The emergence of adolescent onset pain hypersensitivity following neonatal nerve injury

David Vega-Avelaira, Rebecca McKelvey, Gareth Hathway, Maria Fitzgerald, David Vega-Avelaira, Rebecca McKelvey, Gareth Hathway, Maria Fitzgerald

Abstract

Background: Peripheral nerve injuries can trigger neuropathic pain in adults but cause little or no pain when they are sustained in infancy or early childhood. This is confirmed in rodent models where neonatal nerve injury causes no pain behaviour. However, delayed pain can arise in man some considerable time after nerve damage and to examine this following early life nerve injury we have carried out a longer term follow up of rat pain behaviour into adolescence and adulthood.

Results: Spared nerve injury (SNI) or sham surgery was performed on 10 day old (P10) rat pups and mechanical nociceptive reflex thresholds were analysed 3, 7, 14, 21, 28, 38 and 44 days post surgery. While mechanical thresholds on the ipsilateral side are not significantly different from controls for the first 2-3 weeks post P10 surgery, after that time period, beginning at 21 days post surgery (P31), the SNI group developed following early life nerve injury significant hypersensitivity compared to the other groups. Ipsilateral mechanical nociceptive threshold was 2-fold below that of the contralateral and sham thresholds at 21 days post surgery (SNI-ipsilateral 28 (± 5) g control groups 69 (± 9) g, p < 0.001, 3-way ANOVA, n = 6 per group). Importantly, no effect was observed on thermal thresholds. This hypersensitivity was accompanied by macrophage, microglial and astrocyte activation in the DRG and dorsal horn, but no significant change in dorsal horn p38 or JNK expression. Preemptive minocycline (daily 40 mg/kg, s.c) did not prevent the effect. Ketamine (20 mg/kg, s.c), on the other hand, produced a dose-dependent reversal of mechanical nociceptive thresholds ipsilateral to the nerve injury such that thresholds return to control levels at the highest doses of 20 mg/Kg.

Conclusions: We report a novel consequence of early life nerve injury whereby mechanical hypersensitivity only emerges later in life. This delayed adolescent onset in mechanical pain thresholds is accompanied by neuroimmune activation and NMDA dependent central sensitization of spinal nociceptive circuits. This delayed onset in mechanical pain sensitivity may provide clues to understand the long term effects of early injury such as late onset phantom pain and the emergence of complex adolescent chronic pain syndromes.

