Perineural resiniferatoxin selectively inhibits inflammatory hyperalgesia

John K Neubert, Andrew J Mannes, Laszlo J Karai, Alan C Jenkins, Lanel Zawatski, Mones Abu-Asab, Michael J Iadarola, John K Neubert, Andrew J Mannes, Laszlo J Karai, Alan C Jenkins, Lanel Zawatski, Mones Abu-Asab, Michael J Iadarola

Abstract

Resiniferatoxin (RTX) is an ultrapotent capsaicin analog that binds to the transient receptor potential channel, vanilloid subfamily member 1 (TRPV1). There is a large body of evidence supporting a role for TRPV1 in noxious-mediated and inflammatory hyperalgesic responses. In this study, we evaluated low, graded, doses of perineural RTX as a method for regional pain control. We hypothesized that this approach can provide long-term, but reversible, blockade of a portion of nociceptive afferent fibers within peripheral nerves when given at a site remote from the neuronal perikarya in the dorsal root ganglia. Following perineural RTX application to the sciatic nerve, we demonstrated a significant inhibition of inflammatory nociception that was dose- and time-dependent. At the same time, treated animals maintained normal proprioceptive sensations and motor control, and other nociceptive responses were largely unaffected. Using a range of mechanical and thermal algesic tests, we found that the most sensitive measure following perineural RTX administration was inhibition of inflammatory hyperalgesia. Recovery studies showed that physiologic sensory function could return as early as two weeks post-RTX treatment, however, immunohistochemical examination of the DRG revealed a partial, but significant reduction in the number of the TRPV1-positive neurons. We propose that this method could represent a beneficial treatment for a range of chronic pain problems, including neuropathic and inflammatory pain not responding to other therapies.

Figures

Figure 1
Figure 1
Locating the sciatic nerve for electrically-stimulated percutaneous, perineural RTX injection. Note finger palpation locations and direction of the needle (see Methods). Fluorescein dye injection (50 μl, 1 μg/μl) using this technique demonstrates the distribution of dye-containing fluid within the perineurium of the nerve (B).
Figure 2
Figure 2
Percutaneous application of RTX to the sciatic nerve and sciatic plus saphenous nerves minimally effects response to heat stimuli. RTX (250 ng, 50 μl) application to only the sciatic nerve produced a transient change in heat withdrawal latency with respect to testing time (*P < 0.05, 1 way ANOVA, Scheffe post-hoc analysis), as compared to the baseline. Additional application of RTX to the saphenous nerve produced a modest increase in heat latency, but this effect was not significantly different, as compared to the sciatic group. The "sciatic nerve" group refers to animals treated only with percutaneous RTX (N = 24) and the "sciatic and saphenous nerves" group refers to animals that were injected percutaneously near the sciatic nerve and had an open field injection around the saphenous nerve (N = 15). The "control group" represents the uninjected right hindpaw. There was no difference between contralateral controls for the sciatic nerve group (N = 24) and the sciatic/saphenous group (N = 15), therefore these groups were combined (N = 39) for subsequent comparisons.
Figure 3
Figure 3
