Abstract

Vincristine-induced peripheral neuropathy is a common side effect of vincristine treatment, which is accompanied by pain and can be dose-limiting. The molecular mechanisms that underlie vincristine-induced pain are not well understood. We have established an animal model to investigate pathophysiological mechanisms of vincristine-induced pain. Our previous studies have shown that the tetrodotoxin-sensitive voltage-gated sodium channel Nav1.6 in medium-diameter dorsal root ganglion (DRG) neurons contributes to the maintenance of vincristine-induced allodynia.

In this study, we investigated the effects of vincristine administration on excitability in small-diameter DRG neurons and whether the tetrodotoxin-resistant (TTX-R) Nav1.8 channels contribute to mechanical allodynia. Current-clamp recordings demonstrated that small DRG neurons become hyper-excitable following vincristine treatment, with both reduced current threshold and increased firing frequency. Using voltage-clamp recordings in small DRG neurons, we now show an increase in TTX-R current density and a −7.3 mV hyperpolarizing shift in the half-maximal potential (V1/2) of activation of Nav1.8 channels in vincristine-treated animals, which likely contributes to the hyperexcitability that we observed in these neurons.

Notably, vincristine treatment did not enhance excitability of small DRG neurons from Nav1.8 knockout mice, and the development of mechanical allodynia was delayed but not abrogated in these mice. Together, our data suggest that sodium channel Nav1.8 in small DRG neurons contributes to the development of vincristine-induced mechanical allodynia.

Introduction

The microtubule-disrupting agent vincristine often produces a dose-dependent peripheral neuropathy with sensory, autonomic and motor symptoms.1-3 Sensory symptoms are most frequently seen in vincristine-induced peripheral neuropathy (VIPN), including early manifestation of numbness and tingling of hands and feet, distal hyperesthesia and neuropathic pain which can be severe.4 Vincristine-induced pain appears to be associated with a poly-modal neuropathy that involves Aβ, Aδ, and C fibres,5 but its underlying molecular mechanisms are still not well understood.

Multiple mechanisms may contribute to the pathophysiology of VIPN.6-8 Vincristine destabilizes axonal microtubules9-11 and disrupts both kinesin-mediated anterograde and dynein-mediated retrograde microtubule-dependent axonal transport.12 In addition, vincristine treatment induces a strong inflammatory response,13 including the infiltration of monocytes/macrophages into sciatic nerve and dorsal root ganglia (DRG), which leads to release of interleukin (IL)-6,14 and production of reactive oxygen species (ROS) and activation of neuronal TRPA1 channels.15 Minocycline inhibits activation of immune cells and has a protective effect against vincristine-induced mechanical allodynia.16 At peak pain severity, vincristine treatment is associated with a significant increase in spontaneous discharge in A- and C-fibres17 and with central sensitization in spinal cord wide dynamic range (WDR) neurons,18 which suggests the involvement of multiple classes of ligand-gated ion channels and receptors,19-21 as well as voltage-gated sodium (Nav) channels.22-25

Nav channels are essential for the initiation and propagation of action potentials in spontaneously firing neurons and stimulus-evoked activity of these neurons, and previous studies have suggested the involvement of peripheral Nav channels in VIPN. Vincristine has been reported to induce an increase in immunoreactivity for tetrodotoxin-sensitive (TTX-S) sodium channel isoforms Nav1.3 and Nav1.6, as well as the tetrodotoxin-resistant (TTX-R) isoform Nav1.8.23 Nav1.8 is particularly important for the different pain modalities, because it is the major contributor to the upstroke of action potential and repetitive firing in C-fibre nociceptors.26-28 Early pharmacological evidence suggests that TTX-R Nav channels may play a key role in vincristine-induced mechanical allodynia; systemic sodium channel blockers such as lidocaine and mexiletine attenuate tactile allodynia in rodent models of VIPN, while TTX has no effect.24,25 However, neither anti-sense oligodeoxynucleotides targeting Nav1.8 channels nor pharmacological inhibition of Nav1.8 with the blocker A-803467 produce any antinociceptive effects in vincristine-induced mechanical allodynia.24,29 These pharmacological results have been interpreted as suggesting a Nav1.8-independent mechanism for vincristine-induced pain but have not been reconciled with the published immunohistochemical data reporting an increase in the expression of this channel.

We recently developed a mouse model of vincristine-induced mechanical allodynia and showed a significant upregulation of Nav1.6-mediated TTX-S Na+ currents in medium DRG neurons, but not in small DRG neurons, and an attenuation of maintenance but not induction of mechanical allodynia in mice in which Nav1.6 channels are knocked out in Nav1.8-positive neurons [Nav1.6Nav1.8 conditional knockout (KO)].22 Interestingly, we also showed in the same study that the KO of Nav1.7 channels in Nav1.8-positive neurons (Nav1.7Nav1.8 conditional KO), which accounts for the majority (70%) of TTX-S current in these small DRG neurons,30,31 does not ameliorate vincristine-induced mechanical allodynia. However, it has been shown that C-fibre nociceptors (small-diameter DRG neurons) become mechanically hyper-responsive after systemic administration of vincristine.32,33 Thus, the mechanism underlying the hyperactivity of these neurons, and the contribution of Nav1.8 remain to be determined.

In the present study, we investigated whether vincristine in our animal model causes hyper-excitability of small diameter DRG neurons and whether TTX-R sodium channel Nav1.8 contributes to these changes. We showed that vincristine treatment increases the Nav1.8 TTX-R current density and induces a hyperpolarizing shift in the activation of these channels in small DRG neurons, consistent with the increased activity in these neurons. We also demonstrated a delay in the onset of vincristine-induced mechanical allodynia in Nav1.8 KO mice, supporting a role for this channel in the development of vincristine-induced pain.

Materials and methods

Animals

Seventy adult mice (3–4 months old) of both sexes (31 males and 39 females) were used in this study. Wild-type (WT) C57BL/6 mice (n = 46) were obtained from Charles River for the first behaviour experiment. For the second experiment, we used Nav1.8 KO mice (n = 12) and their WT littermates (n = 11), which carry a biallelic Cre insertion in the ATG-carrying exon of Scn10a34 and a Cre-reporter cassette (loxP-stop-loxP-tdTomato fluorescent protein), which produces a red fluorescent protein in neurons that express a functional Cre recombinase.30 Animals were group housed (maximum five per cage) in a controlled environment (22 ± 2°C temperature and 55%–65% humidity) and in artificially lighted rooms on a well-defined 12 h light/dark cycle (lights on at 6:00 a.m.). Food and water were provided ad libitum throughout the experiment except during behavioural testing. Mice were acclimated to the behavioural testing environment for 2 weeks before experimental procedures. The experiments were performed in accordance with the National Institutes of Health Guide for the Care and Use of Laboratory Animals and approved by the Institutional Animal Care and Use Committee of the Veterans Administration Connecticut Healthcare System. All animals were weighed every 3 days and the weight gain was calculated. The experimenter was blinded to treatment and genotype.

Vincristine treatment

Vincristine sulfate (V8388, Sigma-Aldrich) was dissolved in ddH2O (degassed with nitrogen sparging) to a stock solution of 2.5 mg/ml and stored at −20°C. The stock solution was used within 1 month. The stock solution was diluted in saline to 0.25 mg/ml immediately before injections. Mice were injected intraperitoneally (i.p.) with vincristine at 3 ml/kg to a final dose of 0.75 mg/kg twice a week for 4 weeks (accumulated dose 6 mg/kg). Control mice received an equal volume of saline.

Behavioural studies

von Frey test

Animals were placed on an elevated wire grid and the plantar surface of both hind paws was presented with a series of calibrated von Frey filaments (Stoeling). Each filament was applied for 5 s. A nocifensive response was recorded when flinching, withdrawal, paw licking or toe spreading was observed. At least 5 min of rest were included between each stimulus. The 50% withdrawal threshold was determined using the ‘up-down’ method, which requires four stimuli straddling the threshold.

Hargreaves test

Animals were placed on a glass plate, and the plantar paw surface was exposed to radiant heat using a Hargreaves apparatus (IITC) and the paw withdrawal latency was measured. The heat stimulation was repeated three times on both hind paws. At least 5 min of rest were included consecutive tests. The cut-off value was set at 30 s to prevent tissue damage.

Gait analysis

Animals were placed in the tunnel of a Catwalk apparatus (Noldus Information Technology) and could walk freely from one end to the other. Walking patterns were recorded by a high-speed camera collecting reflexive fluorescent light from the contact surface between the paws and glass platform. All experiments were performed in a dark room. Analysis was performed using the Catwalk XT 10.1 software. Three trials were recorded for each animal and time point. The parameters used to analyse gait were stride length and print area.

