Mechanical sensitization of cutaneous sensory fibers in the spared nerve injury mouse model

Similar documents
A role for uninjured afferents in neuropathic pain

Enhanced formalin nociceptive responses following L5 nerve ligation in the rat reveals neuropathy-induced inflammatory hyperalgesia

Received 26 April 2004; revised 21 August 2004; accepted 20 September 2004 Available online 19 November 2004

Venipuncture-induced neuropathic pain: the clinical syndrome, with comparisons to experimental nerve injury models

EFFECTS OF Pueraria mirifica ON NEUROPATHIC PAIN IN RATS

NIH Public Access Author Manuscript J Neuropathic Pain Symptom Palliation. Author manuscript; available in PMC 2007 March 26.

The Conundrum Of Sensitization When Recording From Nociceptors

Centrally Mediated Anti-Hyperalgesic and Anti-Allodynic Effect of Tolperisone in Spared Nerve Injury Model of Neuropathic Pain

Degeneration of Myelinated Efferent Fibers Induces Spontaneous Activity in Uninjured C-Fiber Afferents

The Egyptian Journal of Hospital Medicine (January 2018) Vol. 70 (12), Page

Somatosensory modalities!

Peripheral nerve injury alters excitatory synaptic transmission in lamina II of the rat dorsal horn

CONTRACTING ORGANIZATION: Johns Hopkins University Baltimore MD

Update on the Neurophysiology of Pain Transmission and Modulation: Focus on the NMDA-Receptor

Increased Sensitivity of Injured and Adjacent Uninjured Rat Primary Sensory Neurons to Exogenous Tumor Necrosis Factor- after Spinal Nerve Ligation

Dissecting out mechanisms responsible for peripheral neuropathic pain: Implications for diagnosis and therapy

Diabetic Complications Consortium

Sympathetic fibre sprouting in the skin contributes to pain related behaviour in spared nerve injury and cuff models of neuropathic pain

Mechanisms of Neuropathic Pain

SUPPLEMENTARY INFORMATION

12 Anatomy and Physiology of Peripheral Nerves

Laiche Djouhri 1, Xin Fang 2, Stella Koutsikou, Sally N. Lawson PAIN Ò 153 (2012) article info. abstract. 1.

Pain and Touch. Academic Press. Edited by Lawrence Kruger. Department of Neurobiology University of California, Los Angeles Los Angeles, California

Mechanism of Pain Production

Sensory coding and somatosensory system

Animal Model of Trigeminal Neuralgia Induced by Chronic Constriction Injury Applied to the Ophthalmic Nerve in the Rat

Somatosensation. Recording somatosensory responses. Receptive field response to pressure

Materials and Methods

Special Issue on Pain and Itch

Potential for delta opioid receptor agonists as analgesics in chronic pain therapy

Neurotrophic in uences on neuropathic pain

Lateral view of human brain! Cortical processing of touch!

In Vivo Electrophysiological Recording Techniques for the Study of Neuropathic Pain in Rodent Models

Somatic Sensation (MCB160 Lecture by Mu-ming Poo, Friday March 9, 2007)

Published online October 10, 2016

How strong is it? What is it? Where is it? What must sensory systems encode? 9/8/2010. Spatial Coding: Receptive Fields and Tactile Discrimination

Spatial Coding: Receptive Fields and Tactile Discrimination

Biomechanics of Pain: Dynamics of the Neuromatrix

Male circumcision decreases

Chapter 13 PNS and reflex activity

Coding of Sensory Information

PAIN MANAGEMENT in the CANINE PATIENT

AFFERENT IMPULSES FROM SINGLE MYELINATED FIBERS IN SPLANCHNIC NERVES, ELICITED BY MECHANICAL STIMULATION OF TOAD'S VISCERA

Somatic Sensory System I. Background

Voltage-gated sodium channel expression in mouse DRG after SNI leads to re-evaluation of projections of injured fibers

Pathophysiology of Pain

CHAPTER 10 THE SOMATOSENSORY SYSTEM

Proceedings of the World Small Animal Veterinary Association Sydney, Australia 2007

Dorsal root ganglion compression as an animal model of sciatica and low back pain

Introduction to some interesting research questions: Molecular biology of the primary afferent nociceptor

Supporting Information

Applied physiology: neuropathic pain

SOMATOSENSORY SYSTEMS

Effects of decompression on neuropathic pain behaviors and skin reinnervation in chronic constriction injury

Capsaicin Responses in Heat-Sensitive and Heat-Insensitive A-Fiber Nociceptors


Our senses provide us with wonderful capabilities. If you had to lose one, which would it be?

