You are on page 1of 9

e80(1)

C OPYRIGHT  2021 BY T HE J OURNAL OF B ONE AND J OINT S URGERY, I NCORPORATED

Brief Electrical Stimulation Accelerates Axon


Regeneration and Promotes Recovery Following
Nerve Transection and Repair in Mice
Junichi Sayanagi, MD, Jesús A. Acevedo-Cintrón, BS, Deng Pan, BS, Lauren Schellhardt, BA, Daniel A. Hunter,
Alison K. Snyder-Warwick, MD, Susan E. Mackinnon, MD, and Matthew D. Wood, PhD
Downloaded from http://journals.lww.com/jbjsjournal by BhDMf5ePHKav1zEoum1tQfN4a+kJLhEZgbsIHo4XMi0hCywCX1AWnYQp/IlQrHD3i3D0OdRyi7TvSFl4Cf3VC4/OAVpDDa8KKGKV0Ymy+78= on 04/10/2022

Investigation performed at the Division of Plastic and Reconstructive Surgery, Department of Surgery, Washington University School of Medicine,
St. Louis, Missouri

Background: Clinical outcomes following nerve injury repair can be inadequate. Pulsed-current electrical stimulation
(ES) is a therapeutic method that facilitates functional recovery by accelerating axon regeneration. However, current
clinical ES protocols involve the application of ES for 60 minutes during surgery, which can increase operative complexity
and time. Shorter ES protocols could be a strategy to facilitate broader clinical adoption. The purpose of the present study
was to determine if a 10-minute ES protocol could improve outcomes.
Methods: C57BL/6J mice were randomized to 3 groups: no ES, 10 minutes of ES, and 60 minutes of ES. In all groups, the
sciatic nerve was transected and repaired, and, in the latter 2 groups, ES was applied after repair. Postoperatively, changes to
gene expression from dorsal root ganglia were measured after 24 hours. The number of motoneurons regenerating axons was
determined by retrograde labeling at 7 days. Histomorphological analyses of the nerve were performed at 14 days. Function
was evaluated serially with use of behavioral tests up to 56 days postoperatively, and relative muscle weight was evaluated.
Results: Compared with the no-ES group, both ES groups demonstrated increased regeneration-associated gene
expression within dorsal root ganglia. The 10-minute and 60-minute ES groups demonstrated accelerated axon regen-
eration compared with the no-ES group based on increased numbers of labeled motoneurons regenerating axons (mean
difference, 202.0 [95% confidence interval (CI), 17.5 to 386.5] and 219.4 [95% CI, 34.9 to 403.9], respectively) and
myelinated axon counts (mean difference, 559.3 [95% CI, 241.1 to 877.5] and 339.4 [95% CI, 21.2 to 657.6],
respectively). The 10-minute and 60-minute ES groups had improved behavioral recovery, including on grid-walking
analysis, compared with the no-ES group (mean difference, 11.9% [95% CI, 3.8% to 20.0%] and 10.9% [95% CI, 2.9% to
19.0%], respectively). There was no difference between the ES groups in measured outcomes.
Conclusions: A 10-minute ES protocol accelerated axon regeneration and facilitated functional recovery.
Clinical Relevance: The brief (10-minute) ES protocol provided similar benefits to the 60-minute protocol in an acute
sciatic nerve transection/repair mice model and merits further studies.

P
eripheral nerve injuries are common, affecting 2.6% of Therapeutic electrical stimulation (ES) protocols, with
patients with upper-extremity trauma and 1.2% of those ES being applied intraoperatively during nerve surgery, are a
with lower-extremity trauma1. Despite repair of nerve promising strategy. Animal data have demonstrated that the
injuries, clinical outcomes can be inadequate2-5. Slow axonal application of pulsed-current ES for 60 minutes accelerates the
growth can be a cause of inadequate recovery, and, for severe number of motor and sensory axons regenerating and reaching
axonotmetic (Sunderland III) or neurotmetic (Sunderland IV their appropriate end-organ targets6-10. ES achieves these effects
and V) nerve injuries, axon outgrowth across a repair site by augmenting early expression of regeneration-associated genes
occurs at different rates in a staggered fashion, further delaying (RAGs) within neurons, including brain-derived neurotrophic
regeneration and recovery6. Thus, strategies for accelerating factors (BDNFs)7,11-13. From these promising data, clinical trials
axonal regeneration would improve surgical management. involving the use of a 60-minute ES protocol have demonstrated

Disclosure: The Disclosure of Potential Conflicts of Interest forms are provided with the online version of the article (http://links.lww.com/JBJS/G615).

J Bone Joint Surg Am. 2020;103:e80(1-9) d http://dx.doi.org/10.2106/JBJS.20.01965


e80(2)
TH E JO U R NA L O F B O N E & JO I N T SU RG E RY J B J S . O RG
d
B R I E F E L E C T R I C A L S T I M U L AT I O N I M P R O V E S N E R V E R E PA I R
V O L U M E 103-A N U M B E R 20 O C T O B E R 20, 2 020
d d
OUTCOMES IN MICE

Fig. 1
Schematic illustrating the experimental design and the allocation of animals for the various outcome measures, with end points indicated for each metric.