Electron microscopy analyses of axons of sciatic nerve

We followed our previously described method for preparing sciatic nerve for structural analysis.22 Briefly, mice (two males and two females each for the control and vincristine groups) were anaesthetized with ketamine/xylazine (100/10 mg/kg, i.p.) and transcardially perfused with 0.01 M PBS (pH 7.4) followed by ice-cold 4% paraformaldehyde in 0.14 M Sorensen’s phosphate buffer (pH 7.4). Sciatic nerves at mid-thigh level (distal to the trifurcation) were dissected and transferred to 2.5% glutaraldehyde and 2% paraformaldehyde in 0.1 M sodium cacodylate buffer (pH 7.4) at 4°C overnight. Samples were then post-fixed with 1% osmium tetroxide, 0.8% potassium ferrocyanide in 0.1 M cacodylate buffer, post-stained with 2% uranyl acetate, dehydrated and embedded in fresh EPON. Tissues were sectioned with a Leica UC7 ultramicrotome in 50 nm sections and imaged on an FEI Tecnai G2 Spirit Biotwin transmission electron microscope at 80 kV with an AMT NanoSprint 15 camera. To quantify the number of myelinated nerves, six images were investigated per mouse. Axon and myelin areas of myelinated nerves were measured, and the g-ratio was calculated by dividing the area of the axon by the outer area of the myelinated fibre. All measurements were carried out using ImageJ v.1.50. Statistical significance was determined (P < 0.05) using unpaired Student’s t-tests. All results are presented as mean ± standard error of the mean (SEM).

Primary mouse dorsal root ganglia neuron isolation

DRG neurons from thoracic and lumbar regions were isolated from mice (both male and female) using protocols previously described.35,36 Briefly, DRG neurons were harvested and first incubated at 37°C for 20 min in complete saline solution (CSS) (in mM: 137 NaCl, 5.3 KCl, 1 MgCl2, 25 sorbitol, 3 CaCl2 and 10 HEPES, adjusted to pH 7.2 with NaOH) supplemented with 0.5 U/ml Liberase TM (Roche) and 0.6 nM EDTA. Then the tissue was incubated for 15 min at 37°C in CSS containing 0.5 U/ml Liberase TL (Roche), 0.6 mM EDTA and 30 U/ml papain (Worthington Biochemical). DRG neurons were then centrifuged and triturated in 0.5 ml of DRG culture medium containing 1.5 mg/ml bovine serum albumin (low endotoxin) and 1.5 mg/ml trypsin inhibitor (Sigma). The cell suspension was filtered through a 70 μm nylon mesh cell strainer (Becton Dickinson) to remove debris, and the mesh was then washed twice with 2 ml of DRG culture medium. DRG neurons were pelleted by centrifugation and resuspended in 1 ml of DRG culture medium. Aliquots of 100 μl cell suspension were seeded onto poly-D-lysine/laminin-coated coverslips (BD) and incubated at 37°C in a 95% air/5% CO2 (vol/vol) incubator. After 45 min of incubation, DRG culture medium was added into each well to a final volume of 1 ml. DRG neuron cultures were maintained at 37°C in a 95% air/5% CO2 (vol/vol) incubator for 18–30 h before current-clamp or voltage-clamp recording. For current-clamp recording, nerve growth factor (50 ng/ml) and glial cell line-derived neurotrophic factor (50 ng/ml) were added to the DRG culture medium.

Electrophysiology

Voltage-clamp

Voltage-clamp recordings were conducted within 24 h after DRG neuron isolation using an EPC 10 amplifier and the Patchmaster program (v 53; HEKA Elektronik) at room temperature (22 ± 1°C). Small (25–30 µm) DRG neurons with no apparent neurites were selected for voltage-clamp recording. Fire-polished electrodes were fabricated from 1.6 mm outer diameter borosilicate glass micropipettes (World Precision Instruments) using a Sutter Instruments P-97 puller and had a resistance of 0.6–1.3 MΩ. Pipette potential was adjusted to zero before seal formation. Liquid junction potential was not corrected. To reduce voltage errors, 60%–85% series resistance compensation was applied. Linear leak currents were subtracted out using the P/6 method. Current traces were sampled at 50 kHz and filtered with a low-pass Bessel setting of 10 kHz. The pipette solution contained (in mM): 140 CsF, 10 NaCl, 1 EGTA, 10 dextrose and 10 HEPES, pH 7.3 with CsOH. The extracellular bath solution contained (in mM): 140 NaCl, 3 KCl, 1 MgCl2, 1 CaCl2, 10 HEPES, 5 CsCl, 20 tetraethylammonium chloride (TEA·Cl), pH 7.3 with NaOH. CdCl2 (0.1 mM) and 4-aminopyridine (4 mM) were added to the bath solution to block endogenous calcium currents and potassium currents, respectively. To record TTX-resistant currents, 1 µM TTX was added to the bath. DRG neurons were held at −100 mV. After a 5-min equilibration period once whole-cell configuration was achieved, total voltage-gated sodium currents were recorded using a series of 100-ms step depolarizations (−80 to +40 mV in 5-mV increments at 5-s intervals) from the holding potential. Current density was calculated by normalizing maximal transient peak currents with cell capacitance. For analysis of gating properties, data were excluded if voltage error exceeded 5 mV for small DRG neurons. Peak inward currents obtained from activation protocols were converted to conductance values using the equation

(1)

where G is the conductance, I is the peak inward current, Vm is the membrane potential step used to elicit the response and ENa is the reversal potential for the sodium channel, which is determined for each cell using the x-axis intercept of a linear fit of the peak inward current responses to the last six voltage steps of the activation protocol.

Conductance data were normalized by the maximum conductance value and fit with a Boltzmann equation of the form

(2)

where V1/2 is the midpoint of activation and k is a slope factor.

Steady-state fast inactivation was assessed with a series of 500-ms prepulses (−140 mV to −10 mV in 10-mV increments); the remaining non-inactivated channels were activated by a 40-ms step depolarization to −10 mV. The steady-state slow inactivation was measured with a series of 30-s prepulses, ranging from −130 to +20 mV in a 10-mV increment, followed by a 100-ms step depolarization to −100 mV and then a 20-ms test pulse at −10 mV to assess the available non-inactivated channels. Peak inward currents obtained from steady-state fast or slow inactivation were normalized by the maximum current amplitude and fit with a Boltzmann equation of the form

(3)

where Vm represents the inactivating prepulse membrane potential and V1/2 represents the midpoint of fast or slow inactivation.

Current-clamp

Current-clamp recordings were conducted 24 h after DRG neuron isolation using an EPC 10 amplifier and the Patchmaster program (v. 53; HEKA Elektronik) at room temperature (22 ± 1°C). Small (25–30 µm) DRG neurons were selected for current-clamp recording. Fire-polished electrodes were fabricated from 1.6 mm outer diameter borosilicate glass micropipettes (World Precision Instruments) using a Sutter Instruments P-97 puller and had a resistance of 0.7–1.5 MΩ when filled with the pipette solution, which contained the following (in mM): 140 KCl, 0.5 EGTA, 5 HEPES, 3 MgATP, pH 7.30 with KOH (adjusted to 310 mOsm with dextrose). The extracellular solution contained the following (in mM): 140 NaCl, 3 KCl, 2 MgCl2, 2 CaCl2, 10 HEPES, pH 7.30 with NaOH (adjusted to 320 mOsm with dextrose). Whole-cell configuration was obtained in voltage-clamp mode before proceeding to the current-clamp mode. Cells with stable (<10% variation) resting membrane potentials (RMP) more negative than −35 mV and overshooting action potentials (>85 mV, from RMP to peak) were used for additional data collection. Input resistance was determined by the slope of a linear fit to hyperpolarizing responses to current steps from −5 to −40 pA in −5 pA increments. Threshold was determined by the first action potential elicited by a series of depolarizing current injections (200 ms) that increased in 5 pA increments. Action potential frequency was determined by quantifying the number of action potentials elicited in response to depolarizing current injections (500 ms) of different strengths (25–500 pA in 25 pA increments).

Real-time PCR neuronal ion channels array

Animals (males only) were perfused transcardially with cold PBS on Day 28 after vincristine or vehicle treatment and bilateral L3–L5 DRGs were collected for real-time (RT) PCR array. The DRGs from each animal were pooled. The tissue was frozen with liquid nitrogen and subsequently disrupted and homogenized using the TissueLyser system (Qiagen). The homogenization procedure involved two 1-min runs of the TissueLyser at 50 Hz. Total RNA was extracted and purified using the RNeasy Mini Kit (Qiagen). The concentration and purity of RNA were verified by measuring the absorbance at 260 and 280 nm (NanoDrop 2000, Thermo Scientific). Starting with 1 µg of total RNA, residual genomic DNA removal and reverse transcription into complementary DNA (cDNA) was performed using the RT2 First Strand Kit (Qiagen). A 96-well RT2 Profiler PCR array containing mouse neuronal ion channels (PAMM-036ZD) configured for use in the Bio-Rad CFX96 RT PCR system (Bio-Rad) was used to quantify gene expression. To minimize the variance in sample processing, the samples were handled by the same experimenter. Experiments were performed on tissue from three vincristine-treated animals and three control animals, and the experimenter was blinded.