Figures

Figure 1
Figure 1
Nerve injury at the age of 10 days has no effect over the first 2–3 weeks but gradually ipsilateral mechanical hypersensitivity emerges in adolescence (3-way ANOVA, p B) Thermal pain thresholds show a general developmental reduction in latency in all experimental groups with no significant differences in the SNI group when compared to sham group.
Figure 2
Figure 2
Shows sections through the L4 dorsal horn at various times after sham (top) and SNI (bottom) surgery at P10 (left and middle) and adult (right). Sections are immunostained with the microglia marker IBA-1 (red) and the non peptidergic C-fibre marker isolectin IB-4 (green). SNI surgery at P10 produces non significant microglia activation after 7 days compared to sham in (P10) rats. However, there is a late onset of microglia activation at 21 days post surgery which is significantly greater than sham and similar to that seen in adult rats 7 days post surgery. The gap in isolectin IB-4 is a marker of the terminal region of the nerves damaged by SNI surgery. B) Quantification of microglia activation in the three experimental groups illustrated in A. IBA-1 stained microglial cells were counted and the results are expressed as fold difference of the contralateral side. *: (n = 3 animals per experimental group, 3-way ANOVA, p < 0.001, Tukey, p < 0.05).
Figure 3
Figure 3
Shows sections through the L4 dorsal horn at various times after sham (top) and SNI (bottom) surgery at P10 (left and middle) and adult (right). Sections are immunostained with the astrocytic marker GFAP (red) and the non peptidergic C-fibre marker isolectin IB-4 (green). SNI surgery at P10 produces non significant astrocyte activation after 7 days compared to sham in (P10) rats. However, there is a late onset of astrocyte activation at 21 days post surgery which is significantly greater than sham and similar to that seen in adult rats 7 days post surgery. The gap in isolectin IB-4 is a marker of the terminal region of the nerves damaged by SNI surgery. B) Quantification of astrocyte activation in the three experimental groups illustrated in A. GFAP stained astrocytes with signs of gliosis were counted and the results are expressed as fold difference of the contralateral side. *: (n = 3 animals per experimental group, 3-way ANOVA, p < 0.001, Tukey, p < 0.05).
Figure 4
Figure 4
Shows sections through the L4 dorsal root ganglion at various times after sham (top) and SNI (bottom) surgery at P10 (left and middle) and adult (right). Sections are immunostained with the macrophage marker IBA-1 (red) and the large A cell neuronal population marker NF200 (green). Macrophage activation is observed as macrophages clustering around large neuronal cell bodies in characteristic ‘ring-like’ structures with processes extending from their enlarged cell bodies towards large neurons. SNI surgery at P10 produces non significant macrophage activation after 7 days compared to sham in (P10) rats. However, there is a late onset of macrophage activation at 21 days post surgery which is significantly greater than sham and similar to that seen in adult rats 7 days post surgery. B) Quantification of these activated macrophage ‘ring-like’ structures in the three experimental groups illustrated in A and expressed as a percentage accordingly to large sensory neurons*: (n = 3 animals per experimental group, 3-way ANOVA, p < 0.001, Tukey, p < 0.05).
Figure 5
Figure 5
Mechanical pain thresholds in animals with early-life (P10) nerve injury (S) or sham surgery H) tested in later life (P31). Mechanical pain thresholds were measured in minocycline (M) (40 mg/Kg daily ip injections) after SNI nerve injury compared to SNI saline (S) group and controls (H). n = 6 per experimental group; *: ANOVA, p B) Representative images of western-blot for phospho-p38 and phospho-JNK (pJNK) and the housekeeping gene Gapdh. Quantification was done by densitometry of p38 (C) and pJNK(D). *: n = 4 per experimental group; *: ANOVA, p < 0.001, Tukey, p < 0.05.
Figure 6
Figure 6
Mechanical pain thresholds in animals with early-life (P10) nerve injury (SNI-ipsi) or sham surgery (sham-ipsi) tested in later life (P31), before and after i.p ketamine. SNI surgery animals have lower baseline mechanical pain thresholds but the NMDA antagonist ketamine increases mechanical pain thresholds to normal levels in a dose dependant manner. The clearance of ketamine (72 h post injection) results in the mechanical pain thresholds falling back to pre-ketamine levels. B) The same dose dependent reversal of mechanical hypersensitivity by ketamine is observed when measuring foot paw withdrawal to a von Frey hair. *: (n = 6 per experimental group, 3-way ANOVA, p < 0.001, Tukey, p < 0.05), SNI: spared nerve injury, Ipsi: ipsilateral, Contra: contralateral, ip: intraperitoneal, FPW: frequency of paw withdrawal. Ketamine had no effect upon contralateral (SNI-contra) thresholds or upon sham operated animals.