Electrophysiological Assessment of the Cutaneous Arborization of A -Fiber Nociceptors

Pharmacology of Pain Transmission and Modulation

Repeated intra-articular injections of acidic saline produce long-lasting joint pain and. widespread hyperalgesia

CONTRIBUTION OF FEMORAL AND PROXIMAL SCIATIC NERVE BRANCHES TO THE SENSORY

Pathophysiological Mechanisms of Neuropathic Pain

Seizure: the clinical manifestation of an abnormal and excessive excitation and synchronization of a population of cortical

TMC9 as a novel mechanosensitive ion channel

Original Article Upregulation of neuregulin-1 reverses signs of neuropathic pain in rats

THE EFFECT OF VOLUNATARY EXERCISE ON NEUROPATHIC PAIN. Kevin L. Farmer

The Representation of Peripheral Nerve Inputs in the S-I Hindpaw Cortex of Rats Raised with Incompletely Innervated Hindpaws

Neuronal Circuits and Neuronal Pools

Skin types: hairy and glabrous (e.g. back vs. palm of hand)

Receptor Subtype Mediating the Adrenergic Sensitivity of Pain Behavior and Ectopic Discharges in Neuropathic Lewis Rats

Axotomy increases NADPH-diaphorase activity in the dorsal root ganglia and lumbar spinal cord of the turtle Trachemys dorbigni

The neuropeptide galanin is widely distributed within the

The Role of micrornas in Pain. Phd Work of: Rehman Qureshi Advisor: Ahmet Sacan

Anatomical Substrates of Somatic Sensation

Department of Neurology/Division of Anatomical Sciences

Spared nerve injury model to study orofacial pain

HUMAN MOTOR CONTROL. Emmanuel Guigon

NMDA-Receptor Antagonists and Opioid Receptor Interactions as Related to Analgesia and Tolerance

MYOFASCIAL PAIN. Dr. Janet Travell ( ) credited with bringing MTrPs to the attention of healthcare providers.

Alireza Bakhshaeekia and Sina Ghiasi-hafezi. 1. Introduction. 2. Patients and Methods

Response of Cutaneous A- and C-Fiber Nociceptors in the Monkey to Controlled-Force Stimuli

Supplementary Figure 1 Information on transgenic mouse models and their recording and optogenetic equipment. (a) 108 (b-c) (d) (e) (f) (g)

TREATMENT. Reviews. Clinical features of postherpetic neuralgia The severity of the pain in postherpetic

DR SHRENIK M SHAH SHREY HOSPITAL AHMEDABAD

Compound Action Potential, CAP

Index. Note: Page numbers of article titles are in boldface type.

Pain and endometriosis: How to optimize the medical management?

Spatiotemporal dynamics of re-innervation and hyperinnervation patterns by uninjured CGRP fibers in the rat foot sole epidermis after nerve injury

Accepted Manuscript. Increased expression of cutaneous α 1 -adrenoceptors after chronic constriction injury in rats

EVALUATION OF ANTI-NOCICEPTIVE EFFECT OF TOLPERISONE BY COMPARING ITS EFFECT WITH TRAMADOL IN NEUROPATHIC PAIN

Pre-op Interventions to Mitigate Post-op Acute and Chronic Pain

Lecture 14: The Spinal Cord

On regenerative and collateral sprouting to hind limb digits after sciatic nerve injury in the rat

MANAGEMENT OF CRPS. Brachial Neuralgia n = 81 patients. CRPS type II n = 126 patients CONTENTS. Neuropathic Pain. Introduction.