Statistical analysis

Electrophysiological data were analysed using Fitmaster (HEKA Elektronik) and Origin (Microcal Software) software, and statistical significance was determined by unpaired Student’s t-test when data obeyed normal distribution or non-parametric Mann–Whitney test when data were not normally distributed. Behavioural data were analysed using Prism (GraphPad), and statistical significance was determined (P < 0.05) using two-way ANOVA with Fischer’s least significant difference (LSD) test. Unless otherwise noted, statistical significance for pairwise comparisons was determined (P < 0.05) using an independent t-test. All results are presented as mean ± SEM.

Results

Vincristine induced mechanical allodynia in wild-type mice

A balanced cohort of male (n = 24) and female (n = 22) WT C57/Bl6 mice was used. Baseline mechanical sensitivity measured by von Frey test showed no difference between sexes (males: 1.07 ± 0.09 g; females: 1.13 ± 0.09 g; P = 0.54, unpaired Student’s t-test) (Fig. 1A). Similarly, baseline thermal sensitivity measured by Hargreaves showed no difference between males and females (males: 18.54 ± 0.63 s; females: 16.69 ± 0.78 s; P = 0.07, unpaired Student’s t-test) (Fig. 1B). The vincristine treatment (0.75 mg/kg) twice per week for 4 weeks showed significant effect on the von Frey test [F(1,44) = 21.40; P < 0.0001], reflecting the development of mechanical allodynia at Day 14 (saline: 1.10 ± 0.07 g, vincristine: 0.80 ± 0.06 g; P < 0.05) that persisted through the third (Day 21) (saline: 1.09 ± 0.11 g; vincristine: 0.47 ± 0.08 g; P < 0.001) and the fourth week of treatment (Day 28) (saline: 1.17 ± 0.14 g; vincristine: 0.45 ± 0.06 g; P < 0.001) (Fig. 1C). In addition, when vincristine treatment had the greatest effect (Day 28), we did not detect a significant effect of sex [treatment effect: F(1,40) = 18.15, P = 0.0001; sex effect: F(1,40) = 1.017, P = 0.32, two-way ANOVA]. Hargreaves test revealed a significant interaction between time and treatment [F(41,76) = 3.56, P = 0.0081] with a difference in thermal threshold in the last treatment week (Week 4) (saline: 15.42 ± 0.65 s; vincristine: 12.77 ± 0.61 s; P < 0.05) (Fig. 1D); there was no sex effect: F(1,40) = 1.017, P = 0.32. Vincristine treatment showed an effect on body weight gain. On the last week of treatment, the saline group showed an average weight greater than that of the vincristine group (saline: 25.04 ± 0.84 g; vincristine: 22.35 ± 0.62 g; P < 0.01) (Fig. 1E). Gait parameters [print area (Fig. 1F) and stride length (Fig. 1G)] did not show any abnormality in the vincristine-treated group. Supplementary Fig. 1 illustrates behaviour test data for each sex separately, showing that the vincristine effect was not affected by the sex of the mice.

Vincristine-induced mechanical allodynia in wild-type mice. (A) Baseline mechanical sensitivity was tested by von Frey, which showed no difference between males (n = 24) and females (n = 22). (B) Baseline thermal sensitivity was tested by Hargreaves, which also did not show any difference between males (n = 24) and females (n = 22). (C) Mechanical sensitivity measured weekly by von Frey test in wild-type mice following vincristine treatment (Control group, n = 24, males n = 13, females n = 11; Vincristine group, n = 22, males n = 10, females n = 12). Treated mice showed a significant mechanical allodynia at Day 14 which persisted through Day 28 [*P < 0.05; ***P < 0.001; two-way ANOVA with Fischer’s least significant difference (LSD) test]. (D) A significant reduction in thermal sensitivity at Day 28 (*P < 0.05; two-way ANOVA with Fischer’s LSD test). (E) Time course of change in body weight in vincristine-treated and control mice. Vincristine-treated mice showed a lower weight gain at the end of treatment (**P < 0.01; two-way ANOVA with Fischer’s LSD test). (F) Print area and (G) stride length of hind paws measured on the Catwalk apparatus show no difference in gait analysis between control and treated groups. Error bars indicate standard error of the mean. BSL = baseline.
Figure 1

Vincristine-induced mechanical allodynia in wild-type mice. (A) Baseline mechanical sensitivity was tested by von Frey, which showed no difference between males (n = 24) and females (n = 22). (B) Baseline thermal sensitivity was tested by Hargreaves, which also did not show any difference between males (n = 24) and females (n = 22). (C) Mechanical sensitivity measured weekly by von Frey test in wild-type mice following vincristine treatment (Control group, n = 24, males n = 13, females n = 11; Vincristine group, n = 22, males n = 10, females n = 12). Treated mice showed a significant mechanical allodynia at Day 14 which persisted through Day 28 [*P < 0.05; ***P < 0.001; two-way ANOVA with Fischer’s least significant difference (LSD) test]. (D) A significant reduction in thermal sensitivity at Day 28 (*P < 0.05; two-way ANOVA with Fischer’s LSD test). (E) Time course of change in body weight in vincristine-treated and control mice. Vincristine-treated mice showed a lower weight gain at the end of treatment (**P < 0.01; two-way ANOVA with Fischer’s LSD test). (F) Print area and (G) stride length of hind paws measured on the Catwalk apparatus show no difference in gait analysis between control and treated groups. Error bars indicate standard error of the mean. BSL = baseline.

Vincristine does not cause significant changes in peripheral nerve

We have previously shown that the regimen of vincristine treatment employed here does not produce overt structural changes to the axons in the sciatic nerve.22 We investigated whether the vincristine regimen in this cohort of animals replicated this finding. The morphology of the myelinated axons appeared similar between saline-treated and vincristine-treated groups. The integrity of the sciatic nerve at mid-thigh level was generally not affected by the vincristine treatment. As shown in Fig. 2, the morphology of myelinated axons and integrity of myelin sheaths appeared unaffected in vincristine-treated mice. Bundles of unmyelinated axons with clearly defined structures were visible in cross sections of peripheral nerve in both control and vincristine-treated mice. The morphometry of the transverse sections of sciatic nerve was also analysed. We did not observe a statistically significant difference in the density of myelinated fibres (saline: 29.25 ± 3.54/mm2; vincristine: 19.75 ± 2.39/mm2; P = 0.07) (Fig. 2B), the area of axons (saline: 5.33 ± 0.84 μm2; vincristine: 6.83 ± 0.58 μm2; P = 0.19), myelin sheaths (saline: 8.66 ± 0.99 μm2, vincristine: 12.35 ± 1.33 μm2; P = 0.07) (Fig. 2C) or g-ratio (saline: 0.65 ± 0.05; vincristine: 0.60 ± 0.07; P = 0.49) (Fig. 2D) between groups. These data were consistent with our previous observations22 that vincristine does not cause significant axonal loss or degeneration at the peripheral nerve level.

Vincristine did not cause changes in sciatic nerve structure. [A(i–iv)] Electron micrographs (EM) of transverse ultrathin sections of the sciatic nerve from control and vincristine-treated mice. Scale bar = 2 μm. Sections showing myelinated and unmyelinated fibres of control (i) and vincristine (iii) mice. Scale bar = 1 μm. No apparent changes on myelin sheaths of the axons of treated animal (iv) compared to control (iii). Yellow arrows indicate unmyelinated fibres. m = mitochondria. (B) Density of myelinated fibres (number/mm2), (C) axon and myelin areas (µm2) of myelinated fibres and (D) g-ratio of myelinated fibres. No significant difference among groups, n = 4/group. SAL = saline; VIN = vincristine.
Figure 2

Vincristine did not cause changes in sciatic nerve structure. [A(iiv)] Electron micrographs (EM) of transverse ultrathin sections of the sciatic nerve from control and vincristine-treated mice. Scale bar = 2 μm. Sections showing myelinated and unmyelinated fibres of control (i) and vincristine (iii) mice. Scale bar = 1 μm. No apparent changes on myelin sheaths of the axons of treated animal (iv) compared to control (iii). Yellow arrows indicate unmyelinated fibres. m = mitochondria. (B) Density of myelinated fibres (number/mm2), (C) axon and myelin areas (µm2) of myelinated fibres and (D) g-ratio of myelinated fibres. No significant difference among groups, n = 4/group. SAL = saline; VIN = vincristine.