References

    1. Anand P, Birch R. Restoration of sensory function and lack of long-term chronic pain syndromes after brachial plexus injury in human neonates. Brain. 2002;125:113–122. doi: 10.1093/brain/awf017.
    1. Walco GA, Dworkin RH, Krane EJ, LeBel AA, Treede RD. Neuropathic pain in children: Special considerations. Mayo Clin Proc. 2010;85:S33–S41. doi: 10.4065/mcp.2009.0647.
    1. Ririe D, Eisenach J. Age-dependent responses to nerve injury-induced mechanical allodynia. Anesthesiology. 2006;104:344–350. doi: 10.1097/00000542-200602000-00021.
    1. Lee DH, Chung JM. Neuropathic pain in neonatal rats. Neurosci Lett. 1996;209:140–142. doi: 10.1016/0304-3940(96)12623-9.
    1. Howard R, Walker S, Mota P, Fitzgerald M. The ontogeny of neuropathic pain: postnatal onset of mechanical allodynia in rat spared nerve injury (SNI) and chronic constriction injury (CCI) models. Pain. 2005;115:382–389. doi: 10.1016/j.pain.2005.03.016.
    1. Hermann C, Hohmeister J, Demirakca S, Zohsel K, Flor H. Long-term alteration of pain sensitivity in school-aged children with early pain experiences. Pain. 2006;125:278–285. doi: 10.1016/j.pain.2006.08.026.
    1. Walker SM, Franck LS, Fitzgerald M, Myles J, Stocks J, Marlow N. Long-term impact of neonatal intensive care and surgery on somatosensory perception in children born extremely preterm. Pain. 2009;141:79–87. doi: 10.1016/j.pain.2008.10.012.
    1. Grunau RE, Holsti L, Peters JW. Long-term consequences of pain in human neonates. Semin Fetal Neonatal Med. 2006;11:268–275. doi: 10.1016/j.siny.2006.02.007.
    1. Melzack R, Israel R, Lacroix R, Schultz G. Phantom limbs in people with congenital limb deficiency or amputation in early childhood. Brain. 1997;120(Pt 9):1603–1620.
    1. Sethna NF, Meier PM, Zurakowski D, Berde CB. Cutaneous sensory abnormalities in children and adolescents with complex regional pain syndromes. Pain. 2007;131:153–161. doi: 10.1016/j.pain.2006.12.028.
    1. Decosterd I, Woolf C. Spared nerve injury: an animal model of persistent peripheral neuropathic pain. Pain. 2000;87:149–158. doi: 10.1016/S0304-3959(00)00276-1.
    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. Raghavendra V, Tanga F, DeLeo JA. Inhibition of microglial activation attenuates the development but not existing hypersensitivity in a rat model of neuropathy. J Pharmacol Exp Ther. 2003;306:624–630. doi: 10.1124/jpet.103.052407.
    1. Suzuki R, Matthews EA, Dickenson AH. Comparison of the effects of MK-801, ketamine and memantine on responses of spinal dorsal horn neurones in a rat model of mononeuropathy. Pain. 2001;91:101–109. doi: 10.1016/S0304-3959(00)00423-1.
    1. Plenderleith MB, Wright LL, Snow PJ. Expression of lectin binding in the superficial dorsal horn of the rat spinal cord during pre- and postnatal development. Brain Res Dev Brain Res. 1992;68:103–109.
    1. Narita M, Yoshida T, Nakajima M, Miyatake M, Takagi T, Yajima Y, Suzuki T. Direct evidence for spinal cord microglia in the development of a neuropathic pain-like state in mice. J Neurochem. 2006;97:1337–1348. doi: 10.1111/j.1471-4159.2006.03808.x.
    1. Eng LF, Ghirnikar RS. GFAP and astrogliosis. Brain Pathol. 1994;4:229–237. doi: 10.1111/j.1750-3639.1994.tb00838.x.
    1. Molander C, Wang H, Rivero-Melian C, Grant G. Early decline and late restoration of spinal cord binding and transganglionic transport of isolectin B4 from Griffonia simplicifolia I after peripheral nerve transection and crush. Restor Neurol Neuros. 1996;10:123–133.
    1. Imai Y, Ibata I, Ito D, Ohsawa K, Kohsaka S. A novel gene iba1 in the major histocompatibility complex class III region encoding an EF hand protein expressed in a monocytic lineage. Biochem Biophys Res Commun. 1996;224:855–862. doi: 10.1006/bbrc.1996.1112.
    1. Dahl D, Labkovsky B, Bignami A. Neurofilament phosphorylation in axons and perikarya: immunofluorescence study of the rat spinal cord and dorsal root ganglia with monoclonal antibodies. J Comp Neurol. 1988;271:445–450. doi: 10.1002/cne.902710311.