improved outcomes after nerve surgery from as early as 201014, ticular nerve injury and repair model was unlikely to show dif-
yet only 3 additional trials have been performed since 201015-17. ferences between groups regarding histological findings at late end
A factor that may limit translation of ES is the current points, axon regeneration was measured at early end points as it
utilization of a 60-minute stimulation period. Care providers could detect differences in rates of regeneration25,26. Functional
will find a lengthy ES protocol during surgery to be an obstacle as recovery alone was measured to a longer end point as this longer
it can increase operative time, which negatively affects costs and end point would still capture whether accelerated axon regener-
complication risks18-21, or necessitate more complex recovery ation led to differences in recovery.
procedures to implement ES protocols17. Shorter ES protocols,
therefore, could increase translation. Surgical Procedures
In the present study, we assessed whether a shorter ES Mice were anesthetized with a cocktail of ketamine (100 mg/kg)
protocol could promote improved nerve regeneration and re- (Fort Dodge Animal Health) and dexmedetomidine (0.5 mg/kg)
covery in the context of a repaired traumatic nerve injury. We (Pfizer Animal Health). The right sciatic nerve was exposed under
hypothesized that a shorter ES protocol could improve nerve sterile conditions, transected 2 mm proximal to the sciatic tri-
regeneration and functional recovery and tested our hypothesis furcation, and repaired immediately with 11-0 nylon epineural
with use of a mouse sciatic nerve transection and repair model.

Materials and Methods


Experimental Design

A ll experiments were approved by the Washington University


in St. Louis Institutional Animal Care and Use Committee
and performed in compliance with the National Institutes of
Health guidelines. Ninety-nine male C57BL/6J mice (age, 7 weeks;
weight, 20 to 25 g) (Jackson Laboratories) were used to evaluate
the effects of ES on nerve regeneration (Fig. 1), 20 mice were used
to determine an evaluation end point for axon regeneration, and 5
mice were used as controls for gene analysis. For the primary
experiments, mice were randomized to 3 groups: (1) no ES, (2) 10
minutes of ES, and (3) 60 minutes of ES. Ten minutes was chosen
as the lower limit for ES based on previous studies22-24. Mice were
monitored daily in a temperature-controlled central animal
facility with free access to chow and water.
Previous studies assessing the effects of electrical stimula-
tion on nerve regeneration after repair showed that the primary
effect was to accelerate axon regeneration, which led to functional
recovery7,11. Therefore, we designed the study with specific end Fig. 2
points representing early and still-ongoing axon regeneration in Photograph made during the experimental procedure. The sciatic nerve
order to determine whether the application of ES accelerated axon was transected and repaired 2 mm proximal to the sciatic trifurcation. ES
regeneration compared with that after repair alone. Gene analysis was applied after repair for 10 or 60 minutes at a location 2 mm proximal to
was performed on dorsal root ganglia at 24 hours, as neuron the repair site, with a hook electrode being used to secure the nerve and a
bodies are affected by ES within the first 48 hours. As this par- return electrode placed proximal to the injury in the fascia.
e80(3)
TH E JO U R NA L O F B O N E & JO I N T SU RG E RY J B J S . O RG
d
B R I E F E L E C T R I C A L S T I M U L AT I O N I M P R O V E S N E R V E R E PA I R
V O L U M E 103-A N U M B E R 20 O C T O B E R 20, 2 020
d d
OUTCOMES IN MICE

sutures (Sharpoint). For applicable groups, ES was performed as Actb (Mm02619580_g1) (Thermo Fisher Scientific). Genetic
described previously27. A stainless-steel wire electrode (Compo- expression levels were normalized to Actb. Relative gene expression
nent Supply Company) was hooked around the sciatic nerve changes were calculated from gene expression levels obtained from
2 mm proximal to the repair site. A return electrode was placed dorsal root ganglia of uninjured mice.
into subcutaneous tissue. The electrodes were connected to the ES
device (Checkpoint Stimulator/Locator; Checkpoint Surgical). ES Retrograde Labeling of Motoneurons
was delivered for 10 or 60 minutes (16 Hz, 0.5 mA) (Fig. 2). After Retrograde labeling of motoneurons regenerating their axons was
ES, the electrodes were removed and the fascia and skin were performed on anesthetized mice in which the sciatic nerve was re-
closed with 6-0 nylon sutures (Ethicon). Postoperative pain was explored and transected 3 mm distal to the original repair site. The
managed with buprenorphine SR (sustained-release) (0.05 mg/ proximal cut end was placed in a well of petroleum jelly (Vaseline;
kg) (ZooPharm). At terminal end points, mice were killed with Unilever) filled with 4% Fluoro-Gold (Sigma-Aldrich). After 45
administration of sodium pentobarbital (>200 mg/kg). minutes, the well and Fluoro-Gold were removed, and the wounds
were closed. One week later, spinal cords were harvested and fixed
Gene Analysis (qRT-PCR) of Dorsal Root Ganglia in 4% paraformaldehyde in phosphate-buffered saline solution
Gene expression from dorsal root ganglia was assessed 24 hours (PBS) overnight, followed by immersion in 30% sucrose in PBS for
postoperatively; this end point was chosen on the basis of previ- 24 hours. Tissues were frozen in OCT Compound (VWR) and cut
ous studies11. Total RNA was prepared from combined L3 and L4 into 30-mm longitudinal sections on glass slides. The total number
dorsal root ganglia explants from each animal. RNA was extract- of Fluoro-Gold-labeled cell bodies on every section was counted
ed, measured, and reverse-transcribed to cDNA as described with use of an Olympus IX81 microscope (Olympus).
previously28. Quantitative real-time polymerase chain reaction
(qRT-PCR) was performed with use of a StepOnePlus thermo- Histomorphological Analysis
cycler (Applied Biosystems) using Taqman Master Mix (Applied Histomorphological analyses of the sciatic nerve 3 mm distal to the
Biosystems) reagents with the following primer pairs: Bdnf repair site were performed 14 days postoperatively, as described
(Mm04230607_s1), Atf3 (Mm00476033_m1), Il6 (Mm00446190_ previously29,30. Briefly, harvested sciatic nerves were fixed in 3%
m1), Csf1 (Mm00432684_m1), Ccl2 (Mm00441242_m1), and glutaraldehyde, post-fixed in 1% osmium tetroxide, dehydrated,