Vincristine causes a significant hyperpolarizing shift in voltage-dependence of Nav1.8 channel activation

We previously reported a vincristine-induced small 3-mV hyperpolarizing shift in V1/2 of TTX-S channel activation and no change of TTX-S current amplitude in small DRG neurons.22 We reasoned that the minimal changes in TTX-S current were unlikely to account for the hyperexcitability of small DRG neurons observed in vincristine-treated mice. In this study, we investigated the effect of vincristine treatment on the current density and gating properties of TTX-R channels in small DRG neurons (25–30 µm).

We used voltage-clamp recordings to investigate the effect of vincristine treatment on TTX-R current, primarily that produced by the Nav1.8 channels, in small-diameter DRG neurons (25–30 µm) isolated from control and vincristine-treated mice (both male and female) that had undergone behavioural testing (Fig. 1). After establishing whole-cell patch, DRG neurons were held at −80 mV to inactivate Nav1.9 channels, and TTX (1 µM) was applied in the extracellular solution to block all TTX-S channels. Figure 3A and B shows representative Nav1.8 currents in small DRG neurons from WT mice receiving a 4-week treatment of saline and vincristine, respectively. Vincristine treatment significantly increased the Nav1.8 current density in small DRG neurons (saline: 253.35 ± 40.24 pA/pF, n = 15; vincristine: 448.35 ± 45.01 pA/pF, n = 17; P = 0.003) (Fig. 3C). We previously reported that vincristine treatment did not alter the TTX-R current density.22 The difference between the previous study and our current findings is due to methodological differences between the two studies. We previously used a reduced NaCl gradient (30 mM) to reduce the amplitude of the total sodium current, which permitted good clamping of the neuron. These conditions may have confounded the measurements of the amplitude of the TTX-R currents. In this study, we used full sodium external solution and blocked TTX-S channels using 1 µM TTX, which permitted a more accurate measurement of the Nav1.8 current density.

Vincristine causes a significant increase in current density and a hyperpolarizing shift in Nav1.8 activation in small dorsal root ganglion neurons. (A and B) Representative Nav1.8 currents in small dorsal root ganglion (DRG) neurons from control and vincristine-treated mice. (C) Nav1.8 current density in small DRG neurons from control (n = 15) and vincristine-treated (n = 17) groups show statistically significant increase. (D) Boltzmann fit of conductance of Nav1.8 current for control (n = 11) and vincristine-treated (n = 9) groups shows a hyperpolarizing shift of −7.3 mV in the half-maximal potential (V1/2) of channel activation. (E) Steady-state fast inactivation of Nav1.8 current in control (n = 15) and vincristine-treated (n = 10) groups shows comparable inactivation. (F) Steady-state slow inactivation of Nav1.8 current in control (n = 12) and vincristine-treated (n = 8) groups shows comparable inactivation. Each control group contained: three males and three females; each Vincristine-treated group contained four males and six females. Error bars indicate standard error of the mean. Nav = voltage-gated sodium channel.
Figure 3

Vincristine causes a significant increase in current density and a hyperpolarizing shift in Nav1.8 activation in small dorsal root ganglion neurons. (A and B) Representative Nav1.8 currents in small dorsal root ganglion (DRG) neurons from control and vincristine-treated mice. (C) Nav1.8 current density in small DRG neurons from control (n = 15) and vincristine-treated (n = 17) groups show statistically significant increase. (D) Boltzmann fit of conductance of Nav1.8 current for control (n = 11) and vincristine-treated (n = 9) groups shows a hyperpolarizing shift of −7.3 mV in the half-maximal potential (V1/2) of channel activation. (E) Steady-state fast inactivation of Nav1.8 current in control (n = 15) and vincristine-treated (n = 10) groups shows comparable inactivation. (F) Steady-state slow inactivation of Nav1.8 current in control (n = 12) and vincristine-treated (n = 8) groups shows comparable inactivation. Each control group contained: three males and three females; each Vincristine-treated group contained four males and six females. Error bars indicate standard error of the mean. Nav = voltage-gated sodium channel.

Vincristine treatment also caused a shift in the voltage-dependence of activation (Act) of Nav1.8 channels in a hyperpolarizing direction (saline: V1/2, Act = −10.86 ± 1.45 mV, n = 11; vincristine: V1/2, Act = −17.50 ± 2.14 mV, n = 9; P = 0.022) (Fig. 3D), with no apparent effect on steady-state fast-inactivation (Fast Inact) between control and vincristine-treated groups (saline: V1/2, Fast Inact = −44.72 ± 1.39 mV, n = 15; vincristine: V1/2, Fast Inact = −47.76 ± 1.48 mV, n = 10; P = 0.151). No difference was observed in the voltage-dependence of steady-state slow-inactivation (Slow Inact) either (saline: V1/2, Slow Inact = −61.98 ± 1.38 mV, n = 12; vincristine: V1/2, Fast Inact = −60.61 ± 2.37 mV, n = 8; P = 0.627) (Fig. 3E and F).

Vincristine treatment causes hyper-excitability of small DRG neurons from wild-type mice

We studied the excitability of small DRG neurons (25–30 µm) from control and vincristine-treated mice that had undergone behavioural testing (Fig. 1). DRG neurons were isolated from mice within the first week after the cessation of treatment, a time frame that we have previously shown to maintain the effect of treatment.22 Since the behavioural testing did not uncover a sex effect, we pooled electrophysiological data from cultures of both sexes. Current-clamp recordings were performed 24 h after isolation of DRG neurons from both male and female mice in each condition. Vincristine treatment appeared to increase the percentage of spontaneously firing neurons to 13.04% (3 of 23 small cells) compared to 5.88% for control (1 of 17 cells); however, this increase did not reach statistical significance. Figure 4 shows representative action potential traces in small DRG neurons from control (Fig. 4A) and vincristine-treated mice (Fig. 4B). Vincristine significantly reduced the current threshold of action potential firing (saline: 118.13 ± 35.78 pA, n = 16; vincristine: 41.75 ± 6.31 pA, n = 20; P = 0.025) (Table 1). In addition, vincristine significantly hyperpolarized the afterhyperpolarization potential (AHP) by 7.3 mV (saline: −18.02 ± 2.25 mV, n = 16; vincristine: −25.31 ± 1.81 mV, n = 20; P = 0.017) (Table 1 and Fig. 4C). There was no significant difference in RMP (saline: −50.52 ± 1.69 mV, n = 16; vincristine: −49.71 ± 1.07 mV, n = 20; P = 0.691), input resistance (saline: 0.99 ± 0.10 GΩ, n = 16; vincristine: 0.92 ± 0.11 GΩ, n = 20; P = 0.622) or action potential amplitude, which was measured from RMP to peak (saline: 110.19 ± 3.33 mV, n = 16; vincristine: 109.39 ± 2.76 mV, n = 20; P = 0.853) (Table 1).

Vincristine increases excitability in small dorsal root ganglion neurons. (A) Representative responses of a small dorsal root ganglion (DRG) neuron from a control mouse to a series of current injections for 200 ms. The neuron did not generate an action potential until the stimulus was increased to 135 pA, which was defined as the current threshold for this cell. (B) Representative responses of a small DRG neuron from a vincristine-treated mouse to a series of current injections for 200 ms. The current threshold for this cell was 45 pA. (C) Representative action potential traces from control and vincristine-treated mice were superimposed, showing that the afterhyperpolarization potential was hyperpolarized post vincristine treatment compared with the control. (D) Summary of firing frequency in response to graded stimuli. The total number of action potentials in small DRG neurons elicited by a depolarizing current stimulus from 25 to 500 pA in 25 pA increments from control and vincristine-treated mice. Error bars indicate standard error of the mean. *P < 0.05, **P < 0.01 and ***P < 0.001.
Figure 4

Vincristine increases excitability in small dorsal root ganglion neurons. (A) Representative responses of a small dorsal root ganglion (DRG) neuron from a control mouse to a series of current injections for 200 ms. The neuron did not generate an action potential until the stimulus was increased to 135 pA, which was defined as the current threshold for this cell. (B) Representative responses of a small DRG neuron from a vincristine-treated mouse to a series of current injections for 200 ms. The current threshold for this cell was 45 pA. (C) Representative action potential traces from control and vincristine-treated mice were superimposed, showing that the afterhyperpolarization potential was hyperpolarized post vincristine treatment compared with the control. (D) Summary of firing frequency in response to graded stimuli. The total number of action potentials in small DRG neurons elicited by a depolarizing current stimulus from 25 to 500 pA in 25 pA increments from control and vincristine-treated mice. Error bars indicate standard error of the mean. *P < 0.05, **P < 0.01 and ***P < 0.001.