LONG-TERM postoperative pain follows many common

Cancer-induced bone pain

Neurologic complications - whom to blame? Benno Rehberg Médecin adjoint agrégé Unité d anesthésiologie gynéco-obstétricale, HUG

Bi/CNS/NB 150: Neuroscience. November 11, 2015 SOMATOSENSORY SYSTEM. Ralph Adolphs

Mechanical Allodynia Definition, Assessment and Treatment

Transcription:

Smith et al. Molecular Pain 2013, 9:61 MOLECULAR PAIN SHORT REPORT Open Access Mechanical sensitization of cutaneous sensory fibers in the spared nerve injury mouse model Amanda K Smith, Crystal L O Hara and Cheryl L Stucky * Abstract Background: The spared nerve injury (SNI) model of neuropathic pain produces robust and reproducible behavioral mechanical hypersensitivity. Although this rodent model of neuropathic pain has been well established and widely used, peripheral mechanisms underlying this phenotype remain incompletely understood. Here we investigated the role of cutaneous sensory fibers in the maintenance of mechanical hyperalgesia in mice post-sni. Findings: SNI produced robust, long-lasting behavioral mechanical hypersensitivity compared to sham and naïve controls beginning by post-operative day (POD) 1 and continuing through at least POD 180. We performed teased fiber recordings on single cutaneous fibers from the spared sural nerve using ex vivo skin-nerve preparations. Recordings were made between POD 16 42 after SNI or sham surgery. Aδ-mechanoreceptors (AM) and C fibers, many of which are nociceptors, from SNI mice fired significantly more action potentials in response to suprathreshold mechanical stimulation than did fibers from either sham or naïve control mice. However, there was no increase in spontaneous activity. Conclusions: To our knowledge, this is the first study evaluating the contribution of primary afferent fibers in the SNI model. These data suggest that enhanced suprathreshold firing in AM and C fibers may play a role in the marked, persistent mechanical hypersensitivity observed in this model. These results may provide insight into mechanisms underlying neuropathic pain in humans. Keywords: Neuropathic, Nociceptor, Sensory neuron, C fiber, A fiber, Hyperalgesia, Mechanotransduction Findings Background Peripheral neuropathic pain results from complete or partial lesion to peripheral nerves [1,2]. Occurring in many neurological disorders, neuropathic pain affects 6-8% of the population and is characterized by spontaneous and stimulus-evoked pain [2]. The mechanisms driving nerve injury-induced hyperalgesia are not well understood making treatments sub-optimal [1-4]. Several rodent models of neuropathic pain have been developed including, chronic constriction injury (CCI) [5], partial sciatic nerve injury [6], and spinal nerve ligation (SNL) [7]. These models often involve variability within cohorts [1,4] and present a challenge in determining the role of injured versus non-injured, intact sensory afferents in neuropathic pain because they involve a high degree of co-mingling of intact and injured axons distal to the lesion [8]. * Correspondence: cstucky@mcw.edu Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, USA We used the spared nerve injury (SNI) model of neuropathic pain which produces a pronounced, long-lasting and reproducible behavioral phenotype characterized by intense mechanical allodynia and hyperalgesia that mimics many features of clinical neuropathic pain [1,3,9]. SNI comprises complete transection of two of the three sciatic nerve branches (tibial and common peroneal), leaving the sural nerve intact [1]. Furthermore, SNI involves minimal co-mingling of intact and injured axons distal to the lesion [1], thereby allowing investigators to specifically target non-directly-injured nerve fibers. Although SNI is well established, the peripheral mechanisms contributing to the pain phenotype are not clear. While teased fiber recordings have been used to investigate peripheral sensitization in SNL in monkey and rat [10,11], and in CCI in rat [12,13], to our knowledge, these experiments have not been performed in the SNI model. Thus, the goal of this study was to determine whether intact cutaneous afferent fibers from the spared sural nerve are sensitized to mechanical stimuli 2013 Smith et al.; licensee BioMed Central Ltd. This is an open access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/2.0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

