You are on page 1of 16

| |

Received: 15 January 2019    Revised: 30 August 2019    Accepted: 16 September 2019

DOI: 10.1002/jnr.24538

REVIEW

Peripheral nerve injury and myelination: Potential


therapeutic strategies

Max Modrak1 | M. A. Hassan Talukder2 | Khatuna Gurgenashvili3 |


Mark Noble4 | John C. Elfar2

1
School of Medicine & Dentistry, The
University of Rochester Medical Center, Abstract
Rochester, New York, USA Traumatic peripheral nerve injury represents a major clinical and public health prob‐
2
Department of Orthopaedics &
lem that often leads to significant functional impairment and permanent disability.
Rehabilitation, Penn State Hershey College
of Medicine, Milton S. Hershey Medical Despite modern diagnostic procedures and advanced microsurgical techniques,
Center, Hershey, Pennsylvania, USA
functional recovery after peripheral nerve repair is often unsatisfactory. Therefore,
3
Department of Neurology, Penn State
Hershey College of Medicine, Milton
there is an unmet need for new therapeutic or adjunctive strategies to promote the
S. Hershey Medical Center, Hershey, functional recovery in nerve injury patients. In contrast to the central nervous sys‐
Pennsylvania, USA
4
tem, Schwann cells in the peripheral nervous system play a pivotal role in several
Department of Biomedical Genetics, The
University of Rochester Medical Center, aspects of nerve repair such as degeneration, remyelination, and axonal growth.
Rochester, New York, USA Several non‐surgical approaches, including pharmacological, electrical, cell‐based,
Correspondence and laser therapies, have been employed to promote myelination and enhance func‐
John C. Elfar and M. A. Hassan Talukder, tional recovery after peripheral nerve injury. This review will succinctly discuss the
Department of Orthopaedics and
Rehabilitation, Penn State Hershey College of potential therapeutic strategies in the context of myelination following peripheral
Medicine, Milton S. Hershey Medical Center, neurotrauma.
Mail Code H089, 500 University Drive, P.O.
Box‐850, Hershey, PA 17033, USA.
Email: openelfar@gmail.com (J. C. E.) and KEYWORDS
mahassantalukder@yahoo.com (M. A. H. T) epigenetics, limitations, myelination, peripheral nerve injury, pharmacotherapy, physical
therapy, therapeutic strategies
Funding information
U.S. Department of Defense, Grant/Award
Number: W81XWH‐16‐1‐0725; National
Institute of Health, Grant/Award Number:
K08 AR060164‐01A

1 |  I NTRO D U C TI O N impairment and permanent disability (Robinson, 2000a). It is estimated


that roughly 3% of all trauma patients have peripheral nerve injuries
Traumatic peripheral nerve injury (TPNI) represents a major clinical (Taylor, Braza, Rice, & Dillingham, 2008) and more than 50,000 pe‐
and public health problem that often leads to significant functional ripheral nerve repair procedures are performed annually in the United

Abbreviations: 4‐AP, 4‐aminopyridine; ADSCs, adipose‐derived stem cells; BDNF, brain‐derived neurotrophic factor; cAMP, cyclic adenosine monophosphate; CMAP, compound muscle
action potential; CNS, central nervous system; DVT, deep vein thrombosis; EDX, electrodiagnostic testing; EMG, electromyography; EPO, erythropoietin; EPOR, erythropoietin
receptor; ES, electrical stimulation; ESCs, embryonic stem cells; HDAC, histone deacetylase; hPSCs, human induced pluripotent stem cells; Krox20, early growth response gene (Egr2);
LLLT, low‐level laser therapy; MPZ, myelin protein zero; MSCs, mesenchymal stem cells; MUAP, motor unit action potential; NAB, NGFI‐A/Egr‐binding protein; NCS, nerve conduction
study; NCV, nerve conduction velocity; Oct6, octamer‐binding transcription factor‐6; PBM, Photobiomodulation; PMP22, peripheral myelin protein 22; PNS, peripheral nervous system;
SKPCs, skin‐derived precursor cells; Sox 10, SRY‐related HMG‐box‐10; TPNI, traumatic peripheral nerve injury; WHI, Women's Health Initiative.

Max Modrak and M. A. Hassan Talukder contributed equally to this study.

Edited by Stephen Crocker. Reviewed by David B. Parkinson.

All peer review communications can be found with the online version of the article.

The peer review history for this article is available at https​://publo​ns.com/publo​n/10.1002/jnr.24538​

J Neuro Res. 2019;00:1–16. © 2019 Wiley Periodicals, Inc. |  1


wileyonlinelibrary.com/journal/jnr  
|
2       MODRAK et al.

with fewer and larger fascicles may be more vulnerable to injury as


Significance a result of their inherent deficiency in connective epineurial tissue
Traumatic peripheral nerve injury (TPNI) represents a major (Sunderland, 1951, 1990). Some fibers within any given fascicle con‐
health problem that often leads to significant functional im‐ tain myelin which is compact in distribution around internodes, in con‐
pairment and permanent disability. Despite advanced micro‐ trast to the less myelin‐dense paranodal and myelin‐deficient nodes of
surgical nerve repair techniques, most patients with TPNI Ranvier, where sodium channels are concentrated allowing propaga‐
do not attain full functional recovery and there is no reliable tion of action potentials (saltatory conduction). In practice, injuries to
medical treatment available. Therefore, there is an unmet connective tissue, myelin, and axons all comingle in most TPNI.
need for new therapeutic strategies to promote the func‐ The two commonly used classifications of nerve injury are the Seddon
tional recovery in patients with TPNI. Several non‐surgical and Sunderland Classifications (Kaya & Sarikcioglu, 2015; Sunderland,
approaches, including pharmacological, electrical, cell‐based, 1951). Seddon's scheme, first published in 1943, divides nerve injuries
and laser therapies, have been employed to promote myelina‐ into neurapraxia, axonotmesis, and neurotmesis in order of increasing
tion and enhance functional recovery after TPNI. This review severity. Neurapraxia, the mildest class of nerve injury, is characterized
succinctly discusses the benefits and limitations of potential as a conduction block from a seemingly isolated injury to myelin, usu‐
therapeutic strategies in TPNIs. ally along a particular section of the nerve (segmental demyelination).
In practice, myelin is not likely the only tissue type to be disrupted in
these injuries, as nerve dysfunction from focal ischemia and subtle inju‐
States alone (Evans, 2001). TPNIs are generally associated with motor
ries to other tissue types may also play a role. Nonetheless, neuropraxic
vehicle collisions, penetrating injuries, lacerations, gun‐shots, falls,
injuries are believed to spare most, if not all axons and these injuries
burns, fractures, ischemia, traction and crush injuries (Campbell, 2008),
often result from compression or traction on the nerve. In axonotmesis,
and have been studied in both civilian (Noble, Munro, Prasad, & Midha,
the hallmark is axonal disruption and resulting Wallerian degeneration,
1998; Robinson, 2004; Sullivan, Dailey, Duncan, Abel, & Borlongan,
but some connective tissue structures are preserved. Axonotmesis is
2016; Taylor et al., 2008) and military populations (Birch et al., 2012).
typical after peripheral nerve crush and stretch injuries. Neurotmesis,
TPNIs are also more common in children than previously identified and
where the nerve is severed, is the most severe class of nerve injury and is
can result in major social and economic burdens (Missios, Bekelis, &
often caused by substantial trauma to the nerve and adjoining structures
Spinner, 2014). Despite progress in understanding the pathophysiol‐
(Kaya & Sarikcioglu, 2015; Sunderland, 1951, 1990).
ogy and the biological factors involved in of nerve injury and recovery,
Sunderland's classification breaks down nerve injury into five cate‐
as well as the availability of modern diagnostic procedures and ad‐
gories: (Sunderland, 1990) the first degree of nerve injury corresponds
vanced microsurgical techniques, few treatment options exist to alter
to neurapraxia, here with the distinction that the distal segment of
the course of recovery after TPNI (Campbell, 2008; Menorca, Fussell,
the nerve, which is outside of the zone of injury, can conduct nor‐
& Elfar, 2013). This review will discuss the current knowledge of TPNI
mally. Here too, the axonal structure is believed to be preserved. The
and recovery in terms of pathophysiology of nerve injury, classifica‐
second degree of nerve injury consists of axon degeneration distally
tion, regeneration, diagnostic consideration, and therapeutic strate‐
with preserved endoneurial tubes with a good chance of near nor‐
gies. The main focus is to provide a better understanding of potential
mal recovery if the injuries are not too distant from the targets. The
non‐surgical therapeutic approaches in promoting myelination follow‐
third degree of nerve injury consists of the destruction of endoneur‐
ing peripheral neurotrauma.
ial tubes and nerve fibers with preserved fascicles and perineurium.
Intrafascicular structures are disorganized but fascicular continuity is
preserved. The fourth degree of nerve injury is associated with fascic‐
2 |  PATH O PH YS I O LO G Y A N D
ular and perineurial destruction with only epineurium spared. The fifth
C L A S S I FI C ATI O N O F N E RV E I N J U RY
degree of nerve injury is of complete loss of nerve continuity and com‐
plete destruction of axon and surrounding connective tissue, which
At the cellular level, TPNIs exist in varying severities, which relate di‐
corresponds to neurotmesis in Seddon's classification. In neurotmesis
rectly to the classification of these injuries. The severity of injury is
the prognosis of spontaneous recovery is poor, and nerve continuity
related to the extent of anatomic disruption to the constituents of the
is believed unlikely or even impossible without surgical intervention.
nerve. On an anatomic level, the nerves grossly consist of axons, my‐
elin (in myelinated fibers), Schwann cells, blood vessels, fat and con‐
nective tissue (Menorca et al., 2013; Sunderland, 1951, 1990). The
3 | R EG E N E R ATI O N
outer covering of the nerve is a connective tissue epineurium, which is
the site of most microsurgical techniques for repair. Within the nerve
Recovery of nerve function after injury depends on several factors in‐
there are fascicles embedded in an internal connective tissue epineu‐
cluding the degree and location of the nerve injury and patient‐related
rium. Each individual fascicle is surrounded by perineurial connective
factors. The degree of nerve injury itself depends on the number and
tissue, and each fascicle contains many nerve fibers, which in turn are
size of fascicles within the injured nerve (e.g., partial vs. complete sev‐
encapsulated by longitudinally oriented endoneurial tubes. Nerves
erance of fascicles). Not all nerves have the same capacity for recovery,
MODRAK et al. |
      3

