (A) and in 0/12 after placebo (B).

References

    1. Woolf CJ (2011) Central sensitization: implications for the diagnosis and treatment of pain. Pain 152: S2–15. S0304-3959(10)00584-1 [pii]; 10.1016/j.pain.2010.09.030
    1. Latremoliere A, Woolf CJ (2009) Central sensitization: a generator of pain hypersensitivity by central neural plasticity. J Pain 10: 895–926. 10.1016/j.jpain.2009.06.012
    1. Corder G, Doolen S, Donahue RR, Winter MK, Jutras BL, He Y, et al. (2013) Constitutive mu-opioid receptor activity leads to long-term endogenous analgesia and dependence. Science 341: 1394–1399. 341/6152/1394 [pii]; 10.1126/science.1239403
    1. Schweinhardt P, Sauro KM, Bushnell MC (2008) Fibromyalgia: a disorder of the brain? Neuroscientist 14: 415–421. 1073858407312521 [pii]; 10.1177/1073858407312521
    1. Staud R, Weyl EE, Price DD, Robinson ME (2012) Mechanical and heat hyperalgesia highly predict clinical pain intensity in patients with chronic musculoskeletal pain syndromes. J Pain 13: 725–735. S1526-5900(12)00602-5 [pii]; 10.1016/j.jpain.2012.04.006
    1. Watson M, Lucas C, Hoy A, Back I (2005) Principles of drug use in palliative medicine In: Watson M, Lucas C, Hoy A, Back I, editors. Oxford handbook of palliative care. Oxford University Press; pp. 35–79.
    1. Cohen MR, Cohen RM, Pickar D, Sunderland T, Mueller EA III, Murphy DL (1984) High dose naloxone in depression. Biol Psychiatry 19: 825–832.
    1. Cohen MR, Pickar D, Cohen RM (1985) High-dose naloxone administration in chronic schizophrenia. Biol Psychiatry 20: 573–575. 0006-3223(85)90030-7 [pii].
    1. Edwards RR, Ness TJ, Fillingim RB (2004) Endogenous opioids, blood pressure, and diffuse noxious inhibitory controls: a preliminary study. Percept Mot Skills 99: 679–687.
    1. Cohen MR, Cohen RM, Pickar D, Murphy DL, Bunney WE Jr. (1982) Physiological effects of high dose naloxone administration to normal adults. Life Sci 30: 2025–2031.
    1. Nozaki-Taguchi N, Yaksh TL (1998) A novel model of primary and secondary hyperalgesia after mild thermal injury in the rat. Neurosci Lett 254: 25–28. S0304-3940(98)00648-X [pii].
    1. Ravn P, Secher EL, Skram U, Therkildsen T, Christrup LL, Werner MU (2013) Morphine- and buprenorphine-induced analgesia and antihyperalgesia in a human inflammatory pain model: a double-blind, randomized, placebo-controlled, five-arm crossover study. J Pain Res 6: 23–38. 10.2147/JPR.S36827 jpr-6-023 [pii].
    1. Pereira MP, Werner MU, Ringsted TK, Rowbotham MC, Taylor BK, Dahl JB (2013) Does naloxone reinstate secondary hyperalgesia in humans after resolution of a burn injury? A placebo-controlled, double-blind, randomized, cross-over study. PLoS One 8: e64608 10.1371/journal.pone.0064608 PONE-D-13-08611 [pii].
    1. Werner MU, Petersen KL, Rowbotham MC, Dahl JB (2013) Healthy volunteers can be phenotyped using cutaneous sensitization pain models. PLoS One 8: e62733 10.1371/journal.pone.0062733 PONE-D-13-02721 [pii].
    1. Dixon WJ (1991) Staircase bioassay: the up-and-down method. Neurosci Biobehav Rev 15: 47–50. S0149-7634(05)80090-9 [pii].