Fig. 3
Bar graphs illustrating that ES elevates regeneration-associated gene expression from dorsal root ganglia. Expression levels of Bdnf, Atf3, IL-6, Csf1, and Ccl2 were
measured from pooled L3-L4 dorsal root ganglia harvested at 24 hours postoperatively. Uninjured nerve dorsal root ganglia were used for comparison. All data are
expressed as the mean and the standard deviation (n = 8 for the experimental groups and n = 5 for the control group [Normal]). *P < 0.05. **P < 0.01. ***P < 0.001.
e80(4)
TH E JO U R NA L O F B O N E & JO I N T SU RG E RY J B J S . O RG
d
B R I E F E L E C T R I C A L S T I M U L AT I O N I M P R O V E S N E R V E R E PA I R
V O L U M E 103-A N U M B E R 20 O C T O B E R 20, 2 020
d d
OUTCOMES IN MICE

Fig. 4
Figs. 4-A through 4-E Illustrations depicting how ES improves motor axon regeneration. Fig. 4-A Schematic of retrograde labeling, with the Fluoro-Gold label
being applied 3 mm distal to the nerve repair site. Fig. 4-B Fluorescence photomicrograph showing representative longitudinal sections of spinal cords with Fluoro-
Gold-labeled motoneurons (yellow) for the no-ES group. Scale bars = 200 mm. Fig. 4-C Line graph showing the increase in the number of labeled motoneurons over
time in the no-ES group. Uninjured mice were used as controls (Normal group), represented as the dotted line. The data are expressed as the mean and the
standard deviation. ***P < 0.001 compared with Day 7. Fig. 4-D Fluorescence photomicrographs showing longitudinal sections of spinal cords with Fluoro-Gold-
labeled motoneurons (yellow) for all experimental groups at 7 days postoperatively. Scale bars = 200 mm. Fig. 4-E Line graph showing the number of motoneurons
at Day 7 in the no-ES group and both ES groups. The data are expressed as the mean and the standard deviation. N = 5 per group. *P < 0.05.

embedded in epoxy resin, and sectioned on an ultramicrotome to Grid-Walking Analysis


yield 1-mm cross-sections. Slides were counterstained with 1% A grid-walking test was performed twice per week from pre-
toluidine blue and analyzed at 1,000· with a Laborlux S micro- operatively (Day 0) to 56 days postoperatively28,31. Mice were
scope (Leitz). Slides were assessed with use of custom histomor- acclimated to an elevated wire mesh with a grid size measuring
phometry software (Clemex Vision Professional; Clemex 2.5 · 2.5 cm for 5 minutes before recording of the total number
Technologies). Histological measures were calculated with use of of steps of the ipsilateral hindlimb for 3 minutes. The number
>75% of the cross-sectional area of the nerve. of total steps and slipped steps missing the mesh and going
e80(5)
TH E JO U R NA L O F B O N E & JO I N T SU RG E RY J B J S . O RG
d
B R I E F E L E C T R I C A L S T I M U L AT I O N I M P R O V E S N E R V E R E PA I R
V O L U M E 103-A N U M B E R 20 O C T O B E R 20, 2 020
d d
OUTCOMES IN MICE

Fig. 5
Histomorphological sections of the sciatic nerve, made 3 mm distal to the repair site at 14 days postoperatively, illustrating that ES improved myelinated
axon regeneration. Toluidine blue staining of cross-sectional slices showed myelinated axons in all groups. Scale bars = 20 mm.

through the grid were counted, and the proportion of foot required to detect significant differences. Therefore, the group
faults was reported. size for each experiment was at least 5 mice.
For comparisons of retrograde labeling, histomorphometry,
Mechanical and Cold Sensitivity Analyses sensitivity tests, and muscle wet weight, 1-way analysis of variance
Mechanical and cold sensitivity analyses were performed at a 56- (ANOVA) followed by Tukey-Kramer post hoc testing was used. In
day end point. Mice were acclimated to an elevated meshed metal the grid-walking analysis, following 2-way repeated-measures
grid for 1 hour before measuring paw-withdrawal thresholds ANOVA, Tukey-Kramer post hoc testing was used for multiple
with use of von Frey filaments (0.02 to 2.56 g) (TouchTest; North comparisons between groups at each time point. All statistical
Coast Medical), where diminished sensation was indicated by an analyses were performed with use of JMP software (version 13; SAS
ipsilateral:contralateral ratio of <126,32. Cold sensitivity was as- Institute). The level of significance was set at p < 0.05. The data in
sessed by means of acetone-evoked evaporative cooling27. A drop the figures are expressed as the mean and the standard deviation
of acetone was released onto the plantar surface of the hindlimb, (SD), whereas the data in the Results section are expressed as the
and the time of response was measured. mean difference and the 95% confidence interval (CI).