and therefore the type and location of nerve injured (e.g., proximal vs. increasing disorganization of fascicular structure and scar formation
distal nerve segment, mixed nerve, etc.) are critical prognostic factors inside the nerve. This is perhaps related to the random and disorga‐
for regeneration (Campbell, 2008; Miller, 1987; Post, Boer, & Malessy, nized growth of regenerating axons toward the end organ (Robinson,
2012; Sunderland, 1990). Finally, advanced patient age and the chro‐ 2000a; Sunderland, 1990). This may predispose more severe injuries to
nicity of the injury each negatively affect the capacity for regeneration incomplete recovery of muscle function. Endoneurial tubes, Schwann
with older patients and those with delayed presentations or interven‐ cells, and muscle fibers may indeed be viable for 12–18 months after
tion often faring most poorly. Regeneration and repair processes of prolonged axotomy (Campbell, 2008; Fu & Gordon, 1995; Robinson,
injured nerve occur at multiple levels including the nerve cell body, 2000a); however, motor recovery is rarely possible after this time pe‐
the segment between the neuron and the injury site (proximal stump), riod even with surgical repair because of reduced capacity of motor
the injury site itself, the segment between the injury site and the end axons to regenerate (Sulaiman & Gordon, 2013). Sensory fibers, with‐
organ (distal stump), and the end organ (Campbell, 2008). One critical out their inherent dependency on a viable muscle end organ, may
factor in regeneration is the process of Wallerian degeneration, which enjoy a slightly longer period of viable recovery (Fu & Gordon, 1995).
affects the segment of nerve distal to transected axons. This process
starts within 48 hr and is usually complete by day 7–9 for motor axons
and by day 11 for sensory nerves, with the difference in times ascribed 4 | E LEC TRO D I AG N OS TI C E VA LUATI O N
to special consequences to dysfunction and early failure in neuromus‐ O F TPN I
cular transmission in motor fibers. Wallerian degeneration itself is an
early means to distinguish neurapraxic from axonal injuries provided The nature of the cellular and structural injury in the nerve de‐
electrodiagnostic studies are performed after this period. fies imaging in most scenarios, and electrodiagnostic testing (EDX)
There are generally accepted three mechanisms of repair: remy‐ is currently the most sensitive and specific method to evaluate
elination, collateral sprouting, and axon regrowth (Campbell, 2008; TPNI. However, EDXs such as nerve conduction study (NCS) and
Sulaiman & Gordon, 2013). In neurapraxic lesions, Schwann cells must electromyography (EMG) depend critically on the time‐dependent
de‐differentiate to a stage where cell division is possible to proliferate post‐injury sequelae to the nerve and end‐organ denervation, for a
and make new myelin, and this stage may take 3 months. Beyond this sensitivity that evolves over time after the injury. NCS determines
time period, persistent deficits imply axonal damage. The partial axon nerve conduction velocities (NCVs)  to help evaluate the axonal de‐
lesions recover through the mode of collateral sprouting that can take generation from demyelinating disorders and EMG evaluates the
up to 6 months. Terminal sprouts are chemotrophic outgrowths from motor unit action potential (MUAP) of a motor unit. Unlike surgi‐
the intact nerve endings which grow from the conical growth‐cone cal intervention, which can give some immediate information about
which emanates often from myelin‐free nodes of Ranvier within the the nature of the nerve injury, EDX has the key benefit of offering
intramuscular nerves (Robinson, 2000a). This process sacrifices single information about the functional status of the nerve and distally in‐
fiber motor‐specificity by placing more muscle fibers under the control nervated units and carries far less risk of damage to the nerve or
of remaining axons. Therefore, the motor unit size (the number of mus‐ perioperative complications. It is for these reasons EDX plays a cru‐
cle fibers dependent on one nerve fiber) can increase fivefold through cial ongoing role in the evaluation of these patients. The main aim
the process of collateral sprouting (Miller, 1987). In complete lesions, of EDX in TPNI is to classify the pathophysiology of nerve injury as
recovery depends on an axon regrowth rate of approximately 1 mm/ to axonal or demyelinating in nature, as well as to identify the loca‐
day, with some variation in this rate related to patient factor (age) and tion, grade, and prognosis of the injury. This information is critical
injury factors (injury site, mechanism of injury, proximity of the injury in deciding the plans for an intervention. Despite the importance of
to the nerve cell body, time and type of repair) (Miller, 1987; Post et these studies, it is also crucial to recognize the limitations and time
al., 2012), with the fastest reported regeneration rates associated with dependency of EDX data, which typically include information from
proximal limb and younger patients (Miller, 1987; Sunderland, 1990). distinct NCSs and EMG studies obtained in a single session.
During the regeneration process, the gap caused by the retracted Neurapraxia and axonotmesis are indistinguishable using EDX
stumps of a fully transected nerve is bridged by new tissue termed as testing for the first few days after nerve injury even when informa‐
“nerve bridge” and the regrowing axons from the proximal stump fol‐ tion is combined for sensory and motor nerves. The distal segment
low blood vessels through newly formed nerve bridge to reach the dis‐ of an injured nerve remains excitable during this time, and therefore
tal stump and their target organs (Cattin et al., 2015; Parrinello et al., EDX may not provide sufficient information to inform prognosis for
2010). However, there can be scar formation within the nerve bridge approximately 11 days after injury. Testing during this period can
that can lead to misdirection and aberrant regeneration of the sprout‐ nonetheless aid in localization of the injury and grading severity with
ing axons (Campbell, 2008). Therefore proximal lesions and those that the severest lesions (whether neurapraxic or axonotmetic) failing to
must regenerate through scar hold the worst prognosis owing to the evidence MUAP recruitment. In neurapraxia, the distal compound
time lost during which reversible target muscle denervation atrophy muscle action potential (CMAP) is normal, but stimulation proximal to
becomes irreversible (12–18  months) in axotomy state (Campbell, the lesion will not result in responses in the case of complete conduc‐
2008; Fu & Gordon, 1995). In the most severe forms of axonotmeses tion block. EMG in these cases will show no MUAP (indicating a com‐
and neurotmeses (Sunderland class 3–5), recovery is encumbered by plete neurapraxic lesion) or reduced recruitment (indicating a partial
|
4       MODRAK et al.

neurapraxic lesion)—all without abnormal spontaneous activity. These adjunctive strategies to promote functional recovery in TPNI patients.
changes usually resolve by 12 weeks, but delayed conduction may In contrast to the central nervous system (CNS), Schwann cells in the
persist owing to thinner and shorter internodes associated with remy‐ peripheral nervous system (PNS) produce a growth‐permissive envi‐
elination (Campbell, 2008; Preston & Shapiro, 2013). In axonotmesis, ronment for the injured nerve and play a pivotal role in several aspects
distal CMAP may be decreased or absent, but only after 7–9 days for of nerve repair such as degeneration, remyelination, and axonal growth
motor and 11 days for sensory fiber injury. A decrease in amplitude of (Sulaiman & Gordon, 2013; Sullivan et al., 2016). Several treatment
CMAP is proportional to the degree of axonal loss. EMG examination strategies have been employed to enhance the recovery process after
will show reduced recruitment or absent MUAP in the case of neurot‐ TPNI, including pharmacological, electrical, and cell‐based therapies
mesis. MUAP recruitment can also serve to indicate axonal continu‐ (Blits, Boer, & Verhaagen, 2002; Faroni, Mobasseri, Kingham, & Reid,
ity after Wallerian degeneration is complete. Abnormal spontaneous 2015; Magnaghi, Procacci, & Tata, 2009; Sullivan et al., 2016). While
activity will begin approximately 2–3 weeks after injury; however, each has shown some promise in treating patients, none has provided
the extent of this activity may not correlate with severity (Preston & a single universally applicable cure for the consequences of TPNI and
Shapiro, 2013). There are some notable shortcomings of commonly they are also not without significant pitfalls or potent side effect pro‐
used EDX techniques. EDX does not allow the critical distinction of files. Surgical interventions have been extensively reviewed by sev‐
subtypes of intermediate (Sunderland class 3–5) injuries (Robinson, eral authors (Campbell, 2008; Dvali & Mackinnon, 2007; Mackinnon,
2000a). The absent MUAP recruitment can be erroneously ascribed to 2018; Siemionow & Brzezicki, 2009); this section will highlight the
the loss of axonal continuity in mixed neurapraxic and axonotmetic le‐ usefulness of novel non‐surgical therapeutic approaches in promoting
sions when NCS‐EMG is performed before remyelination takes place. myelination following peripheral neurotrauma (Table 1).
Reinnervation changes first appear 2–3 months after axonotmesis
and are typified by nominal increases in CMAP amplitude; however, they
are probably better predicted based on MUAP configuration. Collateral 6 | PH A R M ACOTH E R A PY
sprouting results in longer duration polyphasic complexes and unstable
motor units in the subacute phase followed by large amplitude and long At present, there is no clinically approved pharmacological agent
duration motor unit potentials with persistently reduced recruitment. available for the treatment of TPNI. However, several agents have
The very first sign of axon regrowth can be nascent units, which are reported to be potential candidates to improve nerve regeneration
small, polyphasic motor unit potentials (Preston & Shapiro, 2013). by promoting myelination in the peripheral nerves.
Taken together, the nature of EDX combined with the nature of
nerve recovery allows the assessment of valuable clinical information,
albeit with constraints on timing. The more detailed information is 7 | S TE RO I D H O R M O N E S
obtained once denervation has taken place with EDX yielding more
definitive answers over time after TPNI (Campbell, 2008). Studies Steroid hormones, such as estrogen and progesterone, have been
performed within a week of injury allow localization of the injured investigated for use in TPNI (Chen et al., 2016; Koenig et al., 2000;
segment in addition to some information about the grade or complete‐ Nobakhti‐Afshar et al., 2016), with both in vivo and in vitro evidence
ness of the injury. After 1 week, studies that focus on motor nerves demonstrating the receptors for these hormones in the constituent
can distinguish between axonotmesis and neurapraxia, in addition to cells of the peripheral nerve such as Schwann cells, dorsal root ganglia
the information about completeness of the injury. This same distinc‐ neurons, sensory and autonomic neurons (Magnaghi et al., 2009). Both
tion is possible for sensory fibers at 11 days post‐injury. Studies per‐ hormones are known to be neuroprotective and promote myelination,
formed at 3–4 weeks allow all of the above distinctions in addition to with estrogen (17β‐Estradiol) promotes nerve recovery in both the
the assessment of abnormal spontaneous activity. Studies performed CNS and PNS (Schumacher et al., 2001; Zhu & Glaser, 2008). In the
at 3–4 months allow the documentation of reinnervation. PNS, estrogen exerts its effects through the modulation of steroid nu‐
clear receptors, which in turn upregulate the PI3K/AKT/mTOR signal‐
ing pathway and thereby promote the myelination (Chen et al., 2016).
5 |  TH E R A PEU TI C S TR ATEG I E S It is reported that PNS dorsal root ganglion neurons activate mTOR fol‐
lowing a sciatic nerve injury and this activity enhances axonal growth
Currently the treatment of choice for peripheral nerve injury that re‐ capacity with the increased expression of growth‐associated protein,
sults in laceration is advanced microsurgical end‐to‐end repair with GAP‐43 (Abe, Borson, Gambello, Wang, & Cavalli, 2010). Activation
tensionless epineurial sutures or autologous nerve grafting where of mTOR can also lead to downstream phosphorylation of S6 riboso‐
end‐to‐end anastomosis is not possible (Table 1) (Isaacs & Browne, mal protein and 4EBP‐1 to initiate protein synthesis (Park, Liu, Hu,
2014; Millesi, 2007; Muheremu & Ao, 2015; Siemionow & Brzezicki, Kanter, & He, 2010). Since GAP‐43 plays a key role in axon sprouting
2009). However, functional recovery after peripheral nerve repair is and outgrowth in regenerating axons (Aigner et al., 1995), it is possible
often unsatisfactory and it is apparent that microsurgical approaches that GAP‐43 is involved in estrogen‐induced upregulation of mTOR
fail to address the complex cellular and molecular events associated signaling and myelination. Progesterone may act in an independent
with TPNI. Therefore, there is an unmet need for new therapeutic or manner following binding to its specific progesterone receptor, which
TA B L E 1   Current surgical and potential therapeutic strategies for traumatic peripheral nerve injury

Limitations/comments/next
Strategies Intervention Timing of intervention Benefit References step
MODRAK et al.

Surgical End‐to‐end neurorrhaphy Variable (Days to Yes Campbell (2008), Dvali and Mackinnon (2007), Isaacs For selected nerve laceration
months) and Browne (2014), Mackinnon (2018), Millesi (2007), injuries
Muheremu and Ao (2015), Siemionow and Brzezicki
(2009)
  Nerve grafting       Slow rate of nerve regeneration
  Nerve transfer       Suboptimal functional outcome
  Conduit repair       Causes of poor outcome
unknown
          Possible benefit from adjunct
therapy
Non‐Surgical          
Physical agents Physical therapy Variable (Days to weeks) Yes Al‐Majed, Neumann, Brushart, and Gordon (2000), Mainly for post‐surgical
Haastert‐Talini et al. (2011), Nix and Hopf (1983), rehabilitation
Pockett and Gavin (1985), MacEwan, Gamble, Stephen,
and Ray (2019), Mendez et al. (2018), Gordon (2016),
Huang et al. (2012), Dow, Dennis, and Faulkner (2005),
Eberstein and Eberstein (1996), Kosman, Wood, and
Osborne (1948), Armada‐da‐Silva, Pereira, Amado, and
Veloso (2013), Marqueste, Alliez, Alluin, Jammes, and
Decherchi (2004), Asensio‐Pinilla, Udina, Jaramillo, and
Navarro (2009), Gordon and English (2016), Gordon,
Sulaiman, and Ladak (2009), Senger et al. (2019, 2018),
Sabatier, Redmon, Schwartz, and English (2008), Wan,
L. D., Xia, R., and Ding, W. L. (2010), Williams (1996),
de Oliveira et al. (2015), Hashmi et al. (2010), Schindl,
Schindl, Pernerstorfer‐Schön, and Schindl (2000),
Anders, Geuna, and Rochkind (2004), Barbosa (2010),
Gigo‐Benato et al. (2004), Mohammed, Al‐Mustawfi,
and Kaka (2007), Moges et al. (2011), Rochkind, Leider‐
Trejo, et al. (2007), Rochkind, Drory, Alon, Nissan, and
Ouaknine (2007)
  Electrical stimulation       Device‐dependent
  Low‐level laser therapy       Compliance issues
  Therapeutic ultrasound       Requires user‐friendly portable
devices
          Large clinical studies required
(Continues)
|
      5
TA B L E 1   (Continued)
|

Limitations/comments/next
6      

Strategies Intervention Timing of intervention Benefit References step

Pharmacological agents Steroid hormones Immediate/Early Yes Nobakhti‐Afshar, Najafpour, Mohammadi, and Zarei Mostly pre‐clinical findings
(2016), Koenig, Gong, and Pelissier (2000), Chen et
al. (2016), Zhu and Glaser (2008), Schumacher et al.
(2001), Magnaghi, Cavarretta, Galbiati, Martini, and
Melcangi (2001); Gronseth and Paduga (2012), Stark
and Amirfeyz (2013); Bernstein, Weiner, Tasciotti, and
Mathis (2016), Galloway, Jensen, Dailey, Thompson,
and Shelton (2000); Mohammadi, Azad‐Tirgan, and
Amini (2013), Suslu, Altun, Erdivanli, and Turan (2013),
Sakanaka et al. (1998), Sundem et al. (2016), Sargin,
Friedrichs, El‐Kordi, and Ehrenreich (2010), Elfar,
Jacobson, Puzas, Rosier, and Zuscik (2008), Toth et al.
(2008), Geary et al. (2017), Grasso et al. (2007), Tseng et
al. (2016), Clark, Hsu, Talukder, Noble, and Elfar (2020),
Noble, Tseng, Li, and Elfar (2019), Bowe, Kocsis, Targ,
and Waxman (1987); Sherratt, Bostock, and Sears (1980)
  Erythropoietin       Effective on crush and laceration
injuries
  4‐Aminopyridine       Improves motor function
          Improves myelination
          Improves nerve conduction
          Reduces muscle atrophy
          Proof‐of‐concept clinical studies
required
Miscellaneous Cell‐based therapy Unknown Unknown Walsh and Midha (2009), Cui et al. (2008), Chen et al. Limited in vitro findings
(2007), Dezawa, Takahashi, Esaki, Takano, and Sawada
(2001), Keilhoff, Stang, Goihl, Wolf, and Fansa (2006),
Kingham et al. (2007), Sowa, Imura, Numajiri, Nishino,
and Fushiki (2012), McKenzie, Biernaskie, Toma, Midha,
and Miller (2006), Walsh, Gordon, Addas, Kemp, and
Midha (2010), Penas and Navarro (2018), Arthur‐Farraj
et al. (2017), Oh et al. (2018), Pereira, Lebrun‐Julien, and
Suter (2012), Jagalur et al. (2011), Svaren and Meijer
(2008), Le et al. (2005), Topilko et al. (1994), Ma and
Svaren (2016), Stolt and Wegner (2016), Jacob, Lebrun‐
Julien, and Suter (2011), He et al. (2018)
  Epigenetic target       Proof‐of‐concept pre‐clinical
studies required
          Proof‐of‐concept clinical studies
required
MODRAK et al.