RTX blocks inflammatory heat hyperalgesia when applied directly to the sciatic nerve. Both percutaneous and open field injection of RTX (250 ng, 50 μl) inhibited inflammatory heat hyperalgesia following carrageenan injection (6 mg, 150 μl). There was no significant difference in latency time regarding RTX application method (i.e. percutaneously vs. direct open field application); therefore data were pooled (N = 15). However, for this set of animals, there was a significant increase in normal heat latency as compared to vehicle treated animals (*P < 0.05, 1 way ANOVA, Scheffe post-hoc analysis). Animals pretreated with vehicle (0.25% Tween-80, PBS 50 μl, N = 10) had a significant decrease in their heat withdrawal latency (+P < 0.05, 1 way RM-ANOVA) following induction of inflammation.
Figure 4
Figure 4
Percutaneous RTX produces long-lasting, reversible inflammatory heat hyperalgesic inhibition (A) in a dose-dependent fashion (B). Inflammatory heat hyperalgesia is significantly inhibited 1 day (N = 8), 1 week (N = 6), and 2 weeks (N = 5) following application of RTX (250 ng, 50 μl) to the sciatic nerve (*P < 0.05, 1 way ANOVA). An intermediate response was seen at 1 month (N = 8), and at 3 months (N = 8). Complete recovery with a normal heat withdrawal response following inflammation was observed 6 months (N = 11) following treatment, indicating that the effect of RTX had reversed. The dose-response results (B) indicate that ≥ 125 ng is necessary for a significant anti-inflammatory effect, as compared to Pre-Inflammation values and to vehicle (P < 0.05). Note that the Pre-inflammation testing point represents the same testing day that the animal was to be inflamed. There was no difference between groups treated with RTX (N = 46); therefore data were pooled for the baseline (Baseline) and Pre-inflammation testing sessions. Animals treated with vehicle (N = 16) were also pooled in the Baseline, Pre-inflammation, and Post-inflammation groups for comparison.
Figure 5
Figure 5
RTX does not affect normal mechanical sensitivity or rotarod performance. Withdrawal force latencies were similar for vehicle and RTX treated animals except after inflammation, where there was a significant difference (*P < 0.05, Mann-Whitney rank sum test) between the two groups (A). Data presented as a median normalized threshold (bars: interquartile range). Pre-inflammation values for both vehicle and RTX-treated groups were all significant versus the post-inflammation value (Krukal-Wallis ANOVA, P < 0.05). Values are represented by a median normalized threshold and the interquartile range is denoted by the bars. Rotarod performance was identical for both groups (B).
Figure 6
Figure 6
Dorsal root gangliaTRPV1 immunopositive cells are eliminated following perineural RTX treatment. Lumbar dorsal root ganglia sections (7 μm, paraffin-embedded) were stained for TRPV1 following RTX, vehicle or no treatment of the sciatic nerve. There was a significant difference in the number of TRPV1 positively stained cells in the RTX-treated animals, as compared to vehicle and untreated animals (Table inset).
Figure 7
Figure 7
RTX blocks capsaicin-induced efferent neurogenic inflammation. Following Evans blue injection (i.v., 30 mg/kg, 2% solution), a 1% capsaicin cream was liberally applied to the shaved hind legs and paws. Areas of neurogenic-induced plasma extravasation are seen as blue. Note the delineation between the blue area (positive for plasma extravasation) versus the white skin (negative for plasma extravasation) for the RTX treated side (A, C) and extravasation on only the three medial toes for the RTX-treated side. Perineural vehicle application did not effect plasma extravasation (B). These data indicate selective targeting of the sciatic nerve while preserving the saphenous division via this percutaneous injection.