Muscle Wet Weight Source of Funding


The tibialis anterior and gastrocnemius muscles were dissected This work was funded by an industry-sponsored research
and weighed 56 days postoperatively. The ipsilateral:contra- agreement from Checkpoint Surgical to Washington Uni-
lateral ratio was calculated. versity through one of the authors (M.D.W.).

Statistical Analysis Results


The primary outcome during experimental planning was mye- Gene Analysis of Dorsal Root Ganglia
linated axon counts26. A power analysis (alpha = 0.05, power =
0.8, effect size = 0.86) was performed considering the effect of ES
on regenerated myelinated axon counts27 with use of G*power
D orsal root ganglia harvested at 24 hours from all 3 groups
revealed that nerve injury elevated the expression of
regeneration-associated genes, Bdnf and Atf3, as well as inflam-
3.1.9.7 (Heinrich Heine University). Five mice per group were matory genes, Il6, Csf1, and Ccl2 (Fig. 3). However, both the

TABLE I Histomorphometric Data of Distal Repaired Sciatic Nerves at 14 Days Postoperatively*

Outcome Metric Uninjured No ES 10-Minute ES 60-Minute ES

Myelinated axon count (no.) 4,521 ± 468 496 ± 218 1,055 ± 378§ 835 ± 239‡
Myelinated axon density (axons/mm2) 29,822 ± 2,685 3,133 ± 1,377 6,344 ± 3,024† 5,531 ± 2,029
Percent nerve area (%) 72.14 ± 2.04 3.22 ± 1.62 7.13 ± 3.65† 5.15 ± 1.89
Fiber width (mm) 4.78 ± 0.24 3.21 ± 0.41 3.32 ± 0.14 3.02 ± 0.22
G-ratio 0.60 ± 0.02 0.51 ± 0.06 0.49 ± 0.03 0.54 ± 0.03

*All data are expressed as the mean and the standard deviation (n = 10 for each group). ‡p < 0.05, †p < 0.01, §p < 0.001 compared with the no-ES
group (1-way ANOVA followed by a post hoc Tukey-Kramer test).
e80(6)
TH E JO U R NA L O F B O N E & JO I N T SU RG E RY J B J S . O RG
d
B R I E F E L E C T R I C A L S T I M U L AT I O N I M P R O V E S N E R V E R E PA I R
V O L U M E 103-A N U M B E R 20 O C T O B E R 20, 2 020
d d
OUTCOMES IN MICE

to 4.28]) were significantly higher than that in the no-ES group.


Gene expression levels were not significantly different between
the 10-minute-ES and 60-minute-ES groups.

Motor Nerve Regeneration


Retrograde labeling of motoneurons regenerating their axons
from the repair site without ES revealed staggered axon regen-
eration. There were significantly greater numbers of labeled
motoneurons 14 days compared with 7 days postoperatively
(mean difference, 355.4 [95% CI, 217.7 to 493.1]) (Figs. 4-A, 4-B,
and 4-C), but there were no differences after 14 days. Based
on these results, we assessed the number of motoneurons re-
generating axons in the ES groups at 7 days postoperatively. The
numbers of labeled motoneurons in the 10-minute-ES and 60-
minute ES groups were significantly greater than those in the no-
ES group (mean difference, 202.0 [95% CI, 17.5 to 386.5] and
219.4 [95% CI, 34.9 to 403.9], respectively). No significant dif-
ference was observed between the 10-minute-ES and 60-minute-
ES groups (Figs. 4-D and 4-E).

Histomorphological Analysis
Histomorphological analyses of the sciatic nerve 3 mm distal to
the repair site at 14 days revealed ongoing axon regeneration, as
normal sciatic nerve contains ;4,500 myelinated axons. Mye-
linated axon regeneration in the ES groups was increased com-
pared with the no-ES group (Fig. 5, Table I). The 10-minute-ES
and 60-minute-ES groups had significantly greater myelinated
axon counts than the no-ES group (mean difference, 559.3 [95%
Fig. 6 CI, 241.1 to 877.5] and 339.4 [95% CI, 21.2 to 657.6], respec-
Figs. 6-A through 6-D Illustrations showing that ES improved motor tively). Myelinated axon density and percent nerve area in the
recovery. Foot faults represent steps that miss the grid rungs and enter the 10-minute-ES group were significantly higher than those in the
grid pattern. Preoperative values are represented at Day 0. Fig. 6-A no-ES group (mean difference, 3,211.3 axons/mm2 [95% CI,
Schematic of the grid-walking test, which involves motor coordination as 718.7 to 5,703.9 axons/mm2] and 3.91% [95% CI, 1.08% to
well as proprioceptive and other sensory feedback. Figs. 6-B, 6-C, and 6-D 6.74%], respectively). No significant differences were observed
Quantification of foot faults (proportion of foot fault steps to total steps for among the 3 groups in terms of fiber width and g-ratio.
the affected limb). All data are expressed as the mean and the standard
deviation (n = 10 per group). Fig. 6-B †P < 0.05 between Day 7 and all time
points from Day 28 onward in the no-ES group.^P < 0.05 between Day 7 and
all time points from Day 17 onward in the 10-minute-ES group. ‡P < 0.05
between Day 7 and all time points from Day 17 onward in the 60-minute-ES
group. #P < 0.05 between the no-ES group and both ES groups from Day 17
onward. The arrows indicate that significant differences continued until
56 days postoperatively. Figs. 6-C and 6-D Bar graphs showing the
quantification of foot faults in the no-ES and ES groups at Days 17 and 56,
respectively. *P < 0.05. **P < 0.01.
Fig. 7