Note: For more information, please refer to the listed references.


MODRAK et al. |
      7

in turn induces myelination via upregulation of Krox20 (also termed been extensively studied in translational neuroscience and its role in
early growth response gene: Egr2) (Magnaghi et al., 2001), a transcrip‐ neuroprotection/neuroregeneration has been elegantly reviewed by
tion factor that regulates PNS myelination (Mirsky et al., 2008). This several authors (Magnaghi et al.; 2009; Maiese, 2016; Maiese, Chong,
activation directly drives the expression of myelin proteins like myelin Shang, & Wang, 2012; Sargin et al., 2010). It is documented that EPO
protein zero (MPZ) and peripheral myelin protein 22 (PMP22), both and its receptor (EPOR) are present in a wide variety of non‐erythroid
of which are primary constituents of the myelin sheath (Magnaghi cells throughout the body and may impact many biological functions.
et al., 2001; Schumacher et al., 2001). However, both estrogen and In the nervous system, EPO is produced and secreted by the neurons
progesterone suffer from well‐documented side effect profiles which of hippocampus, cortex, internal capsule, midbrain, and nervous sys‐
can differ from patient to patient depending on the dose and duration tem tumors (Kondyli, Gatzounis, Kyritsis, Varakis, & Assimakopoulou,
of drug treatments. The United States Food and Drug Administration 2010; Lombardero, Kovacs, & Scheithauer, 2011; Maiese, Hou, Chong,
mandated a “black box” warning on all estrogen products based on & Shang, 2009). The EPOR is also expressed on the myelin sheath of
the results of the Women's Health Initiative (WHI) because unopposed radicular nerves in human PNS (Hassan et al., 2004). We and others
estrogen use can increase the risk of endometrial cancer in intact uteri, have shown the presence and upregulation of EPOR at the site of pe‐
invasive breast cancer in post‐menopausal women, and deep vein ripheral nerve injury in mice (Elfar et al., 2008; Toth et al., 2008). EPO
thrombosis (DVT) (Miller & Harman, 2017). Additional risk factors for improves the sciatic function index of mice and rats after crush injury
estrogen include hypertension, obesity, diabetes mellitus, tobacco use, (Elfar et al., 2008; Geary et al., 2017; Grasso et al., 2007; Sundem et al.,
and/or history of venous thromboembolism. Estrogen receptor sign‐ 2016; Toth et al., 2008). and this functional improvement was observed
aling is also involved in prostate carcinogenesis (Bonkhoff, 2018). In with EPO administration prior to injury, immediately after injury, and
WHI studies, an increased risk of DVT, pulmonary embolism, stroke, after 1 week suggesting that the timing is not critical (Elfar et al., 2008;
and myocardial infarction was observed in women ≥65 years with daily Geary et al., 2017; Grasso et al., 2007; Sundem et al., 2016; Toth et
conjugated estrogens combined with medroxyprogesterone. al., 2008). Importantly, EPO‐mediated functional improvement can be
On the other hand, corticosteroids are sometimes used as a treat‐ demonstrated from single doses up to days after injuries in mice and
ment option for nerve injury, including acute spinal cord injury, fa‐ seems to correlate with injury (Elfar et al., 2008). Although the mecha‐
cial nerve paralysis, compressive neuropathy, and neuropathic pain nisms by which EPO works are still poorly understood, it is thought to
(Bracken, 2012; Eker, Cok, Aribogan, & Arslan, 2012; Gronseth & work by promoting the expression of myelin genes, MPZ and PMP22
Paduga, 2012; Stark & Amirfeyz, 2013). A recent clinical study also (Cervellini et al., 2013). We have shown that mice which received sys‐
demonstrated an important role for oral corticosteroids in the recov‐ temic EPO following nerve crush injury maintained more myelinated
ery of motor and sensory function following iatrogenic nerve injuries in axons at the site of injury (Sundem et al., 2016). In vitro EPO treatment
total hip and knee arthroplasty (Bernstein et al., 2016). However, sev‐ also promoted myelin formation and protected myelin from the effect
eral retrospective reviews have shown that long‐term glucocorticoid of nitric oxide (NO) exposure in co‐cultures of Schwann cells and dorsal
use, even in low doses, is a significant independent predictor of diverse root ganglion cells (Sundem et al., 2016). In addition to the fundamen‐
adverse effects, including bone loss or fracture, serious infections, tal myeloprotective role of EPO, these findings also demonstrated an
gastrointestinal bleeding or ulcer, and cataracts (Curtis et al., 2006; anti‐oxidative effect of EPO at the site of nerve injury which is con‐
Huscher et al., 2009; McDougall, Sibley, Haga, & Russell, 1994; Saag et sistent with the direct role of EPO against oxidative stress (Maiese et
al., 1994). The risk incurred with glucocorticoid use is both dose‐ and al., 2012). Given EPO’s widespread and frequent use to treat anemia
duration‐dependent. Although the generalized use of hormonal thera‐ in humans and its favorable side effect profile (Itri, 2002; Straus, 2002),
pies has not been common in the treatment of TPNI, several pre‐clinical early clinical trials have already begun investigating EPO’s therapeutic
studies have shown that topical steroids can improve functional recov‐ potential treating human peripheral nerve trauma associated with joint
ery and morphometric indices of the injured peripheral nerve (Galloway replacement surgery (Bernstein et al., 2016; Costa et al., 2015). It is also
et al., 2000; Mohammadi et al., 2013; Suslu et al., 2013) and this may noteworthy that clinical use of EPO is not without problems, in particu‐
have clinical implications after nerve transection because of its fewer lar the route of administration and the potent side effect profile. Aside
or no adverse effects compared to chronic systemic administration. from blood hyper‐viscosity from increased RBC production, hyperten‐
Therefore, the potential clinical usefulness of topical steroids to reduce sion, and its related problems (Abraham & Macres, 1991; Raine & Roger,
post‐injury nerve dysfunction warrants further investigations. 1991), the most common side effects of EPO therapy are headache and
an influenza‐like syndrome (Bennett, 1991; Eschbach et al., 1989).

8 | E RY TH RO P O I E TI N
9 | 4 ‐A M I N O PY R I D I N E
Erythropoietin (EPO), an endogenous hormone and FDA‐approved drug
for the treatment of anemia, has neuroprotective properties in both the 4‐aminopyridine (4‐AP), a broad‐spectrum potassium (K+) channel
CNS and PNS (Dame, Juul, & Christensen, 2001; Morishita, Masuda, blocker and FDA‐approved drug for the symptomatic treatment of
Nagao, Yasuda, & Sasaki, 1997; Sakanaka et al., 1998; Sundem et al., multiple sclerosis (MS) (Egeberg, Oh, & Bainbridge, 2012; Jensen,
2016). Although originally discovered as hematopoietic agent, EPO has Ravnborg, Dalgas, & Stenager, 2014), has been shown to improve
|
8       MODRAK et al.

neuromuscular function in patients with other demyelinating 10 | E LEC TR I C A L S TI M U L ATI O N


disorders including myasthenia gravis (Lundh, Nilsson, & Rosén,
1979), spinal cord injury (Hansebout, Blight, Fawcett, & Reddy, Electrical stimulation (ES) represents a promising non‐pharmacologi‐
1993), and Lambert‐Eaton syndrome (Sanders, Massey, Sanders, cal approach to accelerate and promote recovery following periph‐
& Edwards, 2000; Wirtz et al., 2009). The neurological benefits eral nerve injury (Al‐Majed, Neumann, Brushart, & Gordon, 2000;
of 4‐AP are believed to result from increases in action potential Haastert‐Talini et al., 2011; MacEwan et al. 2019; Mendez et al.,
duration, calcium influx, neurotransmitter release, synaptic trans‐ 2018; Nix & Hopf, 1983; Pockett & Gavin, 1985). Studies in animals
mission, and direct effects on muscle (Judge & Bever, 2006; van and humans have shown that ES promotes preferential reinnerva‐
Lunteren & Moyer, 2004; Smith, Felts, & John, 2000). 4‐AP may tion of both motor and sensory neurons, allowing for a faster recov‐
enhance cell‐membrane excitability and impulse conduction, and ery (Al‐Majed, Neumann, et al., 2000; Brushart, Jari, Verge, Rohde,
we observed that clinically relevant doses of 4‐AP treatment be‐ & Gordon, 2005; Gordon, 2016). In addition to promoting reinner‐
ginning shortly after injury enhance global functional recovery, vation, ES also aids in the remyelination process following periph‐
promote remyelination and improve nerve conduction velocity eral nerve injury (Huang et al., 2012). ES is also reported to provide
in an established mouse model of peripheral nerve crush injury benefit on nerve injury‐induced muscle atrophy and function (Dow
(Tseng et al., 2016). We also showed that 4‐AP can distinguish or et al., 2005; Eberstein & Eberstein, 1996; Kosman et al. 1948). Like
classify a crush injury from a transection injury by transient motor ES, treadmill running is reported to exert positive effects on nerve
function recovery (Clark et al., 2020; Noble et al., 2019; Tseng et regeneration and functional recovery (Armada‐da‐Silva et al., 2013;
al., 2016), an effect most likely related to its nerve conduction Marqueste et al., 2004). Moreover, the combination of these two
restoration properties because the time course of recovery was activity‐dependent therapies, ES and treadmill running, has been
far too rapid to be explained by regenerative mechanism and it shown to exert positive synergistic effects on nerve regeneration
is demonstrable at time‐points far earlier than those allowed by and muscle reinnervation (Asensio‐Pinilla et al. 2009; Gordon &
EDX (see above). Importantly, at the tissue level, 4‐AP also ap‐ English, 2016). While post‐operative direct low‐frequency (20  Hz)
pears to be myeloprotective and possibly axonoprotective (Clark ES of the proximal nerve stump for 1 hr (2‐ to 3‐fold threshold cur‐
et al., 2020; Tseng et al., 2016). Early studies with 4‐AP showed rent) is a standard regimen (Gordon et al., 2009), experimental stud‐
an increase in action potential duration and amplitude when ies have demonstrated that the same ES parameters 7 days prior to
measured on experimentally demyelinated mammalian peripheral axotomy (conditioning ES, CES) can promote functional nerve regen‐
nerve fibers (Bowe et al., 1987; Sherratt et al., 1980). These find‐ eration with upregulation of regeneration‐associated genes (Senger
ings highlight a possible diagnostic and therapeutic contribution et al., 2019, 2018). Although the exact mechanisms by which ES
to recovery and prognosis owed to excitatory molecules that may and exercise enhance nerve regeneration are poorly defined, cyclic
stabilize impulse conduction within hours to days after injury (Lee, adenosine monophosphate (cAMP) and brain‐derived neurotrophic
Claussen, & Oh, 2004; Robinson, 2000b; Shah & Jebson, 2008). factor (BDNF) are reported to play key roles (Al‐Majed, Brushart, &
Ongoing work on clinical translation of these findings may reveal Gordon, 2000; Gordon, 2009; Sabatier et al., 2008). ES causes an
clinical indications to TPNI, both in diagnosis and treatment (Elfar, increased influx of calcium into the neurons followed by an increase
2016). However, despite the beneficial effects, the clinical ability in intracellular cAMP levels (Gordon, 2009; Wenjin et al., 2011), and
of 4‐AP to restore function has been limited because of its narrow the downstream signaling molecule of cAMP, protein kinase A, pro‐
therapeutic window, the need for frequent dosing throughout the motes the expression of regeneration‐associated genes for axonal
day, and significant adverse side effects such as anxiety, tremors, growth (Gordon, 2009). Calcium‐induced phosphorylation of extra‐
and seizures (Blight, Toombs, Bauer, & Widmer, 1991; Goodman et cellular signal‐regulated kinase, ERK, has been shown to cause in‐
al., 2007; Goodman & Stone, 2013). Clinical trials have shown that creased expression of BDNF and myelination in the PNS (Wan, L. D.,
the efficacy of 4‐AP is related to the total drug exposure whereas et al., 2010; Wan, L., Xia, R., & Ding, W., 2010). Despite the reported
the toxicity is related to the peak serum levels (Bever et al., 1994; benefits with ES, the clinical use of implantable electrical devices re‐
Van Diemen et al., 1993). We have demonstrated that long‐term mains a challenge because of the nature of invasive procedures, the
treatment with 4‐AP  is beneficial following acute TPNI improv‐ lack of proper controls, and patient dissatisfaction with discomfort
ing functional recovery, myelin thickness, as well as important pa‐ (Williams, 1996).
rameter of EDX, such as nerve conduction velocity (NCV) (Clark
et al., 2020; Tseng et al., 2016). Our findings with 4‐AP convinc‐
ingly demonstrated that it can be used either as local, transdermal, 11 | C E LL‐ BA S E D TH E R A PY
or injectable form to exert its beneficial effect on TPNI recovery
(Clark et al., 2020; Noble et al., 2019; Tseng et al., 2016). Given Some of the severest injuries to peripheral nerves, such as avulsions,
the interplay between clinical efficacy, safety profile, and patient lacerations, and contusions may suffer from the additional loss of
compliance, further developments of safe methods of administer‐ Schwann cells needed to mediate regeneration. Such injuries may
ing 4‐AP are a reasonable focus in the near term to allow treat‐ fail to regenerate even at the accepted rate of 1 mm/day (Sulaiman
ment of the demyelinating component of TPNI. & Gordon, 2013). With this slower rate of regeneration, end organ
MODRAK et al. |
      9