Table 1

Action potential characterization in small dorsal root ganglia neurons from control and vincristine-treated mice

GroupsRMP, mVR input, GΩCurrent threshold, pAAmplitude, mVAHP, mV
Control, n = 16−50.52 ± 1.690.99 ± 0.10118.13 ± 35.78110.19 ± 3.33−18.02 ± 2.25
Vincristine-treated, n = 20−49.71 ± 1.070.92 ± 0.1141.75 ± 6.31*109.39 ± 2.76−25.31 ± 1.81*
GroupsRMP, mVR input, GΩCurrent threshold, pAAmplitude, mVAHP, mV
Control, n = 16−50.52 ± 1.690.99 ± 0.10118.13 ± 35.78110.19 ± 3.33−18.02 ± 2.25
Vincristine-treated, n = 20−49.71 ± 1.070.92 ± 0.1141.75 ± 6.31*109.39 ± 2.76−25.31 ± 1.81*

*P < 0.05 versus control.

Each control group contained two males and three females; each vincristine-treated group contained three males and two females. AHP = afterhyperpolarization potential; RMP = resting membrane potential.

Table 1

Action potential characterization in small dorsal root ganglia neurons from control and vincristine-treated mice

GroupsRMP, mVR input, GΩCurrent threshold, pAAmplitude, mVAHP, mV
Control, n = 16−50.52 ± 1.690.99 ± 0.10118.13 ± 35.78110.19 ± 3.33−18.02 ± 2.25
Vincristine-treated, n = 20−49.71 ± 1.070.92 ± 0.1141.75 ± 6.31*109.39 ± 2.76−25.31 ± 1.81*
GroupsRMP, mVR input, GΩCurrent threshold, pAAmplitude, mVAHP, mV
Control, n = 16−50.52 ± 1.690.99 ± 0.10118.13 ± 35.78110.19 ± 3.33−18.02 ± 2.25
Vincristine-treated, n = 20−49.71 ± 1.070.92 ± 0.1141.75 ± 6.31*109.39 ± 2.76−25.31 ± 1.81*

*P < 0.05 versus control.

Each control group contained two males and three females; each vincristine-treated group contained three males and two females. AHP = afterhyperpolarization potential; RMP = resting membrane potential.

To assess the excitability of small DRG neurons, a series of 500-ms current injections ranging from 25 to 500 pA was applied, and the firing frequency was determined. Compared to the control group, vincristine significantly increased the firing frequency of small DRG neurons (Fig. 4D). The number of animals and cells used for all electrophysiology experiments is listed in Table 1.

Knockout of Nav1.8 delays the development of vincristine-induced mechanical allodynia

To investigate whether genetic deletion of Nav1.8 channels in DRG neurons attenuates the development or maintenance of vincristine-induced pain, we examined behavioural phenotypes of Nav1.8 KO mice and their WT littermates following vincristine treatment (Fig. 5). Nav1.8 KO and WT littermate mice exhibited similar mechanical (WT: 0.99 ± 0.10 g; Nav1.8 KO: 1.03 ± 0.11 g) and thermal baseline thresholds (WT: 12.3 ± 0.8 s; Nav1.8 KO: 12.3 ± 0.6 s). Although vincristine treatment caused mechanical allodynia in mice of both genotypes, this effect was delayed in Nav1.8 KO mice (Fig. 5). A significant mechanical allodynia developed as early as Day 7 and was maintained to Day 28 in WT mice (Fig. 5A) (P = 0.012 at Day 7, P = 0.025 at Day 14, = 0.01 at Day 21, P < 0.0001 at Day 28; two-way ANOVA with Fischer’s LSD test; statistically significant difference compared to WT baseline). In the Nav1.8 KO group; however, the reduction in mechanical threshold following vincristine treatment did not reach statistical significance until Day 28 (Fig. 5A) (P < 0.01; two-way ANOVA with Fischer’s LSD test; statistically significant difference compared to Nav1.8 KO baseline). Compared to the WT control group, KO of Nav1.8 significantly attenuated vincristine-induced mechanical allodynia at Day 7 (Fig. 5A) (Nav1.8 KO: 0.98 ± 0.10 g; WT saline: 0.676 ± 0.09 g; P = 0.046, two-way ANOVA with Fischer’s LSD test) but not at other measurement points. These observations suggested that the absence of Nav1.8 delays the onset but does not prevent the eventual development of vincristine-induced mechanical allodynia. Vincristine treatment did not cause thermal allodynia in either WT control or Nav1.8 KO mice (Fig. 5B). Weight loss was apparent in WT mice after vincristine treatment (baseline: 25.7 ± 1.16 g; Day 28: 24.1 ± 0.84 g). Nav1.8 KO mice appeared to have less weight change compared to WT controls (baseline: 24.5 ± 0.9 g; Day 28: 23.7 ± 0.74 g) (Fig. 5C).

Nav1.8 knockout delays the development of vincristine-induced mechanical allodynia. (A) Mechanical sensitivity measured weekly by von Frey test in wild-type control (WT CTRL) and Nav1.8 knockout (KO) mice following vincristine administration (0.75 mg/kg, twice a week for 4 weeks). Mechanical allodynia developed as early as Day 7 and was maintained to Day 28 in WT mice [*P = 0.012 at Day 7, *P = 0.025 at Day 14, *P = 0.010 at Day 21, ****P < 0.0001 at Day 28, compared to baseline (BSL)]. Vincristine induced a reduction in mechanical threshold in the Nav1.8 KO group but only reached statistical significance at the end of the treatment (**P < 0.01 at Day 28, compared to BSL). A significant difference in mechanical threshold between Nav1.8 KO and WT mice was seen only at Day 7 (#P = 0.046). (B) Thermal sensitivity measured weekly by Hargreaves test in WT control and Nav1.8 KO mice following vincristine treatment. Vincristine did not cause thermal hyperalgesia in WT or Nav1.8 KO animals. No difference was noted between control and Nav1.8 KO groups. (C) Body weight of WT control and Nav1.8 KO mice. No significant difference was observed between WT control and Nav1.8 KO groups. For A–C: WT control group, n = 11 (males n = 3, females n = 8); Nav1.8 KO group, n = 12 (males n = 4, females n = 8). Statistical significance was tested using two-way ANOVA with Fischer’s least significant difference test. Error bars indicate standard error of the mean. Nav = voltage-gated sodium channel.
Figure 5

Nav1.8 knockout delays the development of vincristine-induced mechanical allodynia. (A) Mechanical sensitivity measured weekly by von Frey test in wild-type control (WT CTRL) and Nav1.8 knockout (KO) mice following vincristine administration (0.75 mg/kg, twice a week for 4 weeks). Mechanical allodynia developed as early as Day 7 and was maintained to Day 28 in WT mice [*P = 0.012 at Day 7, *P = 0.025 at Day 14, *P = 0.010 at Day 21, ****P < 0.0001 at Day 28, compared to baseline (BSL)]. Vincristine induced a reduction in mechanical threshold in the Nav1.8 KO group but only reached statistical significance at the end of the treatment (**P < 0.01 at Day 28, compared to BSL). A significant difference in mechanical threshold between Nav1.8 KO and WT mice was seen only at Day 7 (#P = 0.046). (B) Thermal sensitivity measured weekly by Hargreaves test in WT control and Nav1.8 KO mice following vincristine treatment. Vincristine did not cause thermal hyperalgesia in WT or Nav1.8 KO animals. No difference was noted between control and Nav1.8 KO groups. (C) Body weight of WT control and Nav1.8 KO mice. No significant difference was observed between WT control and Nav1.8 KO groups. For AC: WT control group, n = 11 (males n = 3, females n = 8); Nav1.8 KO group, n = 12 (males n = 4, females n = 8). Statistical significance was tested using two-way ANOVA with Fischer’s least significant difference test. Error bars indicate standard error of the mean. Nav = voltage-gated sodium channel.

Vincristine does not significantly alter the excitability of small DRG neurons from Nav1.8 KO mice

To further investigate the contribution of Nav1.8 to the hyper-excitability of small DRG neurons, we examined action potential firings in small DRG neurons from Nav1.8 KO mice with or without vincristine treatment. The RMP was similar between control and vincristine-treated Nav1.8-null neurons (saline: −56.3 ± 3.1 mV, n = 7; vincristine: −59.2 ± 2.2 mV, n = 7, P = 0.454). In agreement with our published data,26 in the absence of Nav1.8 channels, small-diameter DRG neurons were unable to fire all-or-none action potentials at their RMP (Fig. 6A and B), with small-amplitude action potentials that crossed 0 mV being observed in a few cells with relatively hyperpolarized membrane potential (Fig. 6C and D). Hyperpolarizing the cell membrane potential in DRG neurons to −80 mV allowed TTX-S channels to recover from inactivation that occurs at RMP, thus more TTX-S channels became available for action potential generation, which restored the ability of those cells to fire all-or-none action potentials with normal amplitude (Fig. 6E and F). The mean current thresholds at −80 mV were 163 ± 22 pA (n = 4) for control neurons and 184 ± 36 pA (n = 7) for vincristine-treated neurons were not significantly different (P = 0.679). Figure 6G shows repetitive firing when DRG neurons were held at −80 mV, with no apparent difference between control and vincristine groups. This result is consistent with the little or limited effect of vincristine on the TTX-S channels in small-diameter DRG neurons, which we reported previously.22 Taken together, these data demonstrated that Nav1.8 KO abolished vincristine-induced hyperexcitability in small DRG neurons.