    1. Vega-Avelaira D, Geranton SM, Fitzgerald M. Differential regulation of immune responses and macrophage/neuron interactions in the dorsal root ganglion in young and adult rats following nerve injury. Mol Pain. 2009;5:70. doi: 10.1186/1744-8069-5-70.
    1. Falcon M, Guendellman D, Stolberg A, Frenk H, Urca G. Development of thermal nociception in rats. Pain. 1996;67:203–208. doi: 10.1016/0304-3959(96)03070-9.
    1. Huang HW, Wang WC, Lin CC. Influence of age on thermal thresholds, thermal pain thresholds, and reaction time. J Clin Neurosci. 2010;17:722–726. doi: 10.1016/j.jocn.2009.10.003.
    1. De Leo J, Tawfik V, LaCroix-Fralish M. The tetrapartite synapse: path to CNS sensitization and chronic pain. Pain. 2006;122:17–21. doi: 10.1016/j.pain.2006.02.034.
    1. DeLeo J, Sorkin L, Watkins L. Immune and glial regulation of pain. 2007. pp. 67–106.
    1. DeLeo J, Yezierski R. The role of neuroinflammation and neuroimmune activation in persistent pain. Pain. 2001;90:1–6. doi: 10.1016/S0304-3959(00)00490-5.
    1. Hu P, McLachlan E. Macrophage and lymphocyte invasion of dorsal root ganglia after peripheral nerve lesions in the rat. Neuroscience. 2002;112:23–38. doi: 10.1016/S0306-4522(02)00065-9.
    1. Ji R, Suter M. p38 MAPK, microglial signaling, and neuropathic pain. Mol Pain. 2007;3:33. doi: 10.1186/1744-8069-3-33.
    1. Scholz J, Woolf C. The neuropathic pain triad: neurons, immune cells and glia. Nat Neurosci. 2007;10:1361–1368. doi: 10.1038/nn1992.
    1. Moss A, Beggs S, Vega-Avelaira D, Costigan M, Hathway GJ, Salter MW, Fitzgerald M. Spinal microglia and neuropathic pain in young rats. Pain. 2007;128:215–224. doi: 10.1016/j.pain.2006.09.018.
    1. Vega-Avelaira D, Moss A, Fitzgerald M. Age-related changes in the spinal cord microglial and astrocytic response profile to nerve injury. Brain Behav Immun. 2007;21:617–623. doi: 10.1016/j.bbi.2006.10.007.
    1. Bailey AL, Ribeiro-da-Silva A. Transient loss of terminals from non-peptidergic nociceptive fibers in the substantia gelatinosa of spinal cord following chronic constriction injury of the sciatic nerve. Neuroscience. 2006;138:675–690. doi: 10.1016/j.neuroscience.2005.11.051.
    1. Vulchanova L, Olson TH, Stone LS, Riedl MS, Elde R, Honda CN. Cytotoxic targeting of isolectin IB4-binding sensory neurons. Neuroscience. 2001;108:143–155. doi: 10.1016/S0306-4522(01)00377-3.
    1. Beggs S, Salter MW. Stereological and somatotopic analysis of the spinal microglial response to peripheral nerve injury. Brain Behav Immun. 2007;21:624–633. doi: 10.1016/j.bbi.2006.10.017.
    1. Chang YW, Waxman SG. Minocycline attenuates mechanical allodynia and central sensitization following peripheral second-degree burn injury. J Pain. 2010;11:1146–1154.
    1. Piao ZG, Cho IH, Park CK, Hong JP, Choi SY, Lee SJ, Lee S, Park K, Kim JS, Oh SB. Activation of glia and microglial p38 MAPK in medullary dorsal horn contributes to tactile hypersensitivity following trigeminal sensory nerve injury. Pain. 2006;121:219–231. doi: 10.1016/j.pain.2005.12.023.
    1. Gao YJ, Zhang L, Samad OA, Suter MR, Yasuhiko K, Xu ZZ, Park JY, Lind AL, Ma Q, Ji RR. JNK-induced MCP-1 production in spinal cord astrocytes contributes to central sensitization and neuropathic pain. J Neurosci. 2009;29:4096–4108. doi: 10.1523/JNEUROSCI.3623-08.2009.
    1. Ji RR, Kawasaki Y, Zhuang ZY, Wen YR, Decosterd I. Possible role of spinal astrocytes in maintaining chronic pain sensitization: review of current evidence with focus on bFGF/JNK pathway. Neuron Glia Biol. 2006;2:259–269. doi: 10.1017/S1740925X07000403.
    1. Gao X, Kim HK, Chung JM, Chung K. Enhancement of NMDA receptor phosphorylation of the spinal dorsal horn and nucleus gracilis neurons in neuropathic rats. Pain. 2005;116:62–72. doi: 10.1016/j.pain.2005.03.045.
    1. Bourquin AF, Suveges M, Pertin M, Gilliard N, Sardy S, Davison AC, Spahn DR, Decosterd I. Assessment and analysis of mechanical allodynia-like behavior induced by spared nerve injury (SNI) in the mouse. Pain. 2006;122(14):e11–e14.
    1. Fitzgerald M. The development of nociceptive circuits. Nat Rev Neurosci. 2005;6:507–520.
    1. Baccei ML. Development of pain: maturation of spinal inhibitory networks. Int Anesthesiol Clin. 2007;45:1–11.