reinnervation may take months or years or may fail eventually (Fu & cells (SKPCs) are neural crest‐related precursor cells found in the
Gordon, 1995; Sulaiman & Gordon, 2013). In the chronic axotomy dermis, which are capable of in vitro differentiation into neural crest
state, the denervated Schwann cells eventually lose their capacity to cells, including those with the features of Schwann cells and periph‐
support growth of the neurons and the lack of healthy Schwann cells eral neurons (McKenzie et al., 2006; Toma, McKenzie, Bagli, & Miller,
is an important issue in nerve regeneration, which spurs interest in 2005; Walsh et al., 2010).
cell‐based therapy (Fu & Gordon, 1995; Li, Terenghi, & Hall, 1997; While there is still much to learn about the role of SCs and their
Sulaiman & Gordon, 2013; You, Petrov, Chung, & Gordon, 1997). therapeutic potential in TPNI, there is now exciting in vivo and in
Cell‐based therapy is a promising branch of regenerative medi‐ vitro evidence indicating that they may be efficacious myelinating
cine and Schwann cell cultures have demonstrated favorable results phenotypes and localization can be maintained. Most importantly,
in the experimental model of TPNI with regeneration and remye‐ clinical studies exploring the feasibility of cell‐based strategies as an
lination (Borlongan, 2010; Guénard, Kleitman, Morrissey, Bunge, & adjunct therapy in chronic nerve injury will require careful investi‐
Aebischer, 1992). However, the process of human Schwann cell col‐ gation to determine the amount and method of cell delivery and the
lection involves invasive nerve biopsy and the culture also has lim‐ fate (cell survival and differentiation) of transplanted cells for safety,
ited in vitro expansion (Guest, Rao, Olson, Bunge, & Bunge, 1997). In reliability, and maximum efficacy.
search for a suitable cell line, stem cells (SCs) have garnered substan‐
tial interests as candidate transplant cells because of their availabil‐
ity, rapid in vitro expansion, survival, and integration within the host 12 | PH OTO B I O M O D U L ATI O N W ITH
tissue (Walsh & Midha, 2009). Based on the differentiation potential, L A S E R TH E R A PY
there are three categories of SC therapy: totipotent, pluripotent, and
multipotent stem cells. Pluripotent and multipotent stem cells have Photobiomodulation (PBM) with low‐level laser therapy (LLLT) has
been the focus of most research to date (Jiang, Jones, & Jia, 2017). been extensively studied in many clinical practices including physi‐
Embryonic stem cells (ESCs) are pluripotent SCs that can differ‐ cal medicine and rehabilitation, stroke, degenerative or traumatic
entiate into all three germ layers and Schwann cells can be generated brain disorders, and nerve repair (Hashmi et al., 2010; de Oliveira
from ESCs with 60% efficiency (Ziegler, Grigoryan, Yang, Thakor, & et al., 2015; Schindl et al., 2000). Laser therapy is a non‐invasive
Goldstein, 2011). In an animal model, injection of ESCs translated treatment modality that induces a photochemical reaction in the
to improved nerve repair and functional ability (Cui et al., 2008). cell and increases the DNA and RNA synthesis in the cell nucleus,
ESCs are limited in their therapeutic potential as they are in short with subsequent cell proliferation and protein synthesis, including
supply, owing to their source in the human embryo and they also changes in nerve cell action potential (Hashmi et al., 2010; de Oliveira
carry a risk of teratoma formation (Rippon & Bishop, 2004). Human et al., 2015). The precise mechanisms underlying PBM with LLLT and
induced pluripotent stem cells (hPSCs) have shown some promise in its therapeutic benefits are not fully understood. It is believed that
the regeneration and protection of myelin, possibly by providing an the initial trigger of PBM is the absorption of light (photons) by cy‐
exogenous source of self‐renewing Schwann cells (Ren, Wang, Peng, tochrome C oxidase in the mitochondrial respiratory pathway. The
Zhao, & Lu, 2012). Multipotent somatic stem cells from bone mar‐ increased activity of cytochrome C oxidase in turn increases the
row, mesenchymal stem cells (MSCs), have also shown some promise production of adenosine triphosphate and thus modulates the cell
with greater number of myelinated axons when used in combination functions (Karu, 2008; Passarella & Karu, 2014). Both clinical and
of artificial conduits and acellular grafts (Chen et al., 2007; Dezawa experimental studies have reported the beneficial effects of LLLT in
et al., 2001; Walsh & Midha, 2009), and they are also capable in my‐ TPNIs. Animal and in vitro studies have shown that LLLT promotes the
elinating cultured PC12 cells in vitro (Keilhoff et al., 2006). regeneration and functional recovery of the injured peripheral nerve,
In addition, adipose tissue and skin have been reported to pro‐ accelerates the myelination of regenerated nerves, increases the
vide easily accessible and less invasive potential sources of SCs. axonal diameter, and stimulates Schwann cell proliferation (Anders
Adipose‐derived stem cells (ADSCs) are a form of multipotent stem et al., 2004; Barbosa, 2010; Gigo‐Benato et al., 2004; Moges et al.,
cells that can differentiate into a Schwann‐like cell with similar func‐ 2011; Mohammed et al., 2007; Shen, Yang, & Liu, 2011; Yazdani et
tional properties (Kingham et al., 2007). It is possible that ADSCs may al., 2012). Double‐blind randomized studies have reported that post‐
exert their effects via the release of growth factors, such as nerve operative LLLT can enhance the regenerative process of the periph‐
growth factor, vascular endothelial growth factor, and BDNF, as well eral nerve with the increased number of myelinated axons (Rochkind,
as through the recruitment of endogenous Schwann cells (Sowa et Leider‐Trejo, et al., 2007) and improve the motor nerve function with
al., 2012, 2016). Although ADSCs are one of the more attractive SC functional recovery (Rochkind, Drory, et al., 2007). However, other
therapies due to their availability in the adipose tissue, one restric‐ studies did not observe any beneficial effects of LLLT (Bagis et al.,
tion is their tendency to differentiate toward adipocytes, which can 2003; Cömelekoğlu et al., 2002). Although LLLT has no reported ad‐
hinder nerve recovery (Faroni, Smith, Lu, & Reid, 2016). The skin and verse effects, there is no standardization in treatment with LLLT and
associated structures have a highly available supply of transplant‐ different irradiation parameters have been used in different mod‐
able cells due to their ability to differentiate into Schwann‐like cells els of PNI, and there is also a paucity in the clinical trials with LLLT.
(Sullivan et al., 2016; Walsh & Midha, 2009). Skin‐derived precursor Therefore, future studies exploring the effects of different variables,
|
10       MODRAK et al.

such as wavelengths, dose, continuous or pulsed mode, application vivo studies demonstrate that addition of HDAC3 inhibitors or the
site, and type of radiation would verify the usefulness of LLLT in TPNI ablation of HDAC3 in Schwann cells significantly increases the pro‐
as an adjunct therapy. duction of myelin, conduction velocity, CMAP amplitude, and en‐
hances sensory and motor function (He et al., 2018). HDAC3 also
antagonizes the myelogenic neuregulin‐PI3K‐AKT signaling axis.
13  | E PI G E N E TI C S A N D S M A LL M O LECU LE While these findings highlight the therapeutic potential of transient
A LTE R ATI O N S I N M Y E LI N ATI O N HDAC3 inhibition for improving peripheral myelin repair, it is un‐
known how different members of the HDAC family interact, their
There is growing evidence that the interplay of environmental risk fac‐ potential compensatory mechanisms, and how their expression and
tors and individual genetic susceptibility modulates disease presen‐ activity are regulated (Jacob et al., 2011).
tation and therapeutic responsiveness (Grolleau‐Julius, Ray, & Yung, It is evident that epigenetic changes can give rise to several sig‐
2010; Hedrich & Tsokos, 2011; Huynh & Casaccia, 2013). Epigenetics nificant disorders and we are just at the beginning of learning and
offers a promising link between genetic and environmental influ‐ understanding the contributions of these molecular genetic alter‐
ences on phenotype development (Egger, Liang, Aparicio, & Jones, ations to human diseases. While the reversible nature of epigene‐
2004; Grolleau‐Julius et al., 2010; Hedrich & Tsokos, 2011; Huynh tic alterations is encouraging in the effort to find therapies that can
& Casaccia, 2013; Picascia et al., 2015). Epigenetic modifications, reverse the molecular silencing early, it is unknown whether the
which include DNA methylation, post‐translational modifications of epigenetic modifications are the cause or the result of disease pro‐
histones, and non‐coding RNAs, result in the heritable silencing of gression. Future epigenetic studies in TPNIs will certainly enrich our
genes without a change in their coding sequence (Berger, Kouzarides, knowledge and pave the way to use epigenetics as a tool to identify
Shiekhattar, & Shilatifard, 2009; Egger et al., 2004). Such modifica‐ a disease biomarker and the potential therapeutic target.
tions can be induced by stress, tissue damage, and diseases, and can
affect both physiological and pathological processes (van den Elsen
14 | FU T U R E D I R EC TI O N S A N D
et al., 2014; Iridoy Zulet, Pulido Fontes, Ayuso Blanco, Lacruz Bescos,
CO N C LU S I O N S
& Mendioroz, 2017; Penas & Navarro, 2018; Picascia et al., 2015).
Epigenetic mechanisms thus play an essential role in transcriptional In the past three decades, tremendous advances were made in
control of genes, maintenance of cellular identity, cell activation, and TPNI diagnosis and management. However, the failure of well‐de‐
cellular repair processes during stress; with purported roles in many fined microsurgical techniques to provide satisfactory functional
disease processes including autoimmune disease, multiple sclerosis, recovery in the presence of complex cellular and molecular events
neuroinflammation, cancer, and normal aging (van den Elsen et al., with TPNI raises the question of using more robust therapeutic
2014; Iridoy Zulet et al., 2017; Penas & Navarro, 2018). approaches or combination of new approaches with current strat‐
The ever‐evolving field of epigenetics also has the potential to egies to promote functional recovery and quality of life in TPNI
play an important role in peripheral nerve recovery and myelination patients. Table 1 shows currently used and the potentially novel
(Arthur‐Farraj et al., 2017; Oh et al., 2018; Pereira et al., 2012; Salzer, therapeutic strategies for TPNI management. Future treatments
2015). Myelination by Schwann cells is under strict transcriptional for TPNI will likely require the development of new pharmaco‐
control (Jagalur et al., 2011; Svaren & Meijer, 2008) involving se‐ logic adjuvant agents as well as uncovering the mechanistic details
quential, feedforward cascades of promyelinating transcription of those currently available. Key to the successful translation of
factors where Sox 10 (SRY‐related HMG‐box‐10) and Oct6 (octamer‐ treatments into widespread use in humans is well‐executed clini‐
binding transcription factor‐6) synergistically induce the expression cal trials, which we and others have begun on several adjuvant
of Krox20 (also termed early growth response gene: Egr2). Krox 20 forms of TPNI treatments. Critical gains in promoting myelin for‐
is considered a master regulator of PNS myelination because it ac‐ mation or myeloprotection, and allowing early diagnosis will ad‐
tivates many myelin genes, suppresses myelination inhibitors, and vance treatment substantially.
maintains the myelinated state (Mirsky et al., 2008). Krox20 together
with NAB (NGFI‐A/Egr‐binding) protein regulates the transcription
AC K N OW L E D G M E N T S
of myelin structural proteins and biosynthetic components of myelin
lipid layer (Le et al., 2005; Topilko et al., 1994). In addition, several This work was supported by grants from the NIH (K08
epigenetic and chromatin modifiers are involved in myelination of AR060164‐01A) and DOD (W81XWH‐16‐1‐0725) in addition
mammalian nervous system and are crucial for SC differentiation, to institutional support from the University of Rochester and
myelin formation, and myelin maintenance (Ma & Svaren, 2016; Stolt Pennsylvania State University Medical Centers.
& Wegner, 2016). For example, histone deacetylase (HDAC) remod‐
els chromatin and condenses chromatic architecture and thus limits
AU T H O R C O N T R I B U T I O N S
DNA access for transcription factors (Jacob et al., 2011). Recently,
an HDAC3‐dependent pathway was identified as a potent inhibi‐ Conceptualization, M.M., M.A.H.T. and J.C.E.; Writing of the
tor of peripheral myelogenesis (He et al., 2018). Both in vitro and in Manuscript, M.M., M.A.H.T., K.G. and J.C.E.; Revision and Editing,
MODRAK et al. |
      11

