Ablation of rat TRPV1-expressing Adelta/C-fibers with resiniferatoxin: analysis of withdrawal behaviors, recovery of function and molecular correlates

Kendall Mitchell, Brian D Bates, Jason M Keller, Matthew Lopez, Lindsey Scholl, Julia Navarro, Nicholas Madian, Gal Haspel, Michael I Nemenov, Michael J Iadarola, Kendall Mitchell, Brian D Bates, Jason M Keller, Matthew Lopez, Lindsey Scholl, Julia Navarro, Nicholas Madian, Gal Haspel, Michael I Nemenov, Michael J Iadarola

Abstract

Background: Ablation of TRPV1-expressing nociceptive fibers with the potent capsaicin analog resiniferatoxin (RTX) results in long lasting pain relief. RTX is particularly adaptable to focal application, and the induced chemical axonopathy leads to analgesia with a duration that is influenced by dose, route of administration, and the rate of fiber regeneration. TRPV1 is expressed in a subpopulation of unmyelinated C- and lightly myelinated Adelta fibers that detect changes in skin temperature at low and high rates of noxious heating, respectively. Here we investigate fiber-type specific behaviors, their time course of recovery and molecular correlates of axon damage and nociception using infrared laser stimuli following an RTX-induced peripheral axonopathy.

Results: RTX was injected into rat hind paws (mid-plantar) to produce thermal hypoalgesia. An infrared diode laser was used to stimulate Adelta fibers in the paw with a small-diameter (1.6 mm), high-energy, 100 msec pulse, or C-fibers with a wide-diameter (5 mm), long-duration, low-energy pulse. We monitored behavioral responses to indicate loss and regeneration of fibers. At the site of injection, responses to C-fiber stimuli were significantly attenuated for two weeks after 5 or 50 ng RTX. Responses to Adelta stimuli were significantly attenuated for two weeks at the highest intensity stimulus, and for 5 weeks to a less intense Adelta stimulus. Stimulation on the toe, a site distal to the injection, showed significant attenuation of Adelta responses for 7- 8 weeks after 5 ng, or 9-10 weeks after 50 ng RTX. In contrast, responses to C-fiber stimuli exhibited basically normal responses at 5 weeks after RTX. During the period of fiber loss and recovery, molecular markers for nerve regeneration (ATF3 and galanin) are upregulated in the dorsal root ganglia (DRG) when behavior is maximally attenuated, but markers of nociceptive activity (c-Fos in spinal cord and MCP-1 in DRG), although induced immediately after RTX treatment, returned to normal.

Conclusion: Behavioral recovery following peripheral RTX treatment is linked to regeneration of TRPV1-expressing Adelta and C-fibers and sustained expression of molecular markers. Infrared laser stimulation is a potentially valuable tool for evaluating the behavioral role of Adelta fibers in pain and pain control.