Vincristine treatment does not significantly alter the excitability of small dorsal root ganglion neurons in Nav1.8-null mice. (A–D) Representative firing of small dorsal root ganglion (DRG) neurons at resting membrane potential (RMP). Lack of Nav1.8 channels impairs action potential (AP) firing in small DRG neurons at RMP. (A and B) Most neurons could not fire all-or-none APs but graded responses; (C and D) small amplitude APs which passed 0 mV were observed in neurons with relatively hyperpolarized membrane potentials. (E and F) Holding membrane potential at −80 mV, which increases the availability of voltage-gated sodium channels in DRG neurons, allowed Nav1.8-null neurons to fire all-or-none APs, with no significant difference in mean current threshold between control (Ctrl) and vincristine-treated (VIN) neurons. (G) Firing frequency in response to graded stimuli for neurons that were held at −80 mV from Nav1.8-null mice treated as controls or with vincristine. There was no significant difference in AP generation between the two groups. Error bars indicate standard error of the mean. Nav = voltage-gated sodium channel.
Figure 6

Vincristine treatment does not significantly alter the excitability of small dorsal root ganglion neurons in Nav1.8-null mice. (AD) Representative firing of small dorsal root ganglion (DRG) neurons at resting membrane potential (RMP). Lack of Nav1.8 channels impairs action potential (AP) firing in small DRG neurons at RMP. (A and B) Most neurons could not fire all-or-none APs but graded responses; (C and D) small amplitude APs which passed 0 mV were observed in neurons with relatively hyperpolarized membrane potentials. (E and F) Holding membrane potential at −80 mV, which increases the availability of voltage-gated sodium channels in DRG neurons, allowed Nav1.8-null neurons to fire all-or-none APs, with no significant difference in mean current threshold between control (Ctrl) and vincristine-treated (VIN) neurons. (G) Firing frequency in response to graded stimuli for neurons that were held at −80 mV from Nav1.8-null mice treated as controls or with vincristine. There was no significant difference in AP generation between the two groups. Error bars indicate standard error of the mean. Nav = voltage-gated sodium channel.

Changes in gene expression of multiple ion channels in DRG neurons after exposure to vincristine

We previously reported that Nav1.6-mediated increase in TTX-S sodium current density contributes to hyper-excitability of DRG neurons after vincristine treatment,22 and our new data show an increase in the Nav1.8 TTX-R current density. We therefore hypothesized that vincristine chemotherapy might lead to upregulation of sodium channel expression. Using an RT-PCR array, we systematically examined gene expression changes of neuronal ion channels, including sodium channels, HCN channels, potassium channels and TRP channels. We used DRG neurons from male WT mice for this analysis. Vincristine did not alter sodium channel expression, including expression of Nav1.6, Nav1.7, Nav1.8 or Nav1.9; however, small but statistically significant downregulation of expression was observed in five potassium channels and TRPA1 (Supplementary Table 1).

Discussion

We have previously shown that Nav1.6 channels in medium-diameter DRG neurons contribute to the maintenance of mechanical allodynia in mice.22 We have now shown in this model that vincristine-induced mechanical allodynia is not sex-dependent. We also show a significant increase in current density and hyperpolarized activation of Nav1.8 TTX-R channels in small-diameter DRG neurons from mice of both sexes treated with vincristine. These changes in channel properties would be expected to contribute to the hyperexcitability of these neurons. Notably, deletion of Nav1.8 in DRG neurons delayed the development of vincristine-induced mechanical allodynia, but while there is a pattern of attenuated pain throughout the treatment period, loss of Nav1.8 did not abolish allodynia at the end of the 4-week treatment. Together, our data indicate that small-diameter DRG neurons contribute to the development of vincristine-induced mechanical allodynia and that sodium channel Nav1.8 is critical for this effect.

Our published data22 and evidence that we provide here show that Nav1.8 and Nav1.6 channels in small-diameter and medium-diameter DRG neurons, respectively, play an important and possibly complementary role in the induction (Nav1.8) and maintenance (Nav1.6) of mechanical allodynia in mice treated with vincristine. A limitation in our study, however, is that the knockout of Nav1.8 renders the small-diameter DRG neurons hypoexcitable because Nav1.8 channels contribute most of the current underlying the upstroke of action potential26,37; thus we cannot formally rule out the possibility that additional ion channels and receptors in small-diameter DRG neurons play a role in inducing mechanical allodynia. However, even after rescuing firing of all-or-none action potential by hyperpolarizing the neuronal membrane to −80 mV, we did not observe a vincristine-induced increase in repetitive firing, which suggested that the effect of vincristine on other targets in small diameter DRG neurons may not be enough to compensate for the loss of Nav1.8 channels during the induction stage of mechanical allodynia.

We have shown here that small-diameter DRG neurons from WT mice treated with vincristine are hyper-excitable, compared to neurons from control mice. Since we did not observe a difference in the allodynia between males and females, we combined recordings from neuronal cultures of both sexes. The hyperexcitability manifested as a 65% reduction in the action potential current threshold, and an enhancement of repetitive firing in response to graded stimuli. We also observed significant hyperpolarization of the AHP in these neurons, but there was no change in other membrane properties including RMP and input resistance (Table 1). At the channel level, we observed an increase in the current density of the Nav1.8 TTX-R currents in neurons from vincristine-treated mice but did not see an increase in the expression of the channel, which suggested a post-transcriptional and possibly post-translational modulation of the channel. Additionally, we observed a significant hyperpolarizing shift of −7.3 mV in the activation of Nav1.8, which would be expected to increase repetitive firing of DRG neurons and may also contribute to the reduction in the threshold for action potential firing. Gain-of-function mutations in Nav1.8 channels from patients with small fibre neuropathy, which shift activation in a hyperpolarizing direction of a similar magnitude, reduce the threshold for action potential firing and increase repetitive firing of DRG neurons consistent with the pain phenotype in these patients.36,38 These data support the contribution of Nav1.8 to the hyper-excitability of small DRG neurons in vincristine treated mice.

Vincristine treatment causes tissue inflammation.39 It is known that the inflammatory mediator prostaglandin E2 causes a hyperpolarizing shift in the activation of Nav1.8 channels via mechanisms that depend on PKA and PKC.40,41 Thus, the −7.3 mV hyperpolarizing shift in activation of Nav1.8 that we observed in this study could have resulted from inflammation-induced signalling cascades. Other inflammatory mediators, for example TNF-α, increase the current density of Nav1.8.42 Thus, the inflammatory milieu that develops during chemotherapy may contribute to the modulation of Nav1.8 channels in small DRG neurons. Although inflammatory mediators are known to increase the current density of the Nav1.7 TTX-S channel and increase axonal trafficking of the channel in sensory neurons,43 we did not detect changes in either current density or gating properties of the TTX-S currents in small DRG neurons from mice treated with vincristine.22 Taken together, these data point to possible Nav1.8 isoform-specific regulation by vincristine-induced inflammatory response.

The majority of small-diameter DRG neurons from Nav1.8 KO mice produce graded responses to depolarizing stimuli, with only 20% producing the all-or-none action potential.26 Hyperpolarizing the resting potential of these neurons to −80 mV could restore the firing of action potentials because enough TTX-S channels could be rescued from inactivation at hyperpolarized membrane potentials.26 Consistent with these earlier findings, our data showed that small DRG neurons from vincristine-treated Nav1.8 KO mice only produced graded responses and that the threshold for firing an all-or-none action potential when neurons were held at −80 mV was not significantly different from control mice (Fig. 6). This is in agreement with our previous data showing that vincristine treatment has no detectable effect on TTX-S channels in small DRG neurons.22 Together with our published data showing that knockout of Nav1.7, which contributes 70% of the TTX-S current in small DRG neurons,30,31 did not diminish vincristine-induced mechanical allodynia,22 we now provide evidence supporting an important role of Nav1.8 activation in the induction phase of vincristine-induced allodynia.