10-minute-ES and 60-minute-ES groups revealed significantly Figs. 7-A and 7-B Bar graphs illustrating that ES improved mechanical
elevated expression of Bdnf (mean difference, 1.04 [95% CI, 0.44 sensitivity but not cold sensitivity. All data are expressed as the mean and the
to 1.64] and 1.54 [95% CI, 0.94 to 2.13], respectively), Atf3 (mean standard deviation (n = 5 per group). **P < 0.01. Fig. 7-A Mechanical
difference, 22.9 [95% CI, 3.6 to 42.1] and 32.3 [95% CI, 13.0 to sensitivity of the affected foot based on von Frey monofilament tests at
51.6], respectively), and Il6 (mean difference, 13.4 [95% CI, 4.5 to 56 days postoperatively. In uninjured nerve (dashed line), there is no ipsi-
22.2] and 11.1 [95% CI, 2.3 to 19.9], respectively), in comparison lateral/contralateral difference in the paw-withdrawal threshold ratio (ratio =
with the no-ES group. Expression levels of Csf1 in the 60-minute- ;1). Fig. 7-B Cold allodynia responses to the application of cold acetone to
ES group (mean difference, 1.78 [95% CI, 0.83 to 2.72]) and Ccl2 the affected foot, measured at 56 days postoperatively. Mean data for the
in the 10-minute-ES group (mean difference, 2.38 [95% CI, 0.47 uninjured (contralateral) nerve (n = 15) are represented by the dashed line.
e80(7)
TH E JO U R NA L O F B O N E & JO I N T SU RG E RY J B J S . O RG
d
B R I E F E L E C T R I C A L S T I M U L AT I O N I M P R O V E S N E R V E R E PA I R
V O L U M E 103-A N U M B E R 20 O C T O B E R 20, 2 020
d d
OUTCOMES IN MICE

Muscle Wet Weight


The relative gastrocnemius muscle weight was significantly
greater for the 10-minute-ES and 60-minute-ES groups as
compared with the no-ES group (mean difference, 0.08 [95%
CI, 0.01 to 0.16] and 0.10, [95% CI, 0.03 to 0.18]). Similarly,
the relative tibialis anterior muscle weight was significantly
greater for the 10-minute-ES and 60-minute-ES groups as
compared with the no-ES group (mean difference, 0.16 [95%
CI, 0.07 to 0.24] and 0.14 [95% CI, 0.05 to 0.22] respectively)
Fig. 8 (Fig. 8).
Figs. 8-A and 8-B Bar graphs illustrating that ES increased muscle recovery as
expressed as relative muscle weight (i.e., the ratio of affected muscle weight Discussion
to the contralateral muscle weight). All data are expressed as the mean and
the standard deviation (n = 10 per group). *P < 0.05. **P < 0.01. ***P <
0.001. Fig. 8-A Gastrocnemius muscle. Fig. 8-B Tibialis anterior muscle.
T he present study demonstrated that both the 10-minute and
60-minute ES protocols accelerated axon regeneration,
thereby promoting improved functional recovery, compared
with that in the no-ES group. Furthermore, there were no
Grid-Walking Test obvious differences between the ES groups in any measured
All animals demonstrated few foot faults prior to injury as outcome.
measured on Day 0 but then showed impaired function based To our knowledge, only 4 clinical trials have investigated
on increased foot faults after the nerve injury (Figs. 6-A and 6- the use of ES as an intervention in nerve surgery since 2010, all
B). Recovery, while incomplete, was appreciated in all animals of which utilized a 60-minute ES protocol14-17, which was re-
during the observation period following surgery. Compared viewed in detail by Ransom et al33. Those results suggested a
with Day 7, the 10-minute-ES and 60-minute-ES groups slow adoption of ES in the clinic, which could be improved by
demonstrated significantly fewer foot faults from Day 17 simplifying the application of ES to the treatment of nerve
onward (mean difference, 14.8% [95% CI, 5.6% to 24.0%] and injuries. The present study is the first to demonstrate that ES
13.3% [95% CI, 4.3% to 22.3%], respectively). In contrast, the protocols of <60 minutes can be effective for promoting
no-ES group demonstrated significantly fewer foot faults from regeneration and recovery22-24,34,35. A 10-minute ES protocol
Day 28 onward (mean difference, 13.1% [95% CI, 2.5% to involving the use of direct current has been previously shown
23.7%]) (Fig. 6-B). By Day 17, the 10-minute-ES and 60- to promote improved axon regeneration in vitro and in vivo22-24.
minute-ES groups demonstrated significantly fewer foot faults The present study was different from those studies in that we
(mean difference, 10.5% [95% CI, 1.1% to 19.9%] and 10.9% considered a 10-minute pulsed-current ES protocol using a
[95% CI, 1.5% to 20.3%]) compared with the no-ES group clinically available nerve stimulator and compared the results
(Fig. 6-C). At Day 56, this improved functional recovery based with the more common 60-minute ES protocol.
on foot faults was maintained, with the 10-minute-ES and 60- The findings observed in association with the 10-
minute-ES groups being significantly different (mean differ- minute ES protocol are supported by previous literature6-10.
ence, 11.9% [95% CI, 3.8% to 20.0%] and 10.9% [95% CI, Pulsed-current ES protocols applied for 60 minutes increase
2.9% to 19.0%], respectively) as compared with the no-ES regeneration-associated gene expression earlier than that after
group (Fig. 6-D). There was no significant difference between injury alone11 and accelerate the rate at which axons cross a
the 2 ES groups on the grid-walking test. nerve repair site6,7, similar to our observations. These out-
comes have been associated with improved functional
Mechanical and Cold Sensitivity recovery6,7. Therefore, our findings suggest that ES facilitating
Prior to nerve injury, the groups demonstrated no ipsilateral/ earlier axon regeneration results in axons reaching end-organ
contralateral difference in terms of the paw-withdrawal thresh- targets earlier and thereby improves functional recovery.
old on mechanical sensitivity analysis (ratio, ;1). After nerve The mechanism promoting the regenerative effects for
injury, the groups demonstrated decreased mechanical sensi- the 10-minute ES protocol is unclear. The present study sug-
tivity (ratio, >1). Fifty-six days postoperatively, the 10-minute- gests that regeneration was improved via a similar mechanism
ES and 60-minute-ES groups demonstrated significant differ- as that associated with 60-minute ES, with ES-generated action
ences in the ipsilateral/contralateral ratio of the paw-withdrawal potentials leading to increased expression of growth-associated
threshold as compared with the no-ES group (mean difference, genes and, in turn, accelerated axon regeneration11,13,36. While
2.21 [95% CI, 0.90 to 3.52] and 2.16 [95% CI, 0.85 to 3.48], this direct connection cannot yet be proven, studies applying
respectively) (Fig. 7-A). optogenetics have offered insights. In optogenetic studies, the
Prior to nerve injury, all groups had some sensitivity to number of pulses generating action potentials, rather than
cold acetone (;3.2 seconds of response). After nerve injury, duration, was the predominant cue for promoting axon growth
the groups demonstrated increased cold sensitivity. No signif- from neurons37. Optical stimulation of neurons generating far
icant differences among the groups in terms of cold sensitivity fewer pulses than 1 hour at 20 Hz still resulted in increased
were found by Day 56 (Fig. 7-B). axon growth in vitro compared with no pulses37. Therefore, a
e80(8)
TH E JO U R NA L O F B O N E & JO I N T SU RG E RY J B J S . O RG
d
B R I E F E L E C T R I C A L S T I M U L AT I O N I M P R O V E S N E R V E R E PA I R
V O L U M E 103-A N U M B E R 20 O C T O B E R 20, 2 020
d d
OUTCOMES IN MICE