M.M., M.A.H.T., G.K. and J.C.E. All authors read and approved the on the intact skin of the injured rat sciatic nerve. Lasers in Medical
final version of the manuscript. Science, 18(2), 83–88. https​://doi.org/10.1007/s10103-003-0258-6
Barbosa, R. I., Marcolino, A. M., de Jesus Guirro, R. R., Mazzer, N.,
Barbieri, C. H., & de Cássia Registro Fonseca, M. (2010). Comparative
effects of wavelengths of low‐power laser in regeneration of sciatic
C O N FL I C T O F I N T E R E S T
nerve in rats following crushing lesion. Lasers in Medical Science,
The authors have no conflict of interest to disclose. 25(3), 423–430. https​://doi.org/10.1007/s10103-009-0750-8
Bennett, W. M. (1991). Side effects of erythropoietin therapy. American
Journal of Kidney Diseases, 18(4 Suppl. 1), 84–86.
E T H I C A L P U B L I C AT I O N S TAT E M E N T Berger, S. L., Kouzarides, T., Shiekhattar, R., & Shilatifard, A. (2009). An
operational definition of epigenetics. Genes & Development, 23(7),
We confirm that we have read the Journal's position on issues in‐ 781–783. https​://doi.org/10.1101/gad.1787609
Bernstein, D. T., Weiner, B. K., Tasciotti, E., & Mathis, K. B. (2016). Does
volved in ethical publication and affirm that this report is consistent
the combination of erythropoietin and tapered oral corticosteroids
with those guidelines. improve recovery following iatrogenic nerve injury? Injury, 47(8),
1819–1823. https​://doi.org/10.1016/j.injury.2016.05.034
Bever, C. T., Young, D., Anderson, P. A., Krumholz, A., Conway, K., Leslie,
ORCID J., … Dhib‐Jalbut, S. (1994). The effects of 4‐aminopyridine in multi‐
ple sclerosis patients: Results of a randomized, placebo‐controlled,
John C. Elfar  https://orcid.org/0000-0002-4438-7118
double‐blind, concentration‐controlled, crossover trial. Neurology,
44(6), 1054–1059. https​://doi.org/10.1212/WNL.44.6.1054
Birch, R., Misra, P., Stewart, M. P., Eardley, W. G., Ramasamy, A., Brown,
REFERENCES K., … Etherington, J. (2012). Nerve injuries sustained during warfare:
Part I‐Epidemiology. Journal of Bone and Joint Surgery. British Volume,
Abe, N., Borson, S. H., Gambello, M. J., Wang, F., & Cavalli, V. (2010). 94(4), 523–528. https​://doi.org/10.1302/0301-620X.94B4.28483​
Mammalian target of rapamycin (mTOR) activation increases axonal Blight, A. R., Toombs, J. P., Bauer, M. S., & Widmer, W. R. (1991). The
growth capacity of injured peripheral nerves. Journal of Biological effects of 4‐aminopyridine on neurological deficits in chronic
Chemistry, 285(36), 28034–28043. https​://doi.org/10.1074/jbc. cases of traumatic spinal cord injury in dogs: A phase I clinical trial.
M110.125336 Journal of Neurotrauma, 8(2), 103–119. https​ ://doi.org/10.1089/
Abraham, P. A., & Macres, M. G. (1991). Blood pressure in hemodialysis neu.1991.8.103
patients during amelioration of anemia with erythropoietin. Journal Blits, B., Boer, G. J., & Verhaagen, J. (2002). Pharmacological, cell, and
of the American Society of Nephrology, 2(4), 927–936. gene therapy strategies to promote spinal cord regeneration. Cell
Aigner, L., Arber, S., Kapfhammer, J. P., Laux, T., Schneider, C., Botteri, Transplantation, 11(6), 593–613. https​://doi.org/10.3727/00000​
F., … Caroni, P. (1995). Overexpression of the neural growth‐asso‐ 00027​83985521
ciated protein GAP‐43 induces nerve sprouting in the adult ner‐ Bonkhoff, H. (2018). Estrogen receptor signaling in prostate cancer:
vous system of transgenic mice. Cell, 83(2), 269–278. https​://doi. Implications for carcinogenesis and tumor progression. Prostate,
org/10.1016/0092-8674(95)90168-X 78(1), 2–10. https​://doi.org/10.1002/pros.23446​
Al‐Majed, A. A., Brushart, T. M., & Gordon, T. (2000). Electrical stim‐ Borlongan, C. V. (2010). Remyelinating the transected peripheral nerve
ulation accelerates and increases expression of BDNF and trkB by fabricated Schwann cells derived from bone marrow. Experimental
mRNA in regenerating rat femoral motoneurons. European Journal of Neurology, 225(2), 243–245. https​ ://doi.org/10.1016/j.expne​
Neuroscience, 12(12), 4381–4390. urol.2010.07.008
Al‐Majed, A. A., Neumann, C. M., Brushart, T. M., & Gordon, T. (2000). Bowe, C. M., Kocsis, J. D., Targ, E. F., & Waxman, S. G. (1987). Physiological
Brief electrical stimulation promotes the speed and accuracy of effects of 4‐aminopyridine on demyelinated mammalian motor and
motor axonal regeneration. Journal of Neuroscience, 20(7), 2602– sensory fibers. Annals of Neurology, 22(2), 264–268. https​ ://doi.
2608. https​://doi.org/10.1523/JNEUR​OSCI.20-07-02602.2000 org/10.1002/ana.41022​0212
Anders, J. J., Geuna, S., & Rochkind, S. (2004). Phototherapy promotes Bracken, M. B. (2012). Steroids for acute spinal cord injury.
regeneration and functional recovery of injured peripheral nerve. Cochrane Database Systematic Review, 1, CD001046. https​://doi.
Neurological Research, 26(2), 233–239. https​://doi.org/10.1179/01616​ org/10.1002/14651​858.CD001​0 46.pub2
41042​25013914 Brushart, T. M., Jari, R., Verge, V., Rohde, C., & Gordon, T. (2005).
Armada‐da‐Silva, P. A., Pereira, C., Amado, S., & Veloso, A. P. (2013). Role Electrical stimulation restores the specificity of sensory axon re‐
of physical exercise for improving posttraumatic nerve regenera‐ generation. Experimental Neurology, 194(1), 221–229. https​ ://doi.
tion. International Review of Neurobiology, 109, 125–149. https​://doi. org/10.1016/j.expne​urol.2005.02.007
org/10.1016/B978-0-12-420045-6.00006-7 Campbell, W. W. (2008). Evaluation and management of peripheral
Arthur‐Farraj, P. J., Morgan, C. C., Adamowicz, M., Gomez‐Sanchez, nerve injury. Clinical Neurophysiology, 119(9), 1951–1965. https​://doi.
J. A., Fazal, S. V., Beucher, A., … Aitman, T. J. (2017). Changes in org/10.1016/j.clinph.2008.03.018
the coding and non‐coding transcriptome and DNA methylome Cattin, A. L., Burden, J. J., Van Emmenis, L., Mackenzie, F. E., Hoving,
that define the Schwann cell repair phenotype after nerve in‐ J. J., Garcia Calavia, N., … Lloyd, A. C. (2015). Macrophage‐induced
jury. Cell Reports, 20(11), 2719–2734. https​://doi.org/10.1016/j. blood vessels guide Schwann cell‐mediated regeneration of pe‐
celrep.2017.08.064 ripheral nerves. Cell, 162(5), 1127–1139. https​://doi.org/10.1016/j.
Asensio‐Pinilla, E., Udina, E., Jaramillo, J., & Navarro, X. (2009). Electrical cell.2015.07.021
stimulation combined with exercise increase axonal regeneration Cervellini, I., Annenkov, A., Brenton, T., Chernajovsky, Y., Ghezzi, P., &
after peripheral nerve injury. Experimental Neurology, 219(1), 258– Mengozzi, M. (2013). Erythropoietin (EPO) increases myelin gene
265. https​://doi.org/10.1016/j.expne​urol.2009.05.034 expression in CG4 oligodendrocyte cells through the classical EPO
Bagis, S., Comelekoglu, U., Coskun, B., Milcan, A., Buyukakilli, B., Sahin, receptor. Molecular Medicine, 19, 223–229. https​://doi.org/10.2119/
G., … Erdogan, C. (2003). No effect of GA‐AS (904 nm) laser irradiation molmed.2013.00013​
|
12       MODRAK et al.

Chen, C. J., Ou, Y. C., Liao, S. L., Chen, W. Y., Chen, S. Y., Wu, C. W., … Elfar, J. C., Jacobson, J. A., Puzas, J. E., Rosier, R. N., & Zuscik, M. J.
Hsu, S. H. (2007). Transplantation of bone marrow stromal cells for (2008). Erythropoietin accelerates functional recovery after periph‐
peripheral nerve repair. Experimental Neurology, 204(1), 443–453. eral nerve injury. Journal of Bone and Joint Surgery. American Volume,
https​://doi.org/10.1016/j.expne​urol.2006.12.004 90(8), 1644–1653. https​://doi.org/10.2106/JBJS.G.00557​
Chen, Y., Guo, W., Xu, L., Li, W., Cheng, M., Hu, Y., & Xu, W. (2016). 17β‐ Elfar J NMMMGA. (2016). A clinical trial of 4AP for the treatment of
estradiol promotes Schwann cell proliferation and differentiation, post‐prostatectomy peripheral nerve injury in humans (IND 128117
accelerating early remyelination in a mouse peripheral nerve injury approved 2016).
model. BioMed Research International, 2016, 7891202. Eschbach, J. W., Abdulhadi, M. H., Browne, J. K., Delano, B. G., Downing,
Clark, A. R., Hsu, C. G., Talukder, M. A. H., Noble, M., & Elfar, J. C. (2020). M. R., Egrie, J. C., … Haley, N. R. (1989). Recombinant human erythro‐
Transdermal delivery of 4‐aminopyridine accelerates motor func‐ poietin in anemic patients with end‐stage renal disease. Results of a
tional recovery and improves nerve morphology following sciatic phase III multicenter clinical trial. Annals of Internal Medicine, 111(12),
nerve crush injury in mice. Neural Regeneration Research, 15(1), 136– 992–1000. https​://doi.org/10.7326/0003-4819-111-12-992
144. https​://doi.org/10.4103/1673-5374.264471 Evans, G. R. (2001). Peripheral nerve injury: A review and approach to
Cömelekoğlu, U., Bagiş, S., Büyükakilli, B., Sahin, G., Erdoğan, C., & Kanik, A. tissue engineered constructs. Anatomical Record, 263(4), 396–404.
(2002). Acute electrophysiological effect of pulsed gallium‐arsenide low‐ https​://doi.org/10.1002/ar.1120
energy laser irradiation on isolated frog sciatic nerve. Lasers in Medical Faroni, A., Mobasseri, S. A., Kingham, P. J., & Reid, A. J. (2015). Peripheral
Science, 17(1), 62–67. https​://doi.org/10.1007/s10103-002-8268-3 nerve regeneration: Experimental strategies and future perspec‐
Costa, D. D., Beghi, E., Carignano, P., Pagliacci, C., Faccioli, F., Pupillo, E., tives. Advanced Drug Delivery Reviews, 82–83, 160–167. https​://doi.
… Redaelli, T. (2015). Tolerability and efficacy of erythropoietin (EPO) org/10.1016/j.addr.2014.11.010
treatment in traumatic spinal cord injury: A preliminary randomized Faroni, A., Smith, R. J., Lu, L., & Reid, A. J. (2016). Human Schwann‐like
comparative trial vs. methylprednisolone (MP). Neurological Sciences, cells derived from adipose‐derived mesenchymal stem cells rapidly
36(9), 1567–1574. https​://doi.org/10.1007/s10072-015-2182-5 de‐differentiate in the absence of stimulating medium. European
Cui, L., Jiang, J., Wei, L., Zhou, X., Fraser, J. L., Snider, B. J., & Yu, S. P. Journal of Neuroscience, 43(3), 417–430. https​://doi.org/10.1111/
(2008). Transplantation of embryonic stem cells improves nerve re‐ ejn.13055​
pair and functional recovery after severe sciatic nerve axotomy in Fu, S. Y., & Gordon, T. (1995). Contributing factors to poor functional
rats. Stem Cells, 26(5), 1356–1365. https​ ://doi.org/10.1634/stemc​ recovery after delayed nerve repair: Prolonged axotomy. Journal
ells.2007-0333 of Neuroscience, 15(5 Pt 2), 3876–3885. https​://doi.org/10.1523/
Curtis, J. R., Westfall, A. O., Allison, J., Bijlsma, J. W., Freeman, A., JNEUR​OSCI.15-05-03876.1995
George, V., … Saag, K. G. (2006). Population‐based assessment of ad‐ Galloway, E. B., Jensen, R. L., Dailey, A. T., Thompson, B. G., & Shelton,
verse events associated with long‐term glucocorticoid use. Arthritis C. (2000). Role of topical steroids in reducing dysfunction after
and Rheumatism, 55(3), 420–426. https​://doi.org/10.1002/art.21984​ nerve injury. Laryngoscope, 110(11), 1907–1910. https​://doi.
Dame, C., Juul, S. E., & Christensen, R. D. (2001). The biology of eryth‐ org/10.1097/00005​537-20001​1000-00026​
ropoietin in the central nervous system and its neurotrophic and Geary, M. B., Li, H., Zingman, A., Ketz, J., Zuscik, M., De Mesy Bentley, K.
neuroprotective potential. Biology of the Neonate, 79(3–4), 228–235. L., … Elfar, J. C. (2017). Erythropoietin accelerates functional recov‐
de Oliveira, R. F., de Andrade Salgado, D. M., Trevelin, L. T., Lopes, R. M., ery after moderate sciatic nerve crush injury. Muscle and Nerve, 56(1),
da Cunha, S. R., Aranha, A. C., … de Freitas, P. M. (2015). Benefits of 143–151. https​://doi.org/10.1002/mus.25459​
laser phototherapy on nerve repair. Lasers in Medical Science, 30(4), Gigo‐Benato, D., Geuna, S., de Castro, R. A., Tos, P., Fornaro, M., Boux,
1395–1406. https​://doi.org/10.1007/s10103-014-1531-6 E., … Giacobini‐Robecchi, M. G. (2004). Low‐power laser biostim‐
Dezawa, M., Takahashi, I., Esaki, M., Takano, M., & Sawada, H. (2001). ulation enhances nerve repair after end‐to‐side neurorrhaphy: A
Sciatic nerve regeneration in rats induced by transplantation of in double‐blind randomized study in the rat median nerve model.
vitro differentiated bone‐marrow stromal cells. European Journal of Lasers in Medical Science, 19(1), 57–65. https​://doi.org/10.1007/
Neuroscience, 14(11), 1771–1776. s10103-004-0300-3
Dow, D. E., Dennis, R. G., & Faulkner, J. A. (2005). Electrical stimulation Goodman, A. D., Cohen, J. A., Cross, A., Vollmer, T., Rizzo, M., Cohen, R.,
attenuates denervation and age‐related atrophy in extensor digi‐ … Blight, A. R. (2007). Fampridine‐SR in multiple sclerosis: A random‐
torum longus muscles of old rats. Journals of Gerontology. Series A, ized, double‐blind, placebo‐controlled, dose‐ranging study. Multiple
Biological Sciences and Medical Sciences, 60(4), 416–424. https​://doi. Sclerosis Journal, 13(3), 357–368. https​://doi.org/10.1177/13524​
org/10.1093/geron​a/60.4.416 58506​069538
Dvali, L., & Mackinnon, S. (2007). The role of microsurgery in nerve Goodman, A. D., & Stone, R. T. (2013). Enhancing neural transmission
repair and nerve grafting. Hand Clinics, 23(1), 73–81. https​://doi. in multiple sclerosis (4‐aminopyridine therapy). Neurotherapeutics,
org/10.1016/j.hcl.2007.02.003 10(1), 106–110. https​://doi.org/10.1007/s13311-012-0156-3
Eberstein, A., & Eberstein, S. (1996). Electrical stimulation of denervated Gordon, T. (2009). The role of neurotrophic factors in nerve regen‐
muscle: Is it worthwhile? Medicine and Science in Sports and Exercise, eration. Neurosurgical Focus, 26(2), E3. https​://doi.org/10.3171/
28(12), 1463–1469. https​://doi.org/10.1097/00005​768-19961​ FOC.2009.26.2.E3
2000-00004​ Gordon, T. (2016). Electrical stimulation to enhance axon regener‐
Egeberg, M. D., Oh, C. Y., & Bainbridge, J. L. (2012). Clinical overview of ation after peripheral nerve injuries in animal models and hu‐
dalfampridine: An agent with a novel mechanism of action to help mans. Neurotherapeutics, 13(2), 295–310. https​://doi.org/10.1007/
with gait disturbances. Clinical Therapeutics, 34(11), 2185–2194. s13311-015-0415-1
https​://doi.org/10.1016/j.clint​hera.2012.10.003 Gordon, T., & English, A. W. (2016). Strategies to promote peripheral
Egger, G., Liang, G., Aparicio, A., & Jones, P. A. (2004). Epigenetics in nerve regeneration: Electrical stimulation and/or exercise. European
human disease and prospects for epigenetic therapy. Nature, Journal of Neuroscience, 43(3), 336–350. https​ ://doi.org/10.1111/
429(6990), 457–463. https​://doi.org/10.1038/natur​e 02625 ejn.13005​
Eker, H. E., Cok, O. Y., Aribogan, A., & Arslan, G. (2012). Management Gordon, T., Sulaiman, O. A., & Ladak, A. (2009). Chapter 24: Electrical
of neuropathic pain with methylprednisolone at the site of nerve stimulation for improving nerve regeneration: Where do we stand?
injury. Pain Medicine, 13(3), 443–451. https​ ://doi.org/10.1111/ International Review of Neurobiology, 87, 433–444. https​ ://doi.
j.1526-4637.2011.01323.x org/10.1016/S0074-7742(09)87024-4.
MODRAK et al. |
      13

Grasso, G., Meli, F., Fodale, V., Calapai, G., Buemi, M., & Iacopino, D. G. Itri, L. M. (2002). The use of epoetin alfa in chemotherapy patients: A
(2007). Neuroprotective potential of erythropoietin and darbep‐ consistent profile of efficacy and safety. Seminars in Oncology, 29(3
oetin alfa in an experimental model of sciatic nerve injury. Journal Suppl. 8), 81–87. https​://doi.org/10.1053/sonc.2002.33539​
of Neurosurgery: Spine, 7(6), 645–651. https​://doi.org/10.3171/ Jacob, C., Lebrun‐Julien, F., & Suter, U. (2011). How histone deacetylases
SPI-07/12/645 control myelination. Molecular Neurobiology, 44(3), 303–312. https​://
Grolleau‐Julius, A., Ray, D., & Yung, R. L. (2010). The role of epigenetics doi.org/10.1007/s12035-011-8198-9. Epub 2011 Aug 23.
in aging and autoimmunity. Clinical Reviews in Allergy and Immunology, Jagalur, N. B., Ghazvini, M., Mandemakers, W., Driegen, S., Maas, A.,
39(1), 42–50. https​://doi.org/10.1007/s12016-009-8169-3 Jones, E. A., … Meijer, D. (2011). Functional dissection of the Oct6
Gronseth, G. S., & Paduga, R. (2012). Evidence‐based guideline up‐ Schwann cell enhancer reveals an essential role for dimeric Sox10
date: Steroids and antivirals for Bell palsy: Report of the Guideline binding. Journal of Neuroscience, 31(23), 8585–8594. https​ ://doi.
Development Subcommittee of the American Academy of Neurology. org/10.1523/JNEUR​OSCI.0659-11.2011
Neurology, 79(22), 2209–2213. https​://doi.org/10.1212/WNL.0b013​ Jensen, H. B., Ravnborg, M., Dalgas, U., & Stenager, E. (2014). 4‐
e3182​75978c Aminopyridine for symptomatic treatment of multiple sclerosis: A
Guénard, V., Kleitman, N., Morrissey, T. K., Bunge, R. P., & Aebischer, P. systematic review. Therapeutic Advances in Neurological Disorders,
(1992). Syngeneic Schwann cells derived from adult nerves seeded 7(2), 97–113. https​://doi.org/10.1177/17562​85613​512712
in semipermeable guidance channels enhance peripheral nerve re‐ Jiang, L., Jones, S., & Jia, X. (2017). Stem cell transplantation for pe‐
generation. Journal of Neuroscience, 12(9), 3310–3320. https​://doi. ripheral nerve regeneration: Current options and opportunities.
org/10.1523/JNEUR​OSCI.12-09-03310.1992 International Journal of Molecular Sciences, 18(1), 94. https​ ://doi.
Guest, J. D., Rao, A., Olson, L., Bunge, M. B., & Bunge, R. P. (1997). The org/10.3390/ijms1​8 010094
ability of human Schwann cell grafts to promote regeneration in the Judge, S. I., & Bever, C. T. (2006). Potassium channel blockers in multiple
transected nude rat spinal cord. Experimental Neurology, 148(2), 502– sclerosis: Neuronal Kv channels and effects of symptomatic treat‐
522. https​://doi.org/10.1006/exnr.1997.6693 ment. Pharmacology & Therapeutics, 111(1), 224–259. https​ ://doi.
Haastert‐Talini, K., Schmitte, R., Korte, N., Klode, D., Ratzka, A., & Grothe, org/10.1016/j.pharm​thera.2005.10.006
C. (2011). Electrical stimulation accelerates axonal and functional pe‐ Karu, T. I. (2008). Mitochondrial signaling in mammalian cells activated
ripheral nerve regeneration across long gaps. Journal of Neurotrauma, by red and near‐IR radiation. Photochemistry and Photobiology, 84(5),
28(4), 661–674. https​://doi.org/10.1089/neu.2010.1637 1091–1099. https​://doi.org/10.1111/j.1751-1097.2008.00394.x
Hansebout, R. R., Blight, A. R., Fawcett, S., & Reddy, K. (1993). 4‐ Kaya, Y., & Sarikcioglu, L. (2015). Sir Herbert Seddon (1903–1977) and
Aminopyridine in chronic spinal cord injury: A controlled, double‐ his classification scheme for peripheral nerve injury. Childs Nervous
blind, crossover study in eight patients. Journal of Neurotrauma, 10(1), System, 31(2), 177–180. https​://doi.org/10.1007/s00381-014-2560-y
1–18. https​://doi.org/10.1089/neu.1993.10.1 Keilhoff, G., Stang, F., Goihl, A., Wolf, G., & Fansa, H. (2006).
Hashmi, J. T., Huang, Y. Y., Osmani, B. Z., Sharma, S. K., Naeser, M. A., & Transdifferentiated mesenchymal stem cells as alternative ther‐
Hamblin, M. R. (2010). Role of low‐level laser therapy in neurorehabil‐ apy in supporting nerve regeneration and myelination. Cellular
itation. PM&R, 2(12 Suppl. 2), S292–S305. https​://doi.org/10.1016/j. and Molecular Neurobiology, 26(7–8), 1235–1252. https​://doi.
pmrj.2010.10.013 org/10.1007/s10571-006-9029-9
Hassan, K., Gross, B., Simri, W., Rubinchik, I., Cohen, H., Jacobi, J., … Kingham, P. J., Kalbermatten, D. F., Mahay, D., Armstrong, S. J., Wiberg,
Kristal, B. (2004). The presence of erythropoietin receptors in the M., & Terenghi, G. (2007). Adipose‐derived stem cells differentiate
human peripheral nervous system. Clinical Nephrology, 61(2), 127– into a Schwann cell phenotype and promote neurite outgrowth
129. https​://doi.org/10.5414/CNP61127 in vitro. Experimental Neurology, 207(2), 267–274. https​://doi.
He, X., Zhang, L., Queme, L. F., Liu, X., Lu, A., Waclaw, R. R., … Lu, Q. org/10.1016/j.expne​urol.2007.06.029
R. (2018). A histone deacetylase 3‐dependent pathway delimits pe‐ Koenig, H. L., Gong, W. H., & Pelissier, P. (2000). Role of progesterone in
ripheral myelin growth and functional regeneration. Nature Medicine, peripheral nerve repair. Reviews of Reproduction, 5(3), 189–199. https​
24(3), 338–351. https​://doi.org/10.1038/nm.4483 ://doi.org/10.1530/ror.0.0050189
Hedrich, C. M., & Tsokos, G. C. (2011). Epigenetic mechanisms in sys‐ Kondyli, M., Gatzounis, G., Kyritsis, A., Varakis, J., & Assimakopoulou,
temic lupus erythematosus and other autoimmune diseases. Trends M. (2010). Immunohistochemical detection of phosphorylated
in Molecular Medicine, 17(12), 714–724. https​://doi.org/10.1016/j. JAK‐2 and STAT‐5 proteins and correlation with erythropoietin
molmed.2011.07.005 receptor (EpoR) expression status in human brain tumors. Journal
Huang, J., Lu, L., Zhang, J., Hu, X., Zhang, Y., Liang, W., … Luo, Z. of Neuro‐oncology, 100(2), 157–164. https​://doi.org/10.1007/
(2012). Electrical stimulation to conductive scaffold promotes ax‐ s11060-010-0156-2
onal regeneration and remyelination in a rat model of large nerve Kosman, A. J., Wood, E. C., & Osborne, S. L. (1948). Effect of electrical
defect. PLoS ONE, 7(6), e39526. https​://doi.org/10.1371/journ​al. stimulation upon atrophy of partially denervated skeletal muscle of
pone.0039526 the rat. American Journal of Physiology, 154(3), 451–454. https​://doi.
Huscher, D., Thiele, K., Gromnica‐Ihle, E., Hein, G., Demary, W., Dreher, org/10.1152/ajple​gacy.1948.154.3.451
R., … Buttgereit, F. (2009). Dose‐related patterns of glucocorticoid‐ Le, N., Nagarajan, R., Wang, J. Y., Svaren, J., LaPash, C., Araki, T., …
induced side effects. Annals of the Rheumatic Diseases, 68(7), 1119– Milbrandt, J. (2005). Nab proteins are essential for peripheral ner‐
1124. https​://doi.org/10.1136/ard.2008.092163 vous system myelination. Nature Neuroscience, 8(7), 932–940. https​
Huynh, J. L., & Casaccia, P. (2013). Epigenetic mechanisms in multiple scle‐ ://doi.org/10.1038/nn1490
rosis: Implications for pathogenesis and treatment. Lancet Neurology, Lee, D. H., Claussen, G. C., & Oh, S. (2004). Clinical nerve conduc‐
12(2), 195–206. https​://doi.org/10.1016/S1474-4422(12)70309-5 tion and needle electromyography studies. Journal of American
Iridoy Zulet, M., Pulido Fontes, L., Ayuso Blanco, T., Lacruz Bescos, F., Academy of Orthopaedic Surgeons, 12(4), 276–287. https​://doi.
& Mendioroz, I. M. (2017). Epigenetic changes in neurology: DNA org/10.5435/00124​635-20040​7000-00008​
methylation in multiple sclerosis. Neurologia, 32(7), 463–468. https​ Li, H., Terenghi, G., & Hall, S. M. (1997). Effects of delayed re‐inner‐
://doi.org/10.1016/j.nrl.2015.03.011 vation on the expression of c‐erbB receptors by chronically de‐
Isaacs, J., & Browne, T. (2014). Overcoming short gaps in peripheral nervated rat Schwann cells in vivo. Glia, 20(4), 333–347. https​://
nerve repair: Conduits and human acellular nerve allograft. Hand, doi.org/10.1002/(SICI)1098-1136(19970​8 )20:4<333:AID-GLIA6​
9(2), 131–137. https​://doi.org/10.1007/s11552-014-9601-6 >3.0.CO;2-6
|
14       MODRAK et al.

Lombardero, M., Kovacs, K., & Scheithauer, B. W. (2011). Erythropoietin: Mirsky, R., Woodhoo, A., Parkinson, D. B., Arthur‐Farraj, P., Bhaskaran,
A hormone with multiple functions. Pathobiology, 78(1), 41–53. https​ A., & Jessen, K. R. (2008). Novel signals controlling embryonic
://doi.org/10.1159/00032​2975 Schwann cell development, myelination and dedifferentiation.
Lundh, H., Nilsson, O., & Rosén, I. (1979). Effects of 4‐aminopyridine in Journal of the Peripheral Nervous System, 13(2), 122–135. https​://doi.
myasthenia gravis. Journal of Neurology, Neurosurgery and Psychiatry, org/10.1111/j.1529-8027.2008.00168.x
42(2), 171–175. https​://doi.org/10.1136/jnnp.42.2.171 Missios, S., Bekelis, K., & Spinner, R. J. (2014). Traumatic periph‐
Ma, K. H., & Svaren, J. (2016). Epigenomic reprogramming in peripheral eral nerve injuries in children: Epidemiology and socioeconom‐
nerve injury. Neural Regeneration Research, 11(12), 1930–1931. https​:// ics. Journal of Neurosurgery Pediatrics, 14(6), 688–694. https​://doi.
doi.org/10.4103/1673-5374.197133 org/10.3171/2014.8.PEDS1​4112
MacEwan, M. R., Gamble, P., Stephen, M., & Ray, W. Z. (2019). Moges, H., Wu, X., McCoy, J., Vasconcelos, O. M., Bryant, H., Grunberg,
Therapeutic electrical stimulation of injured peripheral nerve tissue N. E., & Anders, J. J. (2011). Effect of 810 nm light on nerve regenera‐
using implantable thin‐film wireless nerve stimulators. Journal of tion after autograft repair of severely injured rat median nerve. Lasers
Neurosurgery, 130, 486–495. in Surgery and Medicine, 43(9), 901–906. https​://doi.org/10.1002/
Mackinnon, S. E. (2018). Future perspectives in the management of lsm.21117​
nerve injuries. Journal of Reconstructive Microsurgery, 34(9), 672–674. Mohammadi, R., Azad‐Tirgan, M., & Amini, K. (2013). Dexamethasone
https​://doi.org/10.1055/s-0038-1639353 topically accelerates peripheral nerve repair and target organ rein‐
Magnaghi, V., Cavarretta, I., Galbiati, M., Martini, L., & Melcangi, R. C. nervation: A transected sciatic nerve model in rat. Injury, 44(4), 565–
(2001). Neuroactive steroids and peripheral myelin proteins. Brain 569. https​://doi.org/10.1016/j.injury.2012.10.013
Research Reviews, 37(1–3), 360–371. https​://doi.org/10.1016/ Mohammed, I. F., Al‐Mustawfi, N., & Kaka, L. N. (2007). Promotion of
S0165-0173(01)00140-0 regenerative processes in injured peripheral nerve induced by low‐
Magnaghi, V., Procacci, P., & Tata, A. M. (2009). Chapter 15: Novel level laser therapy. Photomedicine and Laser Surgery, 25(2), 107–111.
pharmacological approaches to Schwann cells as neuroprotective https​://doi.org/10.1089/pho.2006.1090
agents for peripheral nerve regeneration. International Review of Morishita, E., Masuda, S., Nagao, M., Yasuda, Y., & Sasaki, R. (1997).
Neurobiology, 87, 295–315. Erythropoietin receptor is expressed in rat hippocampal and cerebral
Maiese, K. (2016). Regeneration in the nervous system with erythropoie‐ cortical neurons, and erythropoietin prevents in vitro glutamate‐in‐
tin. Frontiers in Bioscience, 21, 561–596. https​://doi.org/10.2741/4408 duced neuronal death. Neuroscience, 76(1), 105–116. https​ ://doi.
Maiese, K., Chong, Z. Z., Shang, Y. C., & Wang, S. (2012). Erythropoietin: org/10.1016/S0306-4522(96)00306-5
New directions for the nervous system. International Journal of Muheremu, A., & Ao, Q. (2015). Past, present, and future of nerve con‐
Molecular Sciences, 13(9), 11102–11129. https​ ://doi.org/10.3390/ duits in the treatment of peripheral nerve injury. BioMed Research
ijms1​30911102 International, 2015, 237507. https​://doi.org/10.1155/2015/237507
Maiese, K., Hou, J., Chong, Z. Z., & Shang, Y. C. (2009). Erythropoietin, Nix, W. A., & Hopf, H. C. (1983). Electrical stimulation of regenerating
forkhead proteins, and oxidative injury: Biomarkers and biology. nerve and its effect on motor recovery. Brain Research, 272(1), 21–25.
The Scientific World Journal, 9, 1072–1104. https​://doi.org/10.1100/ https​://doi.org/10.1016/0006-8993(83)90360-8
tsw.2009.121 Nobakhti‐Afshar, A., Najafpour, A., Mohammadi, R., & Zarei, L. (2016).
Marqueste, T., Alliez, J. R., Alluin, O., Jammes, Y., & Decherchi, P. (2004). Assessment of neuroprotective effects of local administration of 17‐
Neuromuscular rehabilitation by treadmill running or electrical stim‐ beta‐ estradiol on peripheral nerve regeneration in ovariectomized
ulation after peripheral nerve injury and repair. Journal of Applied female rats. Bulletin of Emergency & Trauma, 4(3), 141–149.
Physiology, 96(5), 1988–1995. https​://doi.org/10.1152/jappl​physi​ Noble, J., Munro, C. A., Prasad, V. S., & Midha, R. (1998). Analysis of
ol.00775.2003 upper and lower extremity peripheral nerve injuries in a population
McDougall, R., Sibley, J., Haga, M., & Russell, A. (1994). Outcome in pa‐ of patients with multiple injuries. Journal of Trauma, 45(1), 116–122.
tients with rheumatoid arthritis receiving prednisone compared to https​://doi.org/10.1097/00005​373-19980​7000-00025​
matched controls. Journal of Rheumatology, 21(7), 1207–1213. Noble, M., Tseng, K. C., Li, H., & Elfar, J. C. (2019). 4‐Aminopyridine as a
McKenzie, I. A., Biernaskie, J., Toma, J. G., Midha, R., & Miller, F. D. single agent diagnostic and treatment for severe nerve crush injury.
(2006). Skin‐derived precursors generate myelinating Schwann Military Medicine, 184(Suppl. 1), 379–385. https​://doi.org/10.1093/
cells for the injured and dysmyelinated nervous system. Journal of milme​d/usy399
Neuroscience, 26(24), 6651–6660. https​ ://doi.org/10.1523/JNEUR​ Oh, Y. M., Mahar, M., Ewan, E. E., Leahy, K. M., Zhao, G., & Cavalli, V.
OSCI.1007-06.2006 (2018). Epigenetic regulator UHRF1 inactivates REST and growth
Mendez, A., Hopkins, A., Biron, V. L., Seikaly, H., Zhu, L. F., & Côté, D. suppressor gene expression via DNA methylation to promote axon
W. J. (2018). Brief electrical stimulation and synkinesis after facial regeneration. Proceedings of the National Academy of Sciences of
nerve crush injury: A randomized prospective animal study. Journal the United States of America, 115(52), E12417–E12426. https​://doi.
of Otolaryngology ‐ Head & Neck Surgery, 47(1), 20. https​ ://doi. org/10.1073/pnas.18125​18115​
org/10.1186/s40463-018-0264-0 Park, K. K., Liu, K., Hu, Y., Kanter, J. L., & He, Z. (2010). PTEN/mTOR and
Menorca, R. M., Fussell, T. S., & Elfar, J. C. (2013). Nerve physiology: axon regeneration. Experimental Neurology, 223(1), 45–50. https​://
Mechanisms of injury and recovery. Hand Clinics, 29(3), 317–330. doi.org/10.1016/j.expne​urol.2009.12.032
https​://doi.org/10.1016/j.hcl.2013.04.002 Parrinello, S., Napoli, I., Ribeiro, S., Wingfield Digby, P., Fedorova, M.,
Miller, R. G. (1987). AAEE minimonograph #28: Injury to peripheral motor Parkinson, D. B., … Lloyd, A. C. (2010). EphB signaling directs periph‐
nerves. Muscle and Nerve, 10(8), 698–710. https​://doi.org/10.1002/ eral nerve regeneration through Sox2‐dependent Schwann cell sort‐
mus.88010​0 805 ing. Cell, 143(1), 145–155. https​://doi.org/10.1016/j.cell.2010.08.039
Miller, V. M., & Harman, S. M. (2017). An update on hormone ther‐ Passarella, S., & Karu, T. (2014). Absorption of monochromatic and nar‐
apy in postmenopausal women: Mini‐review for the basic scien‐ row band radiation in the visible and near IR by both mitochondrial
tist. American Journal of Physiology‐Heart and Circulatory Physiology, and non‐mitochondrial photoacceptors results in photobiomodula‐
313(5), H1013–H1021. https​://doi.org/10.1152/ajphe​art.00383. tion. Journal of Photochemistry and Photobiology B: Biology, 140, 344–
2017 358. https​://doi.org/10.1016/j.jphot​obiol.2014.07.021
Millesi, H. (2007). Bridging defects: Autologous nerve grafts. Acta Penas, C., & Navarro, X. (2018). Epigenetic modifications associated
Neurochirurgica Supplementum, 100, 37–38. to neuroinflammation and neuropathic pain after neural trauma.
MODRAK et al. |
      15

Frontiers in Cellular Neuroscience, 12, 158. https​://doi.org/10.3389/ syndrome. Neurology, 54(3), 603–607. https​://doi.org/10.1212/
fncel.2018.00158​ WNL.54.3.603
Pereira, J. A., Lebrun‐Julien, F., & Suter, U. (2012). Molecular mech‐ Sargin, D., Friedrichs, H., El‐Kordi, A., & Ehrenreich, H. (2010).
anisms regulating myelination in the peripheral nervous system. Erythropoietin as neuroprotective and neuroregenerative treatment
Trends in Neurosciences, 35(2), 123–134. https​://doi.org/10.1016/j. strategy: Comprehensive overview of 12 years of preclinical and clin‐
tins.2011.11.006 ical research. Best Practice & Research Clinical Anaesthesiology, 24(4),
Picascia, A., Grimaldi, V., Pignalosa, O., De Pascale, M. R., Schiano, C., 573–594. https​://doi.org/10.1016/j.bpa.2010.10.005
& Napoli, C. (2015). Epigenetic control of autoimmune diseases: Schindl, A., Schindl, M., Pernerstorfer‐Schön, H., & Schindl, L. (2000).
From bench to bedside. Clinical Immunology, 157(1), 1–15. https​://doi. Low‐intensity laser therapy: A review. Journal of Investigative
org/10.1016/j.clim.2014.12.013 Medicine, 48(5), 312–326.
Pockett, S., & Gavin, R. M. (1985). Acceleration of peripheral nerve re‐ Schumacher, M., Guennoun, R., Mercier, G., Désarnaud, F., Lacor, P.,
generation after crush injury in rat. Neuroscience Letters, 59(2), 221– Bénavides, J., … Baulieu, E. E. (2001). Progesterone synthesis and
224. https​://doi.org/10.1016/0304-3940(85)90203-4 myelin formation in peripheral nerves. Brain Research Reviews, 37(1–
Post, R., de Boer, K. S., & Malessy, M. J. (2012). Outcome follow‐ 3), 343–359. https​://doi.org/10.1016/S0165-0173(01)00139-4
ing nerve repair of high isolated clean sharp injuries of the ulnar Senger, J. L., Chan, K. M., Macandili, H., Chan, A. W. M., Verge, V. M. K.,
nerve. PLoS ONE, 7(10), e47928. https​://doi.org/10.1371/journ​ Jones, K. E., & Webber, C. A. (2019). Conditioning electrical stimula‐
al.pone.0047928 tion promotes functional nerve regeneration. Experimental Neurology,
Preston, D. C., & Shapiro, B. E. (2013). Anatomy and neurophysiology. 315, 60–71. https​://doi.org/10.1016/j.expne​urol.2019.02.001
Electromyography and neuromuscular disorders (3rd ed., pp. 8–18). Senger, J. L. B., Verge, V. M. K., Macandili, H. S. J., Olson, J. L., Chan, K.
London: W.B. Saunders. M., & Webber, C. A. (2018). Electrical stimulation as a conditioning
Raine, A. E., & Roger, S. D. (1991). Effects of erythropoietin on blood strategy for promoting and accelerating peripheral nerve regenera‐
pressure. American Journal of Kidney Diseases, 18(4 Suppl. 1), 76–83. tion. Experimental Neurology, 302, 75–84. https​://doi.org/10.1016/j.
Ren, Z., Wang, Y., Peng, J., Zhao, Q., & Lu, S. (2012). Role of stem cells expne​urol.2017.12.013
in the regeneration and repair of peripheral nerves. Reviews in the Shah, A., & Jebson, P. J. (2008). Current treatment of radial nerve palsy
Neurosciences, 23(2), 135–143. https​ ://doi.org/10.1515/revne​ following fracture of the humeral shaft. Journal of Hand Surgery,
uro-2011-0069 33(8), 1433–1434. https​://doi.org/10.1016/j.jhsa.2008.05.029
Rippon, H. J., & Bishop, A. E. (2004). Embryonic stem cells. Cell Proliferation, Shen, C. C., Yang, Y. C., & Liu, B. S. (2011). Large‐area irradiated low‐level
37(1), 23–34. https​://doi.org/10.1111/j.1365-2184.2004.00298.x laser effect in a biodegradable nerve guide conduit on neural regen‐
Robinson, L. R. (2000a). Traumatic injury to peripheral nerves. Muscle eration of peripheral nerve injury in rats. Injury, 42(8), 803–813. https​
and Nerve, 23(6), 863–873. https​://doi.org/10.1002/(SICI)1097- ://doi.org/10.1016/j.injury.2011.02.005
4598(20000​6)23:6<863:AID-MUS4>3.0.CO;2-0 Sherratt, R. M., Bostock, H., & Sears, T. A. (1980). Effects of 4‐aminopy‐
Robinson, L. R. (2000b). Role of neurophysiologic evaluation in diagnosis. ridine on normal and demyelinated mammalian nerve fibres. Nature,
Journal of American Academy of Orthopaedic Surgeons, 8(3), 190–199. 283(5747), 570–572. https​://doi.org/10.1038/283570a0
https​://doi.org/10.5435/00124​635-20000​5000-00006​ Siemionow, M., & Brzezicki, G. (2009). Chapter 8: Current techniques
Robinson, L. R. (2004). Traumatic injury to peripheral nerves. Supplements and concepts in peripheral nerve repair. International Review of
to Clinical Neurophysiology, 57, 173–186. Neurobiology, 87, 141–172.
Rochkind, S., Drory, V., Alon, M., Nissan, M., & Ouaknine, G. E. (2007). Smith, K. J., Felts, P. A., & John, G. R. (2000). Effects of 4‐aminopyridine
Laser phototherapy (780 nm), a new modality in treatment of long‐ on demyelinated axons, synapses and muscle tension. Brain, 123(Pt
term incomplete peripheral nerve injury: A randomized double‐blind 1), 171–184. https​://doi.org/10.1093/brain/​123.1.171
placebo‐controlled study. Photomedicine and Laser Surgery, 25(5), Sowa, Y., Imura, T., Numajiri, T., Nishino, K., & Fushiki, S. (2012). Adipose‐
436–442. https​://doi.org/10.1089/pho.2007.2093 derived stem cells produce factors enhancing peripheral nerve re‐
Rochkind, S., Leider‐Trejo, L., Nissan, M., Shamir, M. H., Kharenko, O., & generation: Influence of age and anatomic site of origin. Stem Cells
Alon, M. (2007). Efficacy of 780‐nm laser phototherapy on periph‐ and Development, 21(11), 1852–1862. https​ ://doi.org/10.1089/
eral nerve regeneration after neurotube reconstruction procedure scd.2011.0403
(double‐blind randomized study). Photomedicine and Laser Surgery, Sowa, Y., Kishida, T., Imura, T., Numajiri, T., Nishino, K., Tabata, Y., &
25(3), 137–143. https​://doi.org/10.1089/pho.2007.2076 Mazda, O. (2016). Adipose‐derived stem cells promote peripheral
Saag, K. G., Koehnke, R., Caldwell, J. R., Brasington, R., Burmeister, L. F., nerve regeneration in vivo without differentiation into Schwann‐like
Zimmerman, B., … Furst, D. E. (1994). Low dose long‐term corticoste‐ lineage. Plastic and Reconstructive Surgery, 137(2), 318e–330e. https​
roid therapy in rheumatoid arthritis: An analysis of serious adverse ://doi.org/10.1097/01.prs.00004​75762.86580.36
events. American Journal of Medicine, 96(2), 115–123. https​ ://doi. Stark, H., & Amirfeyz, R. (2013). Cochrane corner: Local corticoste‐
org/10.1016/0002-9343(94)90131-7 roid injection for carpal tunnel syndrome. Journal of Hand Surgery
Sabatier, M. J., Redmon, N., Schwartz, G., & English, A. W. (2008). (European Volume), 38(8), 911–914. https​://doi.org/10.1177/17531​
Treadmill training promotes axon regeneration in injured periph‐ 93413​490848
eral nerves. Experimental Neurology, 211(2), 489–493. https​ ://doi. Stolt, C. C., & Wegner, M. (2016). Schwann cells and their transcriptional
org/10.1016/j.expne​urol.2008.02.013 network: Evolution of key regulators of peripheral myelination.
Sakanaka, M., Wen, T. C., Matsuda, S., Masuda, S., Morishita, E., Nagao, Brain Research, 1641(Pt A), 101–110. https​://doi.org/10.1016/j.brain​
M., & Sasaki, R. (1998). In vivo evidence that erythropoietin protects res.2015.09.025
neurons from ischemic damage. Proceedings of the National Academy Straus, D. J. (2002). Epoetin alfa as a supportive measure in hematologic
of Sciences of the United States of America, 95(8), 4635–4640. https​:// malignancies. Seminars in Hematology, 39(4 Suppl. 3), 25–31. https​://
doi.org/10.1073/pnas.95.8.4635 doi.org/10.1053/shem.2002.36926​
Salzer, J. L. (2015). Schwann cell myelination. Cold Spring Harbor Sulaiman, W., & Gordon, T. (2013). Neurobiology of peripheral nerve in‐
Perspectives in Biology, 7(8), a020529. https​://doi.org/10.1101/cshpe​ jury, regeneration, and functional recovery: From bench top research
rspect.a020529 to bedside application. Ochsner Journal, 13(1), 100–108.
Sanders, D. B., Massey, J. M., Sanders, L. L., & Edwards, L. J. (2000). A ran‐ Sullivan, R., Dailey, T., Duncan, K., Abel, N., & Borlongan, C. V. (2016).
domized trial of 3,4‐diaminopyridine in Lambert‐Eaton myasthenic Peripheral nerve injury: Stem cell therapy and peripheral nerve
|
16       MODRAK et al.

transfer. International Journal of Molecular Sciences, 17(12), 2101. Wan, L. D., Xia, R., & Ding, W. L. (2010). Electrical stimulation enhanced
https​://doi.org/10.3390/ijms1​7122101 remyelination of injured sciatic nerves by increasing neurotrophins.
Sundem, L., Chris Tseng, K. C., Li, H., Ketz, J., Noble, M., & Elfar, J. (2016). Neuroscience, 169(3), 1029–1038.
Erythropoietin enhanced recovery after traumatic nerve injury: Wan, L., Xia, R., & Ding, W. (2010). Short‐term low‐frequency electrical
Myelination and localized effects. Journal of Hand Surgery, 41(10), stimulation enhanced remyelination of injured peripheral nerves by
999–1010. https​://doi.org/10.1016/j.jhsa.2016.08.002 inducing the promyelination effect of brain‐derived neurotrophic
Sunderland, S. (1951). A classification of peripheral nerve injuries produc‐ factor on Schwann cell polarization. Journal of Neuroscience Research,
ing loss of function. Brain, 74(4), 491–516. https​://doi.org/10.1093/ 88(12), 2578–2587. https​://doi.org/10.1002/jnr.22426​
brain/​74.4.491 Wenjin, W., Wenchao, L., Hao, Z., Feng, L., Yan, W., Wodong, S., …
Sunderland, S. (1990). The anatomy and physiology of nerve injury. Muscle Wenlong, D. (2011). Electrical stimulation promotes BDNF expres‐
and Nerve, 13(9), 771–784. https​://doi.org/10.1002/mus.88013​0903 sion in spinal cord neurons through Ca(2+)‐ and Erk‐dependent sig‐
Suslu, H., Altun, M., Erdivanli, B., & Turan, S. H. (2013). Comparison of naling pathways. Cellular and Molecular Neurobiology, 31(3), 459–467.
the effects of local and systemic dexamethasone on the rat traumatic https​://doi.org/10.1007/s10571-010-9639-0
sciatic nerve model. Turkish Neurosurgery, 23(5), 623–629. Williams, H. B. (1996). A clinical pilot study to assess functional return fol‐
Svaren, J., & Meijer, D. (2008). The molecular machinery of myelin gene lowing continuous muscle stimulation after nerve injury and repair in
transcription in Schwann cells. Glia, 56(14), 1541–1551. https​://doi. the upper extremity using a completely implantable electrical system.
org/10.1002/glia.20767​ Microsurgery, 17(11), 597–605. https​://doi.org/10.1002/(SICI)1098-
Taylor, C. A., Braza, D., Rice, J. B., & Dillingham, T. (2008). The incidence 2752(1996)17:11<597:AID-MICR6​>3.0.CO;2-M
of peripheral nerve injury in extremity trauma. American Journal Wirtz, P. W., Verschuuren, J. J., van Dijk, J. G., de Kam, M. L., Schoemaker,
of Physical Medicine & Rehabilitation, 87(5), 381–385. https​ ://doi. R. C., van Hasselt, J. G., … van Gerven, J. M. (2009). Efficacy of 3,4‐
org/10.1097/PHM.0b013​e3181​5e6370 diaminopyridine and pyridostigmine in the treatment of Lambert‐
Toma, J. G., McKenzie, I. A., Bagli, D., & Miller, F. D. (2005). Isolation and Eaton myasthenic syndrome: A randomized, double‐blind, placebo‐
characterization of multipotent skin‐derived precursors from human controlled, crossover study. Clinical Pharmacology and Therapeutics,
skin. Stem Cells, 23(6), 727–737. https​://doi.org/10.1634/stemc​ 86(1), 44–48. https​://doi.org/10.1038/clpt.2009.35
ells.2004-0134 Yazdani, S. O., Golestaneh, A. F., Shafiee, A., Hafizi, M., Omrani, H. A., &
Topilko, P., Schneider‐Maunoury, S., Levi, G., Baron‐Van Evercooren, Soleimani, M. (2012). Effects of low level laser therapy on prolifer‐
A., Chennoufi, A. B., Seitanidou, T., … Charnay, P. (1994). Krox‐20 ation and neurotrophic factor gene expression of human Schwann
controls myelination in the peripheral nervous system. Nature, cells in vitro. Journal of Photochemistry and Photobiology B: Biology,
371(6500), 796–799. https​://doi.org/10.1038/371796a0 107, 9–13. https​://doi.org/10.1016/j.jphot​obiol.2011.11.001
Toth, C., Martinez, J. A., Liu, W. Q., Diggle, J., Guo, G. F., Ramji, N., … You, S., Petrov, T., Chung, P. H., & Gordon, T. (1997). The expression of
Zochodne, D. W. (2008). Local erythropoietin signaling enhances re‐ the low affinity nerve growth factor receptor in long‐term dener‐
generation in peripheral axons. Neuroscience, 154(2), 767–783. vated Schwann cells. Glia, 20(2), 87–100. https​://doi.org/10.1002/
Tseng, K. C., Li, H., Clark, A., Sundem, L., Zuscik, M., Noble, M., & Elfar, (SICI)1098-1136(19970​6)20:2<87:AID-GLIA1​>3.0.CO;2-1
J. (2016). 4‐Aminopyridine promotes functional recovery and Zhu, T. S., & Glaser, M. (2008). Neuroprotection and enhancement of re‐
remyelination in acute peripheral nerve injury. EMBO Molecular myelination by estradiol and dexamethasone in cocultures of rat DRG
Medicine, 8(12), 1409–1420. https​://doi.org/10.15252/​e mmm. neurons and Schwann cells. Brain Research, 1206, 20–32. https​://
20150​6 035 doi.org/10.1016/j.brain​res.2008.02.051
van den Elsen, P. J., van Eggermond, M. C., Puentes, F., van der Valk, P., Ziegler, L., Grigoryan, S., Yang, I. H., Thakor, N. V., & Goldstein, R. S.
Baker, D., & Amor, S. (2014). The epigenetics of multiple sclerosis and (2011). Efficient generation of