Vincristine-induced mechanical allodynia is likely a multi-factorial process, which involves multiple classes of ion channels and receptors in peripheral and central neurons. Transient receptor potential vanilloid 1 (TRPV1) and TRPV4,19,20 the 5-hydroxytryptamine receptor 2A20,21 T- and N-type CaV channels,39,44 might be involved. To gain insights into vincristine-mediated effects on the expression levels of ion channels that regulate neuronal excitability in our animal model, we obtained RT-PCR array data showing that vincristine treatment did not cause significant changes in the expression levels of Nav or HCN channels (Supplementary Table 1), while the expression levels of several K+ channels were significantly reduced, including KCa1.1, a large conductance calcium-activated potassium channel encoded by KCNMA1 (also termed BK or BKCa). The Kv2 and calcium-activated potassium channels are prominent contributors to action potential repolarization and afterhyperpolarization.45,46 The BKCa currents in small DRG neurons were reduced and appeared to activate more slowly 3 days after complete Freund’s adjuvant injection.47 Interestingly, Kimm et al.45 demonstrated that inhibiting BKCa channels paradoxically hyperpolarized the AHP, which was attributed to the compensatory enhancement of slowly deactivating Kv2 current during the spike. This suggests that in addition to downregulation of BK channels at the transcription level (Supplementary Table 1), channel activity may also be suppressed by vincristine-induced inflammation, and the crosstalk with the Kv channels might result in the hyperpolarizing shift of AHP (Table 1) in small-diameter DRG neurons.

Nav1.8 channels have an unequivocal role in DRG neuron firing26 and pain,28,48 and our data provide evidence for a role of Nav1.8 in the development of vincristine-induced mechanical allodynia. However, previous studies have yielded conflicting results on the role of Nav1.8 channels in vincristine-induced neuropathic pain. Levels of Nav1.8 were reported to be upregulated using immunohistochemistry,23 and pharmacological studies reported that vincristine-induced pain can be attenuated by pan Nav channel blockers lidocaine and mexiletine but not TTX, findings that were interpreted as suggesting the involvement of TTX-R Na+ channels.24,25 There are studies, however, that did not report amelioration of vincristine-induced pain in rats, in which Nav1.8 expression levels were knocked down using antisense oligonucleotides or after pharmacological block of this channel.29,49 In the current study, we observed an increase in the current density of Nav1.8 and a hyperpolarizing shift in its activation, which is predicted to contribute to hyperexcitability of small DRG neurons and demonstrated a delayed induction of vincristine-induced mechanical allodynia in Nav1.8 KO mice. The difference between our findings and those that did not report an effect of Nav1.8 in vincristine-induced pain may be related to methodological differences including the dose of the drug delivered, the species (mouse versus rat) used, sex (mixed cohort versus male only) or the stages at which mechanical allodynia was assessed.

We have previously shown that the maintenance of vincristine-induced mechanical allodynia but not its induction is attenuated in conditional KO Nav1.6Nav1.8 mice, and, in parallel, an increase in the Nav1.6 current density in medium-diameter but not small DRG neurons.22 Nav1.6 channels are the main isoforms at nodes of Ranvier of myelinated fibres,50 which could explain their critical role in the transmission of nerve impulses in these fibres that contribute to the maintenance of pain in this model. We now show a delay in the onset of vincristine-induced mechanical allodynia in Nav1.8 knockout mice, with an increase in current density and a hyperpolarizing shift in activation of Nav1.8 in small-diameter DRG neurons. It is notable that Nav1.7 channels do not appear to play an important role in either of the two phases of vincristine-induced allodynia.22 Together, these data suggest a different role of these two classes of sensory neurons in the induction and maintenance of vincristine-induced mechanical allodynia and a possible contribution of Nav1.6 (medium neurons) and Nav1.8 (small neurons) in these processes. Recently, the isoform-specific small molecule inhibitor of Nav1.8 VX-548 has shown promising results in treating acute pain in phase 2 clinical trials in patients with bunionectomy and abdominoplasty.51 An isoform-specific Nav1.6 small molecule inhibitor NBI-921352 has shown antiepileptic seizures in rodent models52 and is currently being tested in clinical trials in patients with SCN8A developmental and epileptic encephalopathy syndrome (NCT04873869). These isoform-selective inhibitors could potentially be used as a monotherapy or in combination to treat vincristine-induced pain.

Data availability

All information necessary to evaluate the findings of the paper is included in the paper. Additional data can be provided by the corresponding author upon request.

Funding

This work was supported by grants RX003621 (S.D.H.) and Center Grant B9253-C (S.D.H. and S.G.W.) from the US Department of Veterans Affairs Rehabilitation Research and Development Service, and by a grant from the Thomson Family Foundation. The Center for Neuroscience and Regeneration Research is a collaboration of the Paralyzed Veterans of America with Yale University.

Competing interests

The authors report no competing interests.

Supplementary material

Supplementary material is available at Brain online.

References

1

Windebank
AJ
,
Grisold
W
.
Chemotherapy-induced neuropathy
.
J Peripher Nerv Syst
.
2008
;
13
:
27
46
.

2

Park
SB
,
Goldstein
D
,
Krishnan
AV
, et al.
Chemotherapy-induced peripheral neurotoxicity: A critical analysis
.
CA Cancer J Clin
.
2013
;
63
:
419
437
.

3

Lavoie Smith
EM
,
Li
L
,
Chiang
C
, et al.
Patterns and severity of vincristine-induced peripheral neuropathy in children with acute lymphoblastic leukemia
.
J Peripher Nerv Syst
.
2015
;
20
:
37
46
.

4

Quasthoff
S
,
Hartung
HP
.
Chemotherapy-induced peripheral neuropathy
.
J Neurol
.
2002
;
249
:
9
17
.

5

Dougherty
PM
,
Cata
JP
,
Burton
AW
,
Vu
K
,
Weng
H-R
.
Dysfunction in multiple primary afferent fiber subtypes revealed by quantitative sensory testing in patients with chronic vincristine-induced pain
.
J Pain Symptom Manage
.
2007
;
33
:
166
179
.

6

Boyette-Davis
JA
,
Walters
ET
,
Dougherty
PM
.
Mechanisms involved in the development of chemotherapy-induced neuropathy
.
Pain Manag
.
2015
;
5
:
285
296
.

7

Sisignano
M
,
Baron
R
,
Scholich
K
,
Geisslinger
G
.
Mechanism-based treatment for chemotherapy-induced peripheral neuropathic pain
.
Nat Rev Neurol
.
2014
;
10
:
694
707
.

8

Starobova
H
,
Vetter
I
.
Pathophysiology of chemotherapy-induced peripheral neuropathy
.
Front Mol Neurosci
.
2017
;
10
:
174
.

9

Sahenk
Z
,
Brady
ST
,
Mendell
JR
.
Studies on the pathogenesis of vincristine-induced neuropathy
.
Muscle Nerve
.
1987
;
10
:
80
84
.

10

Tanner
KD
,
Reichling
DB
,
Levine
JD
.
Nociceptor hyper-responsiveness during vincristine-induced painful peripheral neuropathy in the rat
.
J Neurosci
.
1998
;
18
:
6480
6491
.

11

Topp
KS
,
Tanner
KD
,
Levine
JD
.
Damage to the cytoskeleton of large diameter sensory neurons and myelinated axons in vincristine-induced painful peripheral neuropathy in the rat
.
J Comp Neurol
.
2000
;
424
:
563
576
.

12

LaPointe
NE
,
Morfini
G
,
Brady
ST
,
Feinstein
SC
,
Wilson
L
,
Jordan
MA
.
Effects of eribulin, vincristine, paclitaxel and ixabepilone on fast axonal transport and kinesin-1 driven microtubule gliding: Implications for chemotherapy-induced peripheral neuropathy
.
Neurotoxicology
.
2013
;
37
:
231
239
.

13

Kiguchi
N
,
Maeda
T
,
Kobayashi
Y
,
Saika
F
,
Kishioka
S
.
Involvement of inflammatory mediators in neuropathic pain caused by vincristine
.
Int Rev Neurobiol
.
2009
;
85
:
179
190
.

14

Kiguchi
N
,
Maeda
T
,
Kobayashi
Y
,
Kondo
T
,
Ozaki
M
,
Kishioka
S
.
The critical role of invading peripheral macrophage-derived interleukin-6 in vincristine-induced mechanical allodynia in mice
.
Eur J Pharmacol
.
2008
;
592
:
87
92
.

15

Old
EA
,
Nadkarni
S
,
Grist
J
, et al.
Monocytes expressing CX3CR1 orchestrate the development of vincristine-induced pain
.
J Clin Invest
.
2014
;
124
:
2023
2036
.

16

Starobova
H
,
Mueller
A
,
Allavena
R
,
Lohman
RJ
,
Sweet
MJ
,
Vetter
I
.
Minocycline prevents the development of mechanical allodynia in mouse models of vincristine-induced peripheral neuropathy
.
Front Neurosci
.
2019
;
13
:
653
.

17

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
.

18

Weng
H-R
,
Cordella
JV
,
Dougherty
PM
.
Changes in sensory processing in the spinal dorsal horn accompany vincristine-induced hyperalgesia and allodynia
.
Pain
.
2003
;
103
:
131
138
.

19

Alessandri-Haber
N
,
Dina
OA
,
Joseph
EK
,
Reichling
DB
,
Levine
JD
.
Interaction of transient receptor potential vanilloid 4, integrin, and SRC tyrosine kinase in mechanical hyperalgesia
.
J Neurosci
.
2008
;
28
:
1046
1057
.