Figures

Figure 1
Figure 1
Detection of early-onset nocifensive behaviors prior to withdrawal of stimulated limb. A 100 msec thermal pulse at high power (6.08 W/mm2) was used to stimulate the right index toe. Withdrawal responses were recorded at a rate of 500 frames per second. The position of the rat's head before and after stimulation are highlighted in A and B, respectively. The upper and lower arrowheads in each frame bring attention to the eyes and nose, respectively. Abrupt head displacement in (A) versus (B) occurs by 110 msec (140-30 msec, see Methods). Also note in B the orbital "tightening", which was observed only with some rats following laser stimulation. (C) By 210 msec, the head was oriented towards the stimulated foot and toes on the stimulated foot have moved. (D) Withdrawal of the stimulated paw was underway by 310 msec. (E) and (F) are magnifications of (A) and (B), respectively, showing that by 110 msec, toe movement on the non-stimulated hind paw has preceded any movement detected in the stimulated limb. Full videos can be viewed in the Additional file 1 Movie S1 data and show that latency to the first detectable movement was approximately 74 msec.
Figure 2
Figure 2
Effect of RTX on Aδ and C responses at the mid-plantar injection site. Either 5 or 50 ng of RTX was injected into the right while vehicle was injected into the left hind paw. Aδ and C responses before and after injections were followed for several weeks. (A-D and F) Aδ responses following intraplantar RTX-injection. We used probability of withdrawal (A, C and F) and the intensity of withdrawal (B and D) as endpoints for the Aδ assay. Behavioral responses to stimulus intensities at 5.12 W/mm2 (A, B and F) or 6.08 W/mm2 (C and D) are shown. (E) Loss and return of C-fiber responses after RTX-intraplantar injection. We used latency to paw withdrawal as the endpoint for the C-fiber assay; a stimulus intensity was chosen that produced a response latency of ~8 sec in normal rats. A cutoff of 16 sec (dashed line) was imposed to prevent tissue damage. (F and G) Direct comparison of behavioral responses at day 18 post-RTX to Aδ (F) and C (G) stimuli. Data used in week 3, in the line graphs, are the average of tests done on days 16, 18 and 21. Hypoalgesia at day 18 was maintained in Aδ but not C-fibers. The tables under the graphs denote statistical comparisons between vehicle and 5 ng or 50 ng RTX, which are aligned with the post-treatment time points. Each data point is an average of the combined responses from 2 or 3 non-consecutive days of testing during the time period indicated on the x-axis (weeks). For graphical simplicity, the data from all vehicle-treated rats (n = 12) is combined, yielding a single line in the plots, since there was no significant difference between the two vehicle groups. 2-way ANOVA with repeated measures was used to compare the effect of 5 ng or 50 ng RTX versus vehicle. 2-way ANOVA was used to compare the effects of 5 ng versus 50 ng. Fisher's exact test was used in F. *,† p < 0.5, **,†† p < 0.01, ***, ††† p < 0.001, n = 6 rats per group.
Figure 3
Figure 3
Aδ- and C-fiber responses in the toes after mid-plantar RTX injection. The infrared diode laser was used to stimulate Aδ- or C-fibers in the toes of the same rats used in Figure 2. (A-D) Responses to Aδ stimuli are diminished for up to 10 weeks. Plots are similar to those in Figure 2. (E) For 50 ng RTX only, a small but significant difference was measured in the toes at 5 weeks (but not at 10 weeks) for C responses; data reported are an average of 3 test days. Our initial protocol used C-fiber testing only at the mid-plantar footpad; however, since we observed prolonged, diminished Aδ behavioral responses in toes after RTX-treatment, a suppression of C-fiber behavioral responses might occur in tandem. During a separate study, we found that within 1 week after 50 ng intraplantar RTX, rats reached the 16 sec cutoff when testing C-fibers in their toes. 2-way ANOVA with repeated measures was used to compare the effect of 5 ng or 50 ng RTX versus vehicle. 2-way ANOVA was used to compare the effects of 5 ng versus 50 ng. *,† p < 0.5, **,†† p < 0.01, ***,††† p < 0.001, n = 6 rats per group.
Figure 4
Figure 4
Intraplantar injection of RTX leads to a transient increase in gene markers of nociception but a sustained increase in markers of nerve regeneration. Gel image shows ganglionic expression levels of mRNA encoding MCP-1 and ATF3 24 h or 10 days after vehicle or RTX treatment, taken from left and right L4-L5 dorsal root ganglia (n = 4 rats, ipsilateral RTX expression is denoted by an asterisk). MCP-1 (C) and ATF3 (D) transcript levels were normalized to GPDH. Data were obtained from RT-PCR analysis (n = 4/group). All graphs are presented as mean ± SEM. *p < 0.05 and **p < 0.01, and ***p < 0.001 as determined by a one-way ANOVA followed by a Bonferroni correction. (E-H) Immunohistochemistry showing spinal cord c-Fos expression 6 h or 10 d post-RTX. E and F are epifluorescent images depicting an elevation in c-Fos protein 6 h after RTX or vehicle in the ipsilateral and contralateral dorsal horn, respectively. G and H are epifluorescent images representing c-Fos expression 10 d after RTX or vehicle in the ipsilateral and contralateral dorsal horn, respectively. (I) Cell counts demonstrate significantly more c-Fos immunopositive cells in spinal cord ipsilateral to RTX injection as compared to contralateral vehicle injections at 6 h but not at 24 h or 10 days. Data expressed as means ± SEM (n = 4). The fold-change is shown. *p < 0.05 as determined by Student's t-test. Arrows indicate the location of the medial dorsal horn. The most intense c-Fos positive neurons were counted in this area.