Smith et al. Molecular Pain 2013, 9:61 Page 2 of 6 and thereby, may contribute to the maintenance of mechanical hypersensitivity after SNI. SNI mice exhibit long-lasting behavioral mechanical hypersensitivity As previously reported, SNI mice exhibited pronounced hypersensitivity to mechanical stimuli compared to sham and naïve animals beginning by post-operative day (POD) 1 and continuing for at least 6 months post-surgery (Figure 1). The dynamic component of the Light Touch Behavioral Assay [14] was used as a control to ensure adequate denervation of the tibial territory post-sni injury. SNI mice showed significant tibial desensitization, measured by percent response to a puffed cotton swab applied to the tibial territory of the glabrous skin, from POD 1 42 (Figure 1A, p < 0.005), which was expected because of transection of the tibial nerve and subsequent denervation of the skin territory. However, by POD 49, sensation in the tibial area began to return (Figure 1A). In sural nervetargeted behavioral testing, SNI mice showed a significant decrease in paw withdrawal threshold by POD 1 through POD 180 (Figure 1B, p < 0.005) and exhibited a significantly higher percent response to the suprathreshold 3.31 mn monofilament from POD 1 49 (Figure 1C, p < 0.005). Locomotor activity of the mice did not differ between groups (data not shown). Complete transection of the tibial and common peroneal nerves was validated postmortem. Overall, these results parallel those found in rat [1] and previously shown in mouse [3,9]. Aδ and C fibers from SNI mice exhibit enhanced mechanical firing Sensory afferent sensitization is known to contribute to mechanical hypersensitivity observed in diabetic and chemotherapy-induced neuropathies [15,16], and has been shown to contribute to hypersensitivity observed in other models of nerve injury [10-12]. To assess the contribution of cutaneous sensory afferents to SNI-induced mechanical hypersensitivity, we performed ex vivo teased fiber recordings on the spared sural nerve. Fibers from SNI animals exhibited enhanced suprathreshold firing compared to controls. Specifically, Aδ-mechanoreceptor (AM) fibers fired an average of 22% more action potentials across all forces compared to sham or naïve mice, and C fibers exhibited 24% more action potentials across all forces compared to sham and 28% more than naïve mice (AM: Figure 2A, p < 0.01; C: Figure 2C, p < 0.05). Post hoc comparison showed no differences at individual forces for either AM or C fibers (Figure 2). There were no differences in mechanical thresholds or conduction velocity of any fiber subtype across treatment groups (Table 1). There was no difference in the percentage of Aβ, Aδ, andc fibers encountered in preparations from the different surgical groups (Figure 3A, p > 0.05). There was also no Figure 1 SNI mice exhibit prominent behavioral hypersensitivity. A) In response to dynamic stroke of a puffed cotton swab, SNI mice show significant desensitization of the tibial territory beginning POD 1 and continuing through POD 42 compared to sham or naïve animals (***p<0.005). At POD 49 SNI mice begin to regain sensation in the tibial territory, in that SNI mice still showed some sensitization compared to naïve animals (*p<0.05) but not sham animals (p>0.05). Treatments were compared across time using a repeated measure 2-way ANOVA with Tukey s post hoc comparisons. B) SNI mice show a significant decrease in the 50% mechanical withdrawal threshold beginning POD 1 and continuing through POD 49 compared to sham or naïve mice (***p<0.005). Furthermore, SNI mice continue to show a significant decrease in mechanical withdrawal threshold at POD 90 compared to sham (***p<0.005) and at POD 180 compared to sham (*p<0.05). Treatments were compared across time using a 2-way ANOVA with Bonferroni post hoc tests. SNI and sham treatments were compared at POD 90 and POD 180 using Mann Whitney U tests. C) In response to repeated stimulus with a 3.31 mn monofilament, SNI mice exhibit prominent hypersensitivity beginning POD 1 and continuing through at least POD 49 compared to sham or naïve animals (***p<0.005). Treatments were compared across time using a 2 way-anova with Bonferroni post hoc tests. Error bars for all three graphs indicate S.E.M.