10-minute ES protocol may stimulate sufficient evoked-action may result in a larger zone of injury or delayed repair to deter-
potentials to elicit neurons to enhance axon growth. mine if this ES protocol could still provide therapeutic benefit.
Our results also suggest that ES could involve non- In conclusion, a brief 10-minute ES protocol promoted
neuronal cells that may play a role in nerve regeneration, as nerve regeneration and functional recovery, similar to a 60-
inflammatory genes became elevated relative to the no-ES minute ES protocol. Brief (10-minute) ES protocols merit further
group. ES therapies are associated with an increase in macro- studies. n
phage accumulation38,39, and increased macrophage accumu-
lation and the responses of the macrophages can promote
nerve regeneration40,41. However, the current study is limited
in terms of our ability to draw conclusions as the histomor-
phometry techniques that were utilized to describe nerve
Junichi Sayanagi, MD1
regeneration could not quantify inflammatory cells. In addi-
Jesús A. Acevedo-Cintrón, BS1
tion, the assessors of the histomorphometry were not blinded Deng Pan, BS1
to treatment group, representing a potential source of Lauren Schellhardt, BA1
detection bias. Daniel A. Hunter1
The present study had several additional limitations. Only Alison K. Snyder-Warwick, MD1
male sex was considered, and future studies will need to consider Susan E. Mackinnon, MD1
any sex effects due to ES. Additionally, the animal injury and Matthew D. Wood, PhD1
repair model represents a “best-case scenario” in terms of re- 1Divisionof Plastic and Reconstructive Surgery, Department of Surgery,
generation as nerve transection followed by immediate repair Washington University School of Medicine, St. Louis, Missouri
and then application of ES is ideal. Future studies would benefit
from considering clinical scenarios, such as blunt trauma, which Email for corresponding author: woodmd@wustl.edu