References

    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. 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. Belmonte C, Gallar J, Pozo MA, Rebollo I. Excitation by irritant chemical substances of sensory afferent units in the cat's cornea. J Physiol. 1991;437:709–725.
    1. Kaufman MP, Iwamoto GA, Longhurst JC, Mitchell JH. Effects of capsaicin and bradykinin on afferent fibers with ending in skeletal muscle. Circ Res. 1982;50:133–139.
    1. Yeomans DC, Proudfit HK. Nociceptive responses to high and low rates of noxious cutaneous heating are mediated by different nociceptors in the rat: electrophysiological evidence. Pain. 1996;68:141–150. doi: 10.1016/S0304-3959(96)03177-6.
    1. Voight EA, Kort ME. Transient receptor potential vanilloid-1 antagonists: a survey of recent patent literature. Expert Opin Ther Pat. 2010;20:1107–1122. doi: 10.1517/13543776.2010.497756.
    1. Wong GY, Gavva NR. Therapeutic potential of vanilloid receptor TRPV1 agonists and antagonists as analgesics: Recent advances and setbacks. Brain Res Rev. 2009;60:267–277. doi: 10.1016/j.brainresrev.2008.12.006.
    1. Szallasi A, Cortright DN, Blum CA, Eid SR. The vanilloid receptor TRPV1: 10 years from channel cloning to antagonist proof-of-concept. Nat Rev Drug Discov. 2007;6:357–372. doi: 10.1038/nrd2280.
    1. Yaksh TL, Farb DH, Leeman SE, Jessell TM. Intrathecal capsaicin depletes substance P in the rat spinal cord and produces prolonged thermal analgesia. Science. 1979;206:481–483. doi: 10.1126/science.228392.
    1. Szallasi A, Blumberg PM. Vanilloid receptors: new insights enhance potential as a therapeutic target. Pain. 1996;68:195–208. doi: 10.1016/S0304-3959(96)03202-2.
    1. Karai LJ, Russell JT, Iadarola MJ, Olah Z. Vanilloid receptor 1 regulates multiple calcium compartments and contributes to Ca2+-induced Ca2+ release in sensory neurons. J Biol Chem. 2004;279:16377–16387. doi: 10.1074/jbc.M310891200.
    1. Winter J, Dray A, Wood JN, Yeats JC, Bevan S. Cellular mechanism of action of resiniferatoxin: a potent sensory neuron excitotoxin. Brain Res. 1990;520:131–140. doi: 10.1016/0006-8993(90)91698-G.
    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.
    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. Jeffry JA, Yu SQ, Sikand P, Parihar A, Evans MS, Premkumar LS. Selective targeting of TRPV1 expressing sensory nerve terminals in the spinal cord for long lasting analgesia. PLoS One. 2009;4:e7021. doi: 10.1371/journal.pone.0007021.
    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. Bates BD, Mitchell K, Keller JM, Chan CC, Swaim WD, Yaskovich R, Mannes AJ, Iadarola MJ. Prolonged analgesic response of cornea to topical resiniferatoxin, a potent TRPV1 agonist. Pain. 2010;149:522–528. doi: 10.1016/j.pain.2010.03.024.
    1. Szolcsanyi J, Szallasi A, Szallasi Z, Joo F, Blumberg PM. Resiniferatoxin. An ultrapotent neurotoxin of capsaicin-sensitive primary afferent neurons. Ann N Y Acad Sci. 1991;632:473–475. doi: 10.1111/j.1749-6632.1991.tb33161.x.
    1. Cavanaugh DJ, Lee H, Lo L, Shields SD, Zylka MJ, Basbaum AI, Anderson DJ. Distinct subsets of unmyelinated primary sensory fibers mediate behavioral responses to noxious thermal and mechanical stimuli. Proc Natl Acad Sci USA. 2009;106:9075–9080. doi: 10.1073/pnas.0901507106.
    1. Mishra SK, Hoon MA. Ablation of TrpV1 neurons reveals their selective role in thermal pain sensation. Mol Cell Neurosci. 2010;43:157–163. doi: 10.1016/j.mcn.2009.10.006.
    1. Kalliomaki J, Weng HR, Nilsson HJ, Schouenborg J. Nociceptive C fibre input to the primary somatosensory cortex (SI). A field potential study in the rat. Brain Res. 1993;622:262–270. doi: 10.1016/0006-8993(93)90827-A.
    1. Greffrath W, Nemenov MI, Schwarz S, Baumgartner U, Vogel H, Arendt-Nielsen L, Treede RD. Inward currents in primary nociceptive neurons of the rat and pain sensations in humans elicited by infrared diode laser pulses. Pain. 2002;99:145–155. doi: 10.1016/S0304-3959(02)00071-4.
    1. Tzabazis A, Klyukinov M, Manering N, Nemenov MI, Shafer SL, Yeomans DC. Differential activation of trigeminal C or Adelta nociceptors by infrared diode laser in rats: behavioral evidence. Brain Res. 2005;1037:148–156. doi: 10.1016/j.brainres.2005.01.019.
    1. Cuellar JM, Manering NA, Klukinov M, Nemenov MI, Yeomans DC. Thermal nociceptive properties of trigeminal afferent neurons in rats. Mol Pain. 2010;6:39. doi: 10.1186/1744-8069-6-39.
    1. Sun JJ, Yang JW, Shyu BC. Current source density analysis of laser heat-evoked intra-cortical field potentials in the primary somatosensory cortex of rats. Neuroscience. 2006;140:1321–1336. doi: 10.1016/j.neuroscience.2006.03.018.
    1. Devor M, Carmon A, Frostig R. Primary afferent and spinal sensory neurons that respond to brief pulses of intense infrared laser radiation: a preliminary survey in rats. Exp Neurol. 1982;76:483–494. doi: 10.1016/0014-4886(82)90118-2.
    1. Fan RJ, Kung JC, Olausson BA, Shyu BC. Nocifensive behaviors components evoked by brief laser pulses are mediated by C fibers. Physiol Behav. 2009;98:108–117. doi: 10.1016/j.physbeh.2009.04.022.
    1. Lawson SN, Perry MJ, Prabhakar E, McCarthy PW. Primary sensory neurones: neurofilament, neuropeptides, and conduction velocity. Brain Res Bull. 1993;30:239–243. doi: 10.1016/0361-9230(93)90250-F.
    1. Harper AA, Lawson SN. Conduction velocity is related to morphological cell type in rat dorsal root ganglion neurones. J Physiol. 1985;359:31–46.
    1. Lawson JJ, McIlwrath SL, Woodbury CJ, Davis BM, Koerber HR. TRPV1 unlike TRPV2 is restricted to a subset of mechanically insensitive cutaneous nociceptors responding to heat. J Pain. 2008;9:298–308. doi: 10.1016/j.jpain.2007.12.001.
    1. Danneman PJ, Kiritsy-Roy JA, Morrow TJ, Casey KL. Central delay of the laser-activated rat tail-flick reflex. Pain. 1994;58:39–44. doi: 10.1016/0304-3959(94)90183-X.
    1. Bakels R, Kernell D. Matching between motoneurone and muscle unit properties in rat medial gastrocnemius. J Physiol. 1993;463:307–324.
    1. Gosselin RD, Dansereau MA, Pohl M, Kitabgi P, Beaudet N, Sarret P, Melik Parsadaniantz S. Chemokine network in the nervous system: a new target for pain relief. Curr Med Chem. 2008;15:2866–2875. doi: 10.2174/092986708786242822.
    1. Jung H, Toth PT, White FA, Miller RJ. Monocyte chemoattractant protein-1 functions as a neuromodulator in dorsal root ganglia neurons. J Neurochem. 2008;104:254–263.
    1. Yang HY, Mitchell K, Keller JM, Iadarola MJ. Peripheral inflammation increases Scya2 expression in sensory ganglia and cytokine and endothelial related gene expression in inflamed tissue. J Neurochem. 2007;103:1628–1643. doi: 10.1111/j.1471-4159.2007.04874.x.
    1. Tsujino H, Kondo E, Fukuoka T, Dai Y, Tokunaga A, Miki K, Yonenobu K, Ochi T, Noguchi K. Activating transcription factor 3 (ATF3) induction by axotomy in sensory and motoneurons: A novel neuronal marker of nerve injury. Mol Cell Neurosci. 2000;15:170–182. doi: 10.1006/mcne.1999.0814.
    1. Noguchi K, Kowalski K, Traub R, Solodkin A, Iadarola MJ, Ruda MA. Dynorphin expression and Fos-like immunoreactivity following inflammation induced hyperalgesia are colocalized in spinal cord neurons. Brain Res Mol Brain Res. 1991;10:227–233. doi: 10.1016/0169-328X(91)90065-6.
    1. Draisci G, Iadarola MJ. Temporal analysis of increases in c-fos, preprodynorphin and preproenkephalin mRNAs in rat spinal cord. Brain Res Mol Brain Res. 1989;6:31–37. doi: 10.1016/0169-328X(89)90025-9.
    1. Qiao ZM, Wang JY, Han JS, Luo F. Dynamic processing of nociception in cortical network in conscious rats: a laser-evoked field potential study. Cell Mol Neurobiol. 2008;28:671–687. doi: 10.1007/s10571-007-9216-3.
    1. Isseroff RG, Sarne Y, Carmon A, Isseroff A. Cortical potentials evoked by innocuous tactile and noxious thermal stimulation in the rat: differences in localization and latency. Behav Neural Biol. 1982;35:294–307. doi: 10.1016/S0163-1047(82)90725-7.
    1. Price DD, Hu JW, Dubner R, Gracely RH. Peripheral suppression of first pain and central summation of second pain evoked by noxious heat pulses. Pain. 1977;3:57–68. doi: 10.1016/0304-3959(77)90035-5.
    1. Plaghki L, Decruynaere C, Van Dooren P, Le Bars D. The fine tuning of pain thresholds: a sophisticated double alarm system. PLoS One. 2010;5:e10269. doi: 10.1371/journal.pone.0010269.
    1. McIlroy WE, Bent LR, Potvin JR, Brooke JD, Maki BE. Preparatory balance adjustments precede withdrawal response to noxious stimulation in standing humans. Neurosci Lett. 1999;267:197–200. doi: 10.1016/S0304-3940(99)00365-1.
    1. Bent LR, Potvin JR, Brooke JD, McIlroy WE. Medio-lateral balance adjustments preceding reflexive limb withdrawal are modified by postural demands. Brain Res. 2001;914:100–105. doi: 10.1016/S0006-8993(01)02782-2.
    1. Beydoun A, Dyke DB, Morrow TJ, Casey KL. Topical capsaicin selectively attenuates heat pain and A delta fiber-mediated laser-evoked potentials. Pain. 1996;65:189–196. doi: 10.1016/0304-3959(95)00161-1.
    1. Rage M, Van Acker N, Facer P, Shenoy R, Knaapen MW, Timmers M, Streffer J, Anand P, Meert T, Plaghki L. The time course of CO2 laser-evoked responses and of skin nerve fibre markers after topical capsaicin in human volunteers. Clin Neurophysiol. 2010;121:1256–1266. doi: 10.1016/j.clinph.2010.02.159.
    1. Bromm B, Neitzel H, Tecklenburg A, Treede RD. Evoked cerebral potential correlates of C-fibre activity in man. Neurosci Lett. 1983;43:109–114. doi: 10.1016/0304-3940(83)90137-4.
    1. Veldhuijzen DS, Nemenov MI, Keaser M, Zhuo J, Gullapalli RP, Greenspan JD. Differential brain activation associated with laser-evoked burning and pricking pain: An event-related fMRI study. Pain. 2009;141:104–113. doi: 10.1016/j.pain.2008.10.027.
    1. White FA, Sun J, Waters SM, Ma C, Ren D, Ripsch M, Steflik J, Cortright DN, Lamotte RH, Miller RJ. Excitatory monocyte chemoattractant protein-1 signaling is up-regulated in sensory neurons after chronic compression of the dorsal root ganglion. Proc Natl Acad Sci USA. 2005;102:14092–14097. doi: 10.1073/pnas.0503496102.
    1. Bhangoo S, Ren D, Miller RJ, Henry KJ, Lineswala J, Hamdouchi C, Li B, Monahan PE, Chan DM, Ripsch MS, White FA. Delayed functional expression of neuronal chemokine receptors following focal nerve demyelination in the rat: a mechanism for the development of chronic sensitization of peripheral nociceptors. Mol Pain. 2007;3:38. doi: 10.1186/1744-8069-3-38.
    1. Jeon SM, Lee KM, Cho HJ. Expression of monocyte chemoattractant protein-1 in rat dorsal root ganglia and spinal cord in experimental models of neuropathic pain. Brain Res. 2009;1251:103–111. doi: 10.1016/j.brainres.2008.11.046.
    1. Seijffers R, Allchorne AJ, Woolf CJ. The transcription factor ATF-3 promotes neurite outgrowth. Mol Cell Neurosci. 2006;32:143–154. doi: 10.1016/j.mcn.2006.03.005.
    1. Iadarola MJ, Brady LS, Draisci G, Dubner R. Enhancement of dynorphin gene expression in spinal cord following experimental inflammation: stimulus specificity, behavioral parameters and opioid receptor binding. Pain. 1988;35:313–326. doi: 10.1016/0304-3959(88)90141-8.
    1. Fan RJ, Shyu BC, Hsiao S. Analysis of nocifensive behavior induced in rats by CO2 laser pulse stimulation. Physiol Behav. 1995;57:1131–1137. doi: 10.1016/0031-9384(94)00372-C.
    1. Young ST, Porrino LJ, Iadarola MJ. Cocaine induces striatal c-fos-immunoreactive proteins via dopaminergic D1 receptors. Proc Natl Acad Sci USA. 1991;88:1291–1295. doi: 10.1073/pnas.88.4.1291.
    1. Mitchell K, Iadarola MJ. RT-PCR analysis of pain genes: use of gel-based RT-PCR for studying induced and tissue-enriched gene expression. Methods Mol Biol. 2010;617:279–295. full_text.

Source: PubMed

3
订阅