20

Chiba
T
,
Oka
Y
,
Sashida
H
, et al.
Vincristine-induced peripheral neuropathic pain and expression of transient receptor potential vanilloid 1 in rat
.
J Pharmacol Sci
.
2017
;
133
:
254
260
.

21

Thibault
K
,
Van Steenwinckel
J
,
Brisorgueil
M-J
, et al.
Serotonin 5-HT2A receptor involvement and Fos expression at the spinal level in vincristine-induced neuropathy in the rat
.
Pain
.
2008
;
140
:
305
322
.

22

Chen
L
,
Huang
J
,
Benson
C
, et al.
Sodium channel Nav1.6 in sensory neurons contributes to vincristine-induced allodynia
.
Brain
.
2020
;
143
:
2421
2436
.

23

Uceyler
N
,
Kobsar
I
,
Biko
L
, et al.
Heterozygous P0 deficiency protects mice from vincristine-induced polyneuropathy
.
J Neurosci Res
.
2006
;
84
:
37
46
.

24

Kamei
J
,
Nozaki
C
,
Saitoh
A
.
Effect of mexiletine on vincristine-induced painful neuropathy in mice
.
Eur J Pharmacol
.
2006
;
536
:
123
127
.

25

Nozaki-Taguchi
N
,
Chaplan
SR
,
Higuera
ES
,
Ajakwe
RC
,
Yaksh
TL
.
Vincristine-induced allodynia in the rat
.
Pain
.
2001
;
93
:
69
76
.

26

Renganathan
M
,
Cummins
TR
,
Waxman
SG
.
Contribution of Na(v)1.8 sodium channels to action potential electrogenesis in DRG neurons
.
J Neurophysiol
.
2001
;
86
:
629
640
.

27

Rush
AM
,
Cummins
TR
,
Waxman
SG
.
Multiple sodium channels and their roles in electrogenesis within dorsal root ganglion neurons
.
J Physiol
.
2007
;
579
(
Pt 1
):
1
14
.

28

Bennett
DL
,
Clark
AJ
,
Huang
J
,
Waxman
SG
,
Dib-Hajj
SD
.
The role of voltage-gated sodium channels in pain signaling
.
Physiol Rev
.
2019
;
99
:
1079
1151
.

29

Jarvis
MF
,
Honore
P
,
Shieh
C-C
, et al.
A-803467, a potent and selective Nav1.8 sodium channel blocker, attenuates neuropathic and inflammatory pain in the rat
.
Proc Natl Acad Sci U S A
.
2007
;
104
:
8520
8525
.

30

Shields
SD
,
Ahn
H-S
,
Yang
Y
, et al.
Nav1.8 expression is not restricted to nociceptors in mouse peripheral nervous system
.
Pain
.
2012
;
153
:
2017
2030
.

31

Vasylyev
DV
,
Han
C
,
Zhao
P
,
Dib-Hajj
S
,
Waxman
SG
.
Dynamic-clamp analysis of wild-type human Nav1.7 and erythromelalgia mutant channel L858H
.
J Neurophysiol
.
2014
;
111
:
1429
1443
.

32

Tanner
KD
,
Levine
JD
,
Topp
KS
.
Microtubule disorientation and axonal swelling in unmyelinated sensory axons during vincristine-induced painful neuropathy in rat
.
J Comp Neurol
.
1998
;
395
:
481
492
.

33

Tanner
KD
,
Reichling
DB
,
Gear
RW
,
Paul
SM
,
Levine
JD
.
Altered temporal pattern of evoked afferent activity in a rat model of vincristine-induced painful peripheral neuropathy
.
Neuroscience
.
2003
;
118
:
809
817
.

34

Stirling
LC
,
Forlani
G
,
Baker
MD
, et al.
Nociceptor-specific gene deletion using heterozygous NaV1.8-Cre recombinase mice
.
Pain
.
2005
;
113
:
27
36
.

35

Israel
MR
,
Tanaka
BS
,
Castro
J
, et al.
Na(V) 1.6 regulates excitability of mechanosensitive sensory neurons
.
J Physiol
.
2019
;
597
:
3751
3768
.

36

Huang
J
,
Yang
Y
,
Zhao
P
, et al.
Small-fiber neuropathy Nav1.8 mutation shifts activation to hyperpolarized potentials and increases excitability of dorsal root ganglion neurons
.
J Neurosci
.
2013
;
33
:
14087
14097
.

37

Blair
NT
,
Bean
BP
.
Roles of tetrodotoxin (TTX)-sensitive Na+ current, TTX-resistant Na+ current, and Ca2+ current in the action potentials of nociceptive sensory neurons
.
J Neurosci
.
2002
;
22
:
10277
10290
.

38

Faber
CG
,
Lauria
G
,
Merkies
ISJ
, et al.
Gain-of-function Nav1.8 mutations in painful neuropathy
.
Proc Natl Acad Sci U S A
.
2012
;
109
:
19444
19449
.

39

Flatters
SJ
,
Bennett
GJ
.
Ethosuximide reverses paclitaxel- and vincristine-induced painful peripheral neuropathy
.
Pain
.
2004
;
109
:
150
161
.

40

England
S
,
Bevan
S
,
Docherty
RJ
.
PGE2 modulates the tetrodotoxin-resistant sodium current in neonatal rat dorsal root ganglion neurones via the cyclic AMP-protein kinase A cascade
.
J Physiol
.
1996
;
495
(
Pt 2
):
429
440
.

41

Gold
MS
,
Levine
JD
,
Correa
AM
.
Modulation of TTX-R INa by PKC and PKA and their role in PGE2-induced sensitization of rat sensory neurons in vitro
.
J Neurosci
.
1998
;
18
:
10345
10355
.

42

Jin
X
,
Gereau
RW
4th
.
Acute p38-mediated modulation of tetrodotoxin-resistant sodium channels in mouse sensory neurons by tumor necrosis factor-alpha
.
J Neurosci
.
2006
;
26
:
246
255
.

43

Akin
EJ
,
Higerd-Rusli
GP
,
Mis
MA
, et al.
Building sensory axons: Delivery and distribution of Na(V)1.7 channels and effects of inflammatory mediators
.
Sci Adv
.
2019
;
5
:
eaax4755
.

44

Fukuizumi
T
,
Ohkubo
T
,
Kitamura
K
.
Spinal sensitization mechanism in vincristine-induced hyperalgesia in mice
.
Neurosci Lett
.
2003
;
343
:
89
92
.

45

Kimm
T
,
Khaliq
ZM
,
Bean
BP
.
Differential regulation of action potential shape and burst-frequency firing by BK and Kv2 channels in substantia nigra dopaminergic neurons
.
J Neurosci
.
2015
;
35
:
16404
16417
.

46

Tsantoulas
C
,
McMahon
SB
.
Opening paths to novel analgesics: The role of potassium channels in chronic pain
.
Trends Neurosci
.
2014
;
37
:
146
158
.

47

Zhang
X-L
,
Mok
L-P
,
Lee
KY
,
Charbonnet
M
,
Gold
MS
.
Inflammation-induced changes in BK(Ca) currents in cutaneous dorsal root ganglion neurons from the adult rat
.
Mol Pain
.
2012
;
8
:
37
.

48

Dib-Hajj
SD
,
Waxman
SG
.
Sodium channels in human pain disorders: Genetics and pharmacogenomics
.
Annu Rev Neurosci
.
2019
;
42
:
87
106
.

49

Joshi
SK
,
Mikusa
JP
,
Hernandez
G
, et al.
Involvement of the TTX-resistant sodium channel Nav 1.8 in inflammatory and neuropathic, but not post-operative, pain states
.
Pain
.
2006
;
123
:
75
82
.

50

Chen
L
,
Huang
J
,
Zhao
P
, et al.
Conditional knockout of Na(V)1.6 in adult mice ameliorates neuropathic pain
.
Sci Rep
.
2018
;
8
:
3845
.

51

Jones
J
,
Correll
DJ
,
Lechner
SM
, et al.
Selective inhibition of Na(V)1.8 with VX-548 for acute pain
.
N Engl J Med
.
2023
;
389
:
393
405
.

52

Johnson
JP
,
Focken
T
,
Khakh
K
, et al.
NBI-921352, a first-in-class, Na(V)1.6 selective, sodium channel inhibitor that prevents seizures in Scn8a gain-of-function mice, and wild-type mice and rats
.
Elife
.
2022
;
11
:
e72468
.

Author notes

Ana Paula Nascimento de Lima, Huiran Zhang and Lubin Chen contributed equally to this work.

This article is published and distributed under the terms of the Oxford University Press, Standard Journals Publication Model (https://dbpia.nl.go.kr/pages/standard-publication-reuse-rights)

Supplementary data