References
1. Padovano WM, Dengler J, Patterson MM, Yee A, Snyder-Warwick AK, Wood MD, affecting the functional measures in carpal tunnel syndrome patients. Exp Neurol.
Moore AM, Mackinnon SE. Incidence of Nerve Injury After Extremity Trauma in the 2010 May;223(1):192-202. Epub 2009 Oct 1.
United States. Hand (N Y). 2020 Oct 21:1558944720963895. Epub 2020 Oct 21. 15. Wong JN, Olson JL, Morhart MJ, Chan KM. Electrical stimulation enhances
2. Ruijs ACJ, Jaquet JB, Kalmijn S, Giele H, Hovius SER. Median and ulnar nerve sensory recovery: a randomized controlled trial. Ann Neurol. 2015 Jun;77(6):
injuries: a meta-analysis of predictors of motor and sensory recovery after modern 996-1006. Epub 2015 May 4.
microsurgical nerve repair. Plast Reconstr Surg. 2005 Aug;116(2):484-94, discus- 16. Barber B, McNeely M, Chan KM, Beaudry R, Olson J, Harris J, Seikaly H,
sion 495-6. O’Connell D. Intraoperative brief electrical stimulation (BES) for prevention of
3. Grinsell D, Keating CP. Peripheral nerve reconstruction after injury: a review of shoulder dysfunction after oncologic neck dissection: study protocol for a random-
clinical and experimental therapies. Biomed Res Int. 2014;2014:698256. Epub ized controlled trial. Trials. 2015 May 30;16:240.
2014 Sep 3. 17. Power HA, Morhart MJ, Olson JL, Chan KM. Postsurgical Electrical Stimulation
4. Karsy M, Watkins R, Jensen MR, Guan J, Brock AA, Mahan MA. Trends and Cost Enhances Recovery Following Surgery for Severe Cubital Tunnel Syndrome: A Double-
Analysis of Upper Extremity Nerve Injury Using the National (Nationwide) Inpatient Blind Randomized Controlled Trial. Neurosurgery. 2020 Jun 1;86(6):769-77.
Sample. World Neurosurg. 2019 Mar;123:e488-500. Epub 2018 Nov 28. 18. Macario A. What does one minute of operating room time cost? J Clin Anesth.
5. Foster CH, Karsy M, Jensen MR, Guan J, Eli I, Mahan MA. Trends and Cost- 2010 Jun;22(4):233-6.
Analysis of Lower Extremity Nerve Injury Using the National Inpatient Sample. Neu- 19. Hu K, Zhang T, Hutter M, Xu W, Williams Z. Thirty-Day Perioperative Adverse
rosurgery. 2019 Aug 1;85(2):250-6. Outcomes After Peripheral Nerve Surgery: An Analysis of 2351 Patients in the
6. Brushart TM, Hoffman PN, Royall RM, Murinson BB, Witzel C, Gordon T. Electrical American College of Surgeons National Surgical Quality Improvement Program
stimulation promotes motoneuron regeneration without increasing its speed or Database. World Neurosurg. 2016 Oct;94:409-17. Epub 2016 Jul 17.
conditioning the neuron. J Neurosci. 2002 Aug 1;22(15):6631-8. 20. Martin E, Muskens IS, Senders JT, Cote DJ, Smith TR, Broekman MLD. A
7. Al-Majed AA, Neumann CM, Brushart TM, Gordon T. Brief electrical stimulation nationwide analysis of 30-day adverse events, unplanned readmission, and length of
promotes the speed and accuracy of motor axonal regeneration. J Neurosci. 2000 hospital stay after peripheral nerve surgery in extremities and the brachial plexus.
Apr 1;20(7):2602-8. Microsurgery. 2019 Feb;39(2):115-23. Epub 2018 Apr 15.
8. Brushart TM, Jari R, Verge V, Rohde C, Gordon T. Electrical stimulation 21. Childers CP, Maggard-Gibbons M. Understanding costs of care in the operating
restores the specificity of sensory axon regeneration. Exp Neurol. 2005 Jul; room. JAMA Surg. 2018 Apr 18;153(4):e176233. Epub 2018 Apr 18.
194(1):221-9. 22. Wood M, Willits RK. Short-duration, DC electrical stimulation increases chick
9. Geremia NM, Gordon T, Brushart TM, Al-Majed AA, Verge VMK. Electrical stimu- embryo DRG neurite outgrowth. Bioelectromagnetics. 2006 May;27(4):328-31.
lation promotes sensory neuron regeneration and growth-associated gene expres- 23. Wood MD, Willits RK. Applied electric field enhances DRG neurite growth:
sion. Exp Neurol. 2007 Jun;205(2):347-59. Epub 2007 Feb 21. influence of stimulation media, surface coating and growth supplements. J Neural
10. Gordon T, Chan KM, Sulaiman OAR, Udina E, Amirjani N, Brushart TM. Accel- Eng. 2009 Aug;6(4):046003. Epub 2009 Jun 3.
erating axon growth to overcome limitations in functional recovery after peripheral 24. Calvey C, Zhou W, Stakleff KS, Sendelbach-Sloan P, Harkins AB, Lanzinger
nerve injury. Neurosurgery. 2009 Oct;65(4)(Suppl):A132-44. W, Willits RK. Short-term electrical stimulation to promote nerve repair and
11. Al-Majed AA, Brushart TM, Gordon T. Electrical stimulation accelerates and functional recovery in a rat model. J Hand Surg Am. 2015 Feb;40(2):314-22.
increases expression of BDNF and trkB mRNA in regenerating rat femoral moto- Epub 2014 Nov 11.
neurons. Eur J Neurosci. 2000 Dec;12(12):4381-90. 25. Brenner MJ, Moradzadeh A, Myckatyn TM, Tung TH, Mendez AB, Hunter DA,
12. Al-Majed AA, Tam SL, Gordon T. Electrical stimulation accelerates and Mackinnon SE. Role of timing in assessment of nerve regeneration. Microsurgery.
enhances expression of regeneration-associated genes in regenerating rat femoral 2008;28(4):265-72.
motoneurons. Cell Mol Neurobiol. 2004 Jun;24(3):379-402. 26. Wood MD, Kemp SWP, Weber C, Borschel GH, Gordon T. Outcome measures
13. Zuo KJ, Gordon T, Chan KM, Borschel GH. Electrical stimulation to enhance of peripheral nerve regeneration. Ann Anat. 2011 Jul;193(4):321-33. Epub 2011 May 13.
peripheral nerve regeneration: Update in molecular investigations and clinical 27. Jo S, Pan D, Halevi AE, Roh J, Schellhardt L, Hunter Ra DA, Snyder-Warwick AK,
translation. Exp Neurol. 2020 Oct;332:113397. Epub 2020 Jul 3. Moore AM, Mackinnon SE, Wood MD. Comparing electrical stimulation and tacroli-
14. Gordon T, Amirjani N, Edwards DC, Chan KM. Brief post-surgical electrical mus (FK506) to enhance treating nerve injuries. Muscle Nerve. 2019 Nov;60(5):
stimulation accelerates axon regeneration and muscle reinnervation without 629-36. Epub 2019 Aug 21.
e80(9)
TH E JO U R NA L O F B O N E & JO I N T SU RG E RY J B J S . O RG
d
B R I E F E L E C T R I C A L S T I M U L AT I O N I M P R O V E S N E R V E R E PA I R
V O L U M E 103-A N U M B E R 20 O C T O B E R 20, 2 020
d d
OUTCOMES IN MICE