Smith et al. Molecular Pain 2013, 9:61 Page 3 of 6 Figure 2 Aδ-Mechanoreceptor and C fibers in SNI mice exhibit enhanced mechanical firing. Using ex vivo skin nerve recordings, 10 sec mechanical stimuli ranging 5-200 mn were applied to the receptive field of each fiber using a 0.8 mm probe. All recordings were performed on the sural nerve and its innervating territory. A) Aδ-Mechanoreceptor (AM) fibers fired an average 22% more action potentials per second across all forces compared to sham or to naïve animals (**p<0.01). B) Examples of AM fiber action potentials evoked by sustained mechanical stimuli in naïve, sham and SNI treatment groups. C) C fibers fired an average 24% more action potentials per second across all forces compared to sham and 28% more compared to naïve animals (*p<0.05). D) Examples of C fiber action potentials evoked by sustained mechanical stimuli in naïve, sham and SNI treatment groups. Number of action potentials fired per second was compared across all forces and treatment groups using a 2-way ANOVA with Bonferroni post hoc comparisons. Error bars indicate S.E.M. difference in the proportion of slowly adapting (AM) and rapidly adapting (D-hair) Aδ fibers (Figure 3B, p > 0.05). There was an overall difference in the distribution of Aβ fibers among SNI, sham and naïve mice (Figure 3C, p < 0.05). There was a decrease in the distribution of slowly adapting (SA) A-beta fibers from SNI animals compared to sham (Figure 3C, p< 0.5), although no difference was observed between SNI and naive animals (Figure 3C, p> 0.5). We also measured spontaneous activity because spontaneous activity in primary afferent fibers accompanies other nerve injury animal models including SNL and CCI [10-13,17-19]. There was no difference in the percentage of AM or C fibers that exhibited spontaneous activity in SNI versus sham or naïve groups (Figure 4, p > 0.05). Furthermore, preliminary analysis of Aβ fibers from SNI preparations also does Table 1 Summary of fiber properties in Naive, Sham and SNI mice Fiber type Genotype n Median von frey threshold (mn) Lower quartile Upper quartile Mean conduction velocity (m/s) Naive 24 6.82 6.82 13.88 4.30 0.41 Aδ-Mechanoreceptor Sham 25 6.82 5.41 11.70 5.33 0.62 SNI 20 6.82 4.00 11.70 3.95 0.71 Naive 5 0.66 0.66 1.15 4.80 0.86 D-hair Sham 7 0.66 0.27 0.66 4.93 0.80 SNI 7 0.27 0.23 1.63 5.57 0.54 Naive 11 6.82 6.82 11.70 0.66 0.10 C Sham 17 11.70 5.41 14.60 0.68 0.06 SNI 13 6.82 4.00 11.70 0.65 0.08 Naive 7 1.63 0.66 4.00 11.60 1.05 RA-Aβ Sham 10 0.66 0.66 1.63 13.99 1.38 SNI 17 1.63 0.66 1.63 12.85 0.58 Naive 8 1.63 1.63 3.41 13.47 1.46 SA-Aβ Sham 13 1.63.63 2.81 14.49 1.33 SNI 11 1.63 0.66 4.00 14.76 1.15 ±SEM

Smith et al. Molecular Pain 2013, 9:61 Page 4 of 6 Figure 3 Distribution of cutaneous fiber types in SNI mice. Since SNI mice fired significantly more action potentials per second across all forces, we compared the percent distribution of each fiber type across treatment groups. A) There was not a significant difference in percent fiber distribution in SNI mice compared to sham or naïve animals (p>0.05). B) SNI mice have a similar distribution of AM and DH fibers as sham and naïve animals (p>0.05). C) There was an overall difference in the distribution of Aβ fibers among SNI, sham and naïve mice (*p<0.05). There was a decrease in the proportion of slowly adapting (SA) Abeta fibers in SNI mice compared to sham (*p<0.5), although no difference was observed between SNI and naive animals (p>0.5). Distribution data was analyzed using a Chi-square analysis and Fisher s exact post hoc tests. not suggest increased spontaneous firing, as has been observed in SNL or CCI (data not shown). There was also no difference in the frequency of spontaneous action potential firing in SNI versus controls for any fiber type (data not shown). In dividing groups into early and late stages post SNI, there was no significant difference in the spontaneous activity of fibers recorded at POD 16 21 compared to those at POD 37 42 for Aβ, Aδ, orcfibers (data not shown). To our knowledge, this is the first study to assess sensitization of primary afferent fibers in SNI. Our results suggest that enhanced suprathreshold firing in AM and C fibers may contribute to the robust behavioral mechanical hypersensitivity that occurs in the SNI model of neuropathic pain. Sensitized nociceptors might contribute to SNI-induced behavioral hypersensitivity either directly through increased suprathreshold firing in response to external stimuli, or indirectly by driving central sensitization [10,11]. Previous nerve injury studies that used thesnlandccimodelsofnerveinjurysuggestthat Wallerian Degeneration of injured nerves drives sensitization of adjacent intact afferent fibers [10-12]. However, unlike CCI and SNL, Wallerian Degeneration is not a major factor in the SNI model of neuropathic pain as SNI involves minimal co-mingling of intact and injured afferent fibers [1]. Therefore, a different mechanism(s) likely drives the afferent mechanical sensitization observed in this model. One potential mechanism is paracrine signaling between injured and intact cell bodies within the dorsal root ganglia (DRG), a level where co-mingling Figure 4 Spontaneous activity is similar in AM and C fibers from naïve, sham, and SNI mice. SNI mice did not differ from sham or naïve mice in that the percent of non-evoked spontaneous discharges was similar across treatment groups in either AM or C fibers (p>0.05). Data was analyzed using Chi-square analysis.