References

    1. Caterina MJ, Schumacher MA, Tominaga M, Rosen TA, Levine JD, Julius D. The capsaicin receptor: a heat-activated ion channel in the pain pathway. Nature. 1997;389:816–824. doi: 10.1038/39807.
    1. Montell C, Birnbaumer L, Flockerzi V, Bindels RJ, Bruford EA, Caterina MJ, Clapham DE, Harteneck C, Heller S, Julius D, Kojima I, Mori Y, Penner R, Prawitt D, Scharenberg AM, Schultz G, Shimizu N, Zhu MX. A unified nomenclature for the superfamily of TRP cation channels. Mol Cell. 2002;9:229–231. doi: 10.1016/S1097-2765(02)00448-3.
    1. Gunthorpe MJ, Benham CD, Randall A, Davis JB. The diversity in the vanilloid (TRPV) receptor family of ion channels. Trends Pharmacol Sci. 2002;23:183–191. doi: 10.1016/S0165-6147(02)01999-5.
    1. Caterina MJ, Leffler A, Malmberg AB, Martin WJ, Trafton J, Petersen-Zeitz KR, Koltzenburg M, Basbaum AI, Julius D. Impaired nociception and pain sensation in mice lacking the capsaicin receptor. Science. 2000;288:306–313. doi: 10.1126/science.288.5464.306.
    1. Davis JB, Gray J, Gunthorpe MJ, Hatcher JP, Davey PT, Overend P, Harries MH, Latcham J, Clapham C, Atkinson K, Hughes SA, Rance K, Grau E, Harper AJ, Pugh PL, Rogers DC, Bingham S, Randall A, Sheardown SA. Vanilloid receptor-1 is essential for inflammatory thermal hyperalgesia. Nature. 2000;405:183–187. doi: 10.1038/35012076.
    1. Xu XJ, Farkas-Szallasi T, Lundberg JM, Hokfelt T, Wiesenfeld-Hallin Z, Szallasi A. Effects of the capsaicin analogue resiniferatoxin on spinal nociceptive mechanisms in the rat: behavioral, electrophysiological and in situ hybridization studies. Brain Res. 1997;752:52–60. doi: 10.1016/S0006-8993(96)01444-8.
    1. Szabo T, Olah Z, Iadarola MJ, Blumberg PM. Epidural resiniferatoxin induced prolonged regional analgesia to pain. Brain Res. 1999;840:92–98. doi: 10.1016/S0006-8993(99)01763-1.
    1. Neubert JK, Karai L, Jun JH, Kim HS, Olah Z, Iadarola MJ. Peripherally induced resiniferatoxin analgesia. Pain. 2003;104:219–228. doi: 10.1016/S0304-3959(03)00009-5.
    1. Karai L, Brown DC, Mannes AJ, Connelly ST, Brown J, Gandal M, Wellisch OM, Neubert JK, Olah Z, Iadarola MJ. Deletion of vanilloid receptor 1-expressing primary afferent neurons for pain control. J Clin Invest. 2004;113:1344–1352. doi: 10.1172/JCI200420449.
    1. Dray A. Mechanism of action of capsaicin-like molecules on sensory neurons. Life Sci. 1992;51:1759–1765. doi: 10.1016/0024-3205(92)90045-Q.
    1. Appendino G, Cravotto G, Palmisano G, Annunziata R, Szallasi A. Synthesis and evaluation of phorboid 20-homovanillates: discovery of a class of ligands binding to the vanilloid (capsaicin) receptor with different degrees of cooperativity. J Med Chem. 1996;39:3123–3131. doi: 10.1021/jm960063l.
    1. Pomonis JD, Harrison JE, Mark L, Bristol DR, Valenzano KJ, Walker K. N-(4-Tertiarybutylphenyl)-4-(3-cholorphyridin-2-yl)tetrahydropyrazine -1(2H)-carbox-amide (BCTC), a novel, orally effective vanilloid receptor 1 antagonist with analgesic properties: II. in vivo characterization in rat models of inflammatory and neuropathic pain. J Pharmacol Exp Ther. 2003;306:387–393. doi: 10.1124/jpet.102.046268.
    1. Varga A, Nemeth J, Szabo A, McDougall JJ, Zhang C, Elekes K, Pinter E, Szolcsanyi J, Helyes Z. Effects of the novel TRPV1 receptor antagonist SB366791 in vitro and in vivo in the rat. Neurosci Lett. 2005;385:137–142. doi: 10.1016/j.neulet.2005.05.015.
    1. Gavva NR, Tamir R, Qu Y, Klionsky L, Zhang TJ, Immke D, Wang J, Zhu D, Vanderah TW, Porreca F, Doherty EM, Norman MH, Wild KD, Bannon AW, Louis JC, Treanor JJ. AMG 9810 [(E)-3-(4-t-butylphenyl)-N-(2,3-dihydrobenzo[b][1,4] dioxin-6-yl)acrylamide], a novel vanilloid receptor 1 (TRPV1) antagonist with antihyperalgesic properties. J Pharmacol Exp Ther. 2005;313:474–484. doi: 10.1124/jpet.104.079855.
    1. Levine JD, Alessandri-Haber N. TRP channels: targets for the relief of pain. Biochim Biophys Acta. 2007;1772:989–1003.
    1. Cortright DN, Krause JE, Broom DC. TRP channels and pain. Biochim Biophys Acta. 2007;1772:978–988.