28. Pan D, Hunter DA, Schellhardt L, Fuchs A, Halevi AE, Snyder-Warwick AK, 35. Alrashdan MS, Park JC, Sung MA, Yoo SB, Jahng JW, Lee TH, Kim SJ, Lee JH.
Mackinnon SE, Wood MD. T cells modulate IL-4 expression by eosinophil recruitment Thirty minutes of low intensity electrical stimulation promotes nerve regeneration
within decellularized scaffolds to repair nerve defects. Acta Biomater. 2020 Aug; after sciatic nerve crush injury in a rat model. Acta Neurol Belg. 2010 Jun;
112:149-63. Epub 2020 May 17. 110(2):168-79.
29. Hunter DA, Moradzadeh A, Whitlock EL, Brenner MJ, Myckatyn TM, Wei CH, Tung 36. Willand MP, Nguyen MA, Borschel GH, Gordon T. Electrical Stimulation to Pro-
TH, Mackinnon SE. Binary imaging analysis for comprehensive quantitative histo- mote Peripheral Nerve Regeneration. Neurorehabil Neural Repair. 2016 Jun;30(5):
morphometry of peripheral nerve. J Neurosci Methods. 2007 Oct 15;166(1):116-24. 490-6. Epub 2015 Sep 10.
Epub 2007 Jun 30. 37. Park S, Koppes RA, Froriep UP, Jia X, Achyuta AK, McLaughlin BL, Anikeeva P.
30. Hunter DA, Pan D, Wood MD, Snyder-Warwick AK, Moore AM, Feldman EL, Optogenetic control of nerve growth. Sci Rep. 2015 May 18;5:9669.
Mackinnon SE, Brenner MJ. Design-Based stereology and binary image histomor- 38. Keane GC, Pan D, Roh J, Larson EL, Schellhardt L, Hunter DA, Snyder-Warwick
phometry in nerve assessment. J Neurosci Methods. 2020 Apr 15;336:108635. AK, Moore AM, Mackinnon SE, Wood MD. The Effects of Intraoperative Electrical
Epub 2020 Feb 15. Stimulation on Regeneration and Recovery After Nerve Isograft Repair in a Rat
31. Pan D, Acevedo-Cintrón JA, Sayanagi J, Snyder-Warwick AK, Mackinnon SE, Model. Hand (N Y). 2020 Jul 15:1558944720939200. Epub 2020 Jul 15.
Wood MD. The CCL2/CCR2 axis is critical to recruiting macrophages into acellular 39. McLean NA, Verge VMK. Dynamic impact of brief electrical nerve stimulation
nerve allograft bridging a nerve gap to promote angiogenesis and regeneration. Exp on the neural immune axis-polarization of macrophages toward a pro-repair pheno-
Neurol. 2020 Sep;331:113363. Epub 2020 May 23. type in demyelinated peripheral nerve. Glia. 2016 Sep;64(9):1546-61. Epub 2016
32. Dixon WJ. Efficient analysis of experimental observations. Annu Rev Pharmacol Jun 29.
Toxicol. 1980;20:441-62. 40. Bombeiro AL, Santini JC, Thomé R, Ferreira ER, Nunes SL, Moreira BM, Bonet IJ,
33. Ransom SC, Shahrestani S, Lien BV, Tafreshi AR, Brown NJ, Hanst B, Lehrich Sartori CR, Verinaud L, Oliveira AL. Enhanced immune response in immunodeficient
BM, Ransom RC, Sahyouni R. Translational Approaches to Electrical Stimulation for mice improves peripheral nerve regeneration following axotomy. Front Cell Neurosci.
Peripheral Nerve Regeneration. Neurorehabil Neural Repair. 2020 Nov;34(11): 2016 Jun 14;10:151.
979-85. Epub 2020 Oct 10. 41. Bombeiro AL, Pereira BTN, de Oliveira ALR. Granulocyte-macrophage colony-
34. Nix WA, Hopf HC. Electrical stimulation of regenerating nerve and its effect on stimulating factor improves mouse peripheral nerve regeneration following sciatic
motor recovery. Brain Res. 1983 Aug 1;272(1):21-5. nerve crush. Eur J Neurosci. 2018 Sep;48(5):2152-64. Epub 2018 Aug 29.

You might also like