Smith et al. Molecular Pain 2013, 9:61 Page 5 of 6 occurs. A previous study has shown an increase in macrophage infiltration, expression of inflammatory mediators such as IL-6 and TNF-α, and expression of neurotrophins BDNF and NGF in the DRG after sciatic nerve injury [20], and these may be key factors driving afferent sensitization in the SNI model [1]. Alternatively or in addition, at the peripheral terminals, collateral sprouting of intact sensory afferent terminals into the denervated skin territory of the transected nerves has been shown in other neuropathic pain models [21,22], and may also occur and contribute to sensitization in SNI. It has been shown that degeneration of injured fibers induces spontaneous activity in nearby uninjured primary afferent fibers [23]. Furthermore, previous studies have shown that increased spontaneous activity in A and C fibers can contribute to sensitization after nerve injury in other models of neuropathic pain [10-13,17-19]. However, we did not observe more spontaneous activity in either A or C fibers post-sni. One explanation for the absence of spontaneous activity may be that ectopic discharge rates change over time after injury. Previous studies on injured primary afferent fibers show that there is a higher frequency of spontaneous activity early after nerve injury (POD 1 3) and less activity in late stages (POD 11 14) [17,18]. Furthermore, studies on uninjured afferents, which show spontaneous activity, have been performed primarily at early stages after nerve injury [13,23]. Thus, our recordings at later stages (POD 16 42) after injury may have occurred after SNI-induced spontaneous activity subsided. Another likely explanation is that spontaneous activity may not be present in the SNI model due to minimal co-mingling of injured and adjacent fibers. Previous reports of spontaneous activity after nerve injury have been recorded from nerve injury models that involve considerable Wallerian Degeneration and extensive co-mingling of intact and injured axons [10-12]. Sensitizing compounds associated with Wallerian Degeneration, such as TNF-α, have been shown to sensitize primary afferent fibers [24]. However, in the absence of co-mingling of intact and injured axons distal to the site of lesion, and the minimal degeneration of injured fibers proximal to the lesion, these compounds may not affect the intact peripheral afferent fibers in the SNI model. Conclusions These results may provide insight into the mechanisms underlying neuropathic pain in humans with traumatic peripheral nerve injury. Our results show an increase in suprathreshold firing in Aδ-mechanoreceptor (AM) and C fibers, suggesting that enhanced primary afferent drive may contribute to nerve injury-induced hypersensitivity, and peripheral afferent fibers may be targets for pharmacological treatment of neuropathic pain. Materials and methods Animals Male C57BL/6 mice (Jackson Labs), 8 weeks at time of injury, were used for all behavioral and teased fiber skin nerve experiments. Animals were housed individually after surgery and handled equally during all experiments. All experimental protocols were approved by the Medical College of Wisconsin s Institutional Animal Care and Use Committee. Surgery SNI surgery was performed as previously described [1]. Briefly, under ketamine-induced anesthesia, animals underwent surgery to ligate and transect the left tibial and common peroneal nerves, and 2-4 mm of nerve distal to the ligation was removed to prevent regeneration. Care was taken to avoid damage to the sural nerve. Sham animals underwent anesthesia and skin and muscle incisions identical to the SNI animals, without ligation or axotomy of the tibial and peroneal nerves. Naïve animals received no surgical treatment or anesthetic. Although we attempted to blind the experimenter to surgery type, it was possible to distinguish which animals had undergone SNI, sham or naïve treatment. Behavior Behavioral sensitivity to dynamic light touch was assessed on the tibial skin territory of the left paw as previously reported [14]. Mechanical threshold and sensitivity to suprathreshold mechanical force were assessed on the sural territory of the left hind paw as previously reported [14]. The animals were tested twice before surgery and on post-operative-days (POD) 1, 2, 4, 7, 14, 21, 28, 35, 42, 49, 90, and 180. Teased fiber skin-nerve recordings Teased fiber recordings were used to determine the mechanical response properties of cutaneous primary afferent fibers in skin nerve preparations from SNI, sham and naïve mice as previously described [25]. Briefly, the sural nerve and innervated skin of the left hindlimb were dissected and placed corium side up in a recording bath superfused with oxygenated synthetic interstitial fluid at 32 ± 0.5 C. The nerve was desheathed and fascicles were teased apart until single, functionally distinct fibers, could be distinguished. Fibers were characterized by mechanical threshold and conduction velocity. Units with conduction velocities over 10 m/s were classified as Aβ, and Aδ for units with conduction velocities between 1.2 m/s and 10 m/s. C fibers were classified as units that had conduction velocities less than 1.2 m/s. Units were further sub-classified as slowly adapting (SA) or rapidly adapting (RA) based on the rate of adaptation to mechanical force. Following characterization, fibers were recorded for 2 min