    1. Chaplan SR, Bach FW, Pogrel JW, Chung JM, Yaksh TL (1994) Quantitative assessment of tactile allodynia in the rat paw. J Neurosci Methods 53: 55–63. 0165-0270(94)90144-9 [pii].
    1. Snaith RP (2003) The Hospital Anxiety And Depression Scale. Health Qual Life Outcomes 1: 29 10.1186/1477-7525-1-29
    1. Zigmond AS, Snaith RP (1983) The hospital anxiety and depression scale. Acta Psychiatr Scand 67: 361–370.
    1. Sullivan MJL, Bishop SR, Kivik J (1995) The pain catastrophizing scale: development and validation. Psych Assess 7: 524–532.
    1. Campillo A, Cabanero D, Romero A, Garcia-Nogales P, Puig MM (2011) Delayed postoperative latent pain sensitization revealed by the systemic administration of opioid antagonists in mice. Eur J Pharmacol 657: 89–96. S0014-2999(11)00117-8 [pii]; 10.1016/j.ejphar.2011.01.059
    1. Le RC, Laboureyras E, Gavello-Baudy S, Chateauraynaud J, Laulin JP, Simonnet G (2011) Endogenous opioids released during non-nociceptive environmental stress induce latent pain sensitization Via a NMDA-dependent process. J Pain 12: 1069–1079. S1526-5900(11)00592-X [pii]; 10.1016/j.jpain.2011.04.011
    1. Turk DC, Wilson HD, Cahana A (2011) Treatment of chronic non-cancer pain. Lancet 377: 2226–2235. S0140-6736(11)60402-9 [pii]; 10.1016/S0140-6736(11)60402-9
    1. Murray CJ, Vos T, Lozano R, Naghavi M, Flaxman AD, Michaud C, et al. (2012) Disability-adjusted life years (DALYs) for 291 diseases and injuries in 21 regions, 1990–2010: a systematic analysis for the Global Burden of Disease Study 2010. Lancet 380: 2197–2223. S0140-6736(12)61689-4 [pii]; 10.1016/S0140-6736(12)61689-4
    1. Katz J, Seltzer Z (2009) Transition from acute to chronic postsurgical pain: risk factors and protective factors. Expert Rev Neurother 9: 723–744. 10.1586/ern.09.20
    1. Johansen A, Schirmer H, Stubhaug A, Nielsen CS (2014) Persistent post-surgical pain and experimental pain sensitivity in the Tromso study: comorbid pain matters. Pain 155: 341–348. S0304-3959(13)00557-5 [pii]; 10.1016/j.pain.2013.10.013
    1. Mogil JS, Wilson SG, Bon K, Lee SE, Chung K, Raber P, et al. (1999) Heritability of nociception I: responses of 11 inbred mouse strains on 12 measures of nociception. Pain 80: 67–82.
    1. Bryant CD, Zhang NN, Sokoloff G, Fanselow MS, Ennes HS, Palmer AA, et al. (2008) Behavioral differences among C57BL/6 substrains: implications for transgenic and knockout studies. J Neurogenet 22: 315–331. 906582175 [pii]; 10.1080/01677060802357388
    1. Mogil JS, Wilson SG, Bon K, Lee SE, Chung K, Raber P, et al. (1999) Heritability of nociception II. 'Types' of nociception revealed by genetic correlation analysis. Pain 80: 83–93.
    1. Werner MU, Ringsted TK, Kehlet H, Wildgaard K (2013) Sensory testing in patients with postthoracotomy pain syndrome: part 1: mirror-image sensory dysfunction. Clin J Pain 29: 775–783. 10.1097/AJP.0b013e318277b646
    1. Frost JJ, Douglass KH, Mayberg HS, Dannals RF, Links JM, Wilson AA, et al. (1989) Multicompartmental analysis of [11C]-carfentanil binding to opiate receptors in humans measured by positron emission tomography. J Cereb Blood Flow Metab 9: 398–409. 10.1038/jcbfm.1989.59

Source: PubMed

3
Subscribe