    1. Caudle RMK., L.; Mena, N.; Cooper, B.Y.; Mannes, A.J.; Perez, F.M.; Iadarola, M.J.; Olah, Z. Resiniferatoxin induced loss of plasma membrane in vanilloid receptor expressing cells. Neurotoxicology. 2003;24:895–908. doi: 10.1016/S0161-813X(03)00146-3.
    1. Olah Z, Szabo T, Karai L, Hough C, Fields RD, Caudle RM, Blumberg PM, Iadarola MJ. Ligand-induced dynamic membrane changes and cell deletion conferred by vanilloid receptor 1. J Biol Chem. 2001;276:11021–11030. doi: 10.1074/jbc.M008392200.
    1. Karai LC., T.; Olah, Z.; Mannes, A.J.; Iadarola, M.J. Evaluation of intraganglionic resiniferatoxin (RTX) injection for pain control [Abstract] Society for Neuroscience Abstracts. 2001.
    1. Brown DC, Iadarola MJ, Perkowski SZ, Erin H, Shofer F, Laszlo KJ, Olah Z, Mannes AJ. Physiologic and antinociceptive effects of intrathecal resiniferatoxin in a canine bone cancer model. Anesthesiology. 2005;103:1052–1059. doi: 10.1097/00000542-200511000-00020.
    1. Menendez L, Juarez L, Garcia E, Garcia-Suarez O, Hidalgo A, Baamonde A. Analgesic effects of capsazepine and resiniferatoxin on bone cancer pain in mice. Neurosci Lett. 2006;393:70–73. doi: 10.1016/j.neulet.2005.09.046.
    1. Szolcsanyi J. Capsaicin and nociception. Acta Physiol Hung. 1987;69:323–332.
    1. Jancso G, Kiraly E, Jancso-Gabor A. Direct evidence for an axonal site of action of capsaicin. Naunyn Schmiedebergs Arch Pharmacol. 1980;313:91–94. doi: 10.1007/BF00505809.
    1. Kissin I, Bright CA, Bradley EL., Jr. Selective and long-lasting neural blockade with resiniferatoxin prevents inflammatory pain hypersensitivity. Anesth Analg. 2002;94:1253–8, table of contents. doi: 10.1097/00000539-200205000-00038.
    1. Kissin I, Davison N, Bradley EL., Jr. Perineural resiniferatoxin prevents hyperalgesia in a rat model of postoperative pain. Anesth Analg. 2005;100:774–80, table of contents. doi: 10.1213/01.ANE.0000143570.75908.7F.
    1. Iadarola MJ, Douglass J, Civelli O, Naranjo JR. Differential activation of spinal cord dynorphin and enkephalin neurons during hyperalgesia: evidence using cDNA hybridization. Brain Res. 1988;455:205–212. doi: 10.1016/0006-8993(88)90078-9.
    1. Hargreaves K, Dubner R, Brown F, Flores C, Joris J. A new and sensitive method for measuring thermal nociception in cutaneous hyperalgesia. Pain. 1988;32:77–88. doi: 10.1016/0304-3959(88)90026-7.
    1. Wall PD, Devor M, Inbal R, Scadding JW, Schonfeld D, Seltzer Z, Tomkiewicz MM. Autotomy following peripheral nerve lesions: experimental anaesthesia dolorosa. Pain. 1979;7:103–111. doi: 10.1016/0304-3959(79)90002-2.
    1. Jancso G, Lawson SN. Transganglionic degeneration of capsaicin-sensitive C-fiber primary afferent terminals. Neuroscience. 1990;39:501–511. doi: 10.1016/0306-4522(90)90286-D.
    1. Mays KS, Lipman JJ, Schnapp M. Local analgesia without anesthesia using peripheral perineural morphine injections. Anesth Analg. 1987;66:417–420. doi: 10.1213/00000539-198705000-00008.
    1. Chung JM, Lee KH, Hori Y, Willis WD. Effects of capsaicin applied to a peripheral nerve on the responses of primate spinothalamic tract cells. Brain Res. 1985;329:27–38. doi: 10.1016/0006-8993(85)90509-8.
    1. Fitzgerald M, Woolf CJ. The time course and specificity of the changes in the behavioural and dorsal horn cell responses to noxious stimuli following peripheral nerve capsaicin treatment in the rat. Neuroscience. 1982;7:2051–2056. doi: 10.1016/0306-4522(82)90119-1.
    1. Pini A, Baranowski R, Lynn B. Long-Term Reduction in the Number of C-Fibre Nociceptors Following Capsaicin Treatment of a Cutaneous Nerve in Adult Rats. Eur J Neurosci. 1990;2:89–97. doi: 10.1111/j.1460-9568.1990.tb00384.x.
    1. Szallasi A, Cruz F, Geppetti P. TRPV1: a therapeutic target for novel analgesic drugs? Trends Mol Med. 2006;12:545–554. doi: 10.1016/j.molmed.2006.09.001.
    1. Krause JE, Chenard BL, Cortright DN. Transient receptor potential ion channels as targets for the discovery of pain therapeutics. Curr Opin Investig Drugs. 2005;6:48–57.

Source: PubMed

3
구독하다