Smith et al. Molecular Pain 2013, 9:61 Page 6 of 6 to assess spontaneous activity. Next a feedback-controlled mechanical stimulator was used to deliver increasing sustained mechanical forces (5-200 mn) for 10 sec each with 1 min recovery period between stimuli. Action potentials were recorded and analyzed using Lab Chart Data Acquisition Software (AD Instruments, Colorado Springs, CO). Data analysis All data sets were compared between SNI, sham, and naïve groups. Behavioral Data: Percent response to light touch was analyzed across time using repeated measures two-way ANOVA with Tukey s post hoc test. Mechanical withdrawal thresholds and the percent response to a 3.31 mn monofilament were compared across time (Baseline 1 through POD 49) using a 2-way ANOVA with Bonferroni post hoc analysis. Withdrawal thresholds for SNI and sham were compared POD 90 and POD 180 with Mann Whitney U tests. Skin-Nerve Data: Each fiber type was analyzed for: 1) number of action potentials fired across mechanical forces using a two-way ANOVA with Bonferroni post hoc comparisons, 2) conduction velocity using a one-way ANOVA with Tukey s multiple comparisons, 3) von Frey thresholds using Kruskal-Wallis with Dunn s multiple comparisons, and 4) percent spontaneous fibers using Chi-square analysis. Column statistics of each fiber type were analyzed to compare the sum of the average number of action potentials fired across all forces. Percent distribution of the fiber types was compared using Chi-square analysis and Fisher s exact post hoc tests. Data analysis was completed using Prism 6 Software (GraphPad, La Jolla, CA). Abbreviations SNI: Spared nerve injury; CCI: Chronic constriction injury; SNL: Spinal nerve ligation; POD: Post, operative day; AM: Aδ-Mechanoreceptor; DRG: Dorsal root ganglia. Competing interests The authors declare that they have no competing interests. Authors contributions All authors read and approved the final manuscript. AS and CO conducted the experiments and analyzed the data. AS and CS designed the study and wrote the manuscript. Acknowledgements This work was completed with support from the National Institute of Health grants NS040538 and NS070711 to C.L.S. Received: 29 July 2013 Accepted: 22 November 2013 Published: 29 November 2013 References 1. Decosterd I, Woolf CJ: Spared nerve injury: an animal model of persistent peripheral neuropathic pain. Pain 2000, 87:149 158. 2. Magrinelli F, Zanette G, Tamburin S: Neuropathic pain: diagnosis and treatment. Pract Neurol 2013, 13:262 307. 3. Bourquin AF, Suveges M, Pertin M, Gilliard N, Sardy S, Davison AC, et al: Assessment and analysis of mechanical allodynia-like behavior induced by spared nerve injury (SNI) in the mouse. Pain 2006, 122:14. 4. Abe K, Fujii Y, Nojima H: Evaluation of hyperalgesia in spared nerve injury model using mechanical, thermal, and chemical stimuli in the mouse. Neurol Res 2011, 33:656 662. 5. Bennett GJ, Xie YK: A peripheral mononeuropathy in rat that produces disorders of pain sensation like those seen in man. Pain 1988, 33:87 107. 6. Seltzer Z, Dubner R, Shir Y: A novel behavioral model of neuropathic pain disorders produced in rats by partial sciatic nerve injury. Pain 1990, 43:205 218. 7. Kim SH, Chung JM: An experimental model for peripheral neuropathy produced by segmental spinal nerve ligation in the rat. Pain 1992, 50:355 363. 8. Koltzenburg M, Scadding J: Neuropathic pain. Curr Opin Neurol 2001, 14:641 647. 