    1. McCutcheon JE, Marinelli M. Age matters. Eur J Neurosci. 2009;29:997–1014. doi: 10.1111/j.1460-9568.2009.06648.x.
    1. Atherton DD, Taherzadeh O, Elliot D, Anand P. Age-dependent development of chronic neuropathic pain, allodynia and sensory recovery after upper limb nerve injury in children. J Hand Surg Eur Vol. 2008;33:186–191. doi: 10.1177/1753193408087029.
    1. McCann ME, Waters P, Goumnerova LC, Berde C. Self-mutilation in young children following brachial plexus birth injury. Pain. 2004;110:123–129. doi: 10.1016/j.pain.2004.03.020.
    1. Saadah ES, Melzack R. Phantom limb experiences in congenital limb-deficient adults. Cortex. 1994;30:479–485.
    1. Clinch J, Eccleston C. Chronic musculoskeletal pain in children: assessment and management. Rheumatology (Oxford) 2009;48:466–474.
    1. Costigan M, Moss A, Latremoliere A, Johnston C, Verma-Gandhu M, Herbert TA, Barrett L, Brenner GJ, Vardeh D, Woolf CJ, Fitzgerald M. T-cell infiltration and signaling in the adult dorsal spinal cord is a major contributor to neuropathic pain-like hypersensitivity. J Neurosci. 2009;29:14415–14422. doi: 10.1523/JNEUROSCI.4569-09.2009.
    1. Fadel S, Sarzotti M. Cellular immune responses in neonates. Int Rev Immunol. 2000;19:173–193. doi: 10.3109/08830180009088504.
    1. Adkins B. Development of neonatal Th1/Th2 function. Int Rev Immunol. 2000;19:157–171. doi: 10.3109/08830180009088503.
    1. Zhuang ZY, Gerner P, Woolf CJ, Ji RR. ERK is sequentially activated in neurons, microglia, and astrocytes by spinal nerve ligation and contributes to mechanical allodynia in this neuropathic pain model. Pain. 2005;114:149–159. doi: 10.1016/j.pain.2004.12.022.
    1. Zhang J, De Koninck Y. Spatial and temporal relationship between monocyte chemoattractant protein-1 expression and spinal glial activation following peripheral nerve injury. J Neurochem. 2006;97:772–783. doi: 10.1111/j.1471-4159.2006.03746.x.
    1. Colton CA. Heterogeneity of microglial activation in the innate immune response in the brain. J Neuroimmune Pharmacol. 2009;4:399–418. doi: 10.1007/s11481-009-9164-4.
    1. Nimmerjahn A, Kirchhoff F, Helmchen F. Resting microglial cells are highly dynamic surveillants of brain parenchyma in vivo. Science. 2005;308:1314–1318. doi: 10.1126/science.1110647.
    1. Ji R, Strichartz G. Cell signaling and the genesis of neuropathic pain. Sci STKE. 2004;2004:reE14.
    1. Wieseler-Frank J, Maier SF, Watkins LR. Central proinflammatory cytokines and pain enhancement. Neurosignals. 2005;14:166–174. doi: 10.1159/000087655.
    1. Ransohoff RM, Cardona AE. The myeloid cells of the central nervous system parenchyma. Nature. 2010;468:253–262. doi: 10.1038/nature09615.
    1. Woolf CJ. Evidence for a central component of post-injury pain hypersensitivity. Nature. 1983;306:686–688. doi: 10.1038/306686a0.
    1. Latremoliere A, Woolf CJ. Central sensitization: a generator of pain hypersensitivity by central neural plasticity. J Pain. 2009;10:895–926. doi: 10.1016/j.jpain.2009.06.012.
    1. Ma QP, Woolf CJ. Noxious stimuli induce an N-methyl-D-aspartate receptor-dependent hypersensitivity of the flexion withdrawal reflex to touch: implications for the treatment of mechanical allodynia. Pain. 1995;61:383–390. doi: 10.1016/0304-3959(94)00195-K.
    1. Qian J, Brown SD, Carlton SM. Systemic ketamine attenuates nociceptive behaviors in a rat model of peripheral neuropathy. Brain Res. 1996;715:51–62. doi: 10.1016/0006-8993(95)01452-7.
    1. Li J, Walker SM, Fitzgerald M, Baccei ML. Activity-dependent modulation of glutamatergic signaling in the developing rat dorsal horn by early tissue injury. J Neurophysiol. 2009;102:2208–2219. doi: 10.1152/jn.00520.2009.
    1. Hathway GJ, Koch S, Low L, Fitzgerald M. The changing balance of brainstem-spinal cord modulation of pain processing over the first weeks of rat postnatal life. J Physiol. 2009;587:2927–2935. doi: 10.1113/jphysiol.2008.168013.
    1. Beggs S, Currie G, Salter MW, Fitzgerald M. Walker SM: Priming of adult pain responses by neonatal pain experience: maintenance by central neuroimmune activity. Brain. 2011;135(Pt2):404–417.

Source: PubMed

3
Se inscrever