9. Shields SD, Eckert WA III, Basbaum AI: Spared nerve injury model of neuropathic pain in the mouse: a behavioral and anatomic analysis. J Pain 2003, 4:465 470. 10. Ali Z, Ringkamp M, Hartke TV, Chien HF, Flavahan NA, Campbell JN, et al: Uninjured C-fiber nociceptors develop spontaneous activity and alpha-adrenergic sensitivity following L6 spinal nerve ligation in monkey. J Neurophysiol 1999, 81:455 466. 11. Shim B, Kim DW, Kim BH, Nam TS, Leem JW, Chung JM: Mechanical and heat sensitization of cutaneous nociceptors in rats with experimental peripheral neuropathy. Neuroscience 2005, 132:193 201. 12. Koltzenburg M, Kees S, Budweiser S, Ochs G, Toyka KV: The Properties of Unmyelinated Nociceptive Afferents Change in a Painful Chronic Constriction Neuropathy. InProceedings of the 7th World Congress on Pain, Progress in Pain Research and Management, Vol. 2. Edited by Gebhart GF, Hammond DL, Jensen TS. Seattle: IASP Press; 1994:511 522. 13. Tal M, Eliav E: Abnormal discharge originates at the site of nerve injury in experimental constriction neuropathy (CCI) in the rat. Pain 1996, 64:511 518. 14. Garrison SR, Dietrich A, Stucky CL: TRPC1 contributes to light-touch sensation and mechanical responses in low-threshold cutaneous sensory neurons. JNeurophysiol2012, 107:913 922. 15. Lennertz RC, Medler KA, Bain JL, Wright DE, Stucky CL: Impaired sensory nerve function and axon morphology in mice with diabetic neuropathy. J Neurophysiol 2011, 106:905 914. 16. Xiao WH, Bennett GJ: Chemotherapy-evoked neuropathic pain: Abnormal spontaneous discharge in A-fiber and C-fiber primary afferent neurons and its suppression by acetyl-l-carnitine. Pain 2008, 135:262 270. 17. Liu CN, Wall PD, Ben-Dor E, Michaelis M, Amir R, Devor M: Tactile allodynia in the absence of C-fiber activation: altered firing properties of DRG neurons following spinal nerve injury. Pain 2000, 85:503 521. 18. Sun Q, Tu H, Xing GG, Han JS, Wan Y: Ectopic discharges from injured nerve fibers are highly correlated with tactile allodynia only in early, but not late, stage in rats with spinal nerve ligation. Exp Neurol 2005, 191:128 136. 19. Gold MS: Spinal nerve ligation: what to blame for the pain and why. Pain 2000, 84:117 120. 20. Kwon MJ, Kim J, Shin H, Jeong SR, Kang YM, Choi JY, et al: Contribution of macrophages to enhanced regenerative capacity of dorsal root ganglia sensory neurons by conditioning injury. J Neurosci 2013, 33:15095 15108. 21. Kingery WS, Vallin JA: The development of chronic mechanical hyperalgesia, autotomy and collateral sprouting following sciatic nerve section in rat. Pain 1989, 38:321 332. 22. Devor M, Schonfeld D, Seltzer Z, Wall PD: Two modes of cutaneous reinnervation following peripheral nerve injury. J Comp Neurol 1979, 185:211 220. 23. Wu G, Ringkamp M, Hartke TV, Murinson BB, Campbell JN, Griffin JW, et al: Early onset of spontaneous activity in uninjured C-fiber nociceptors after injury to neighboring nerve fibers. J Neurosci 2001, 21:RC140. 24. Schafers M, Lee DH, Brors D, Yaksh TL, Sorkin LS: Increased sensitivity of injured and adjacent uninjured rat primary sensory neurons to exogenous tumor necrosis factor-alpha after spinal nerve ligation. J Neurosci 2003, 23:3028 3038. 25. Lennertz RC, Kossyreva EA, Smith AK, Stucky CL: TRPA1 mediates mechanical sensitization in nociceptors during inflammation. PLoS One 2012, 7:e43597. doi:10.1186/1744-8069-9-61 Cite this article as: Smith et al.: Mechanical sensitization of cutaneous sensory fibers in the spared nerve injury mouse model. Molecular Pain 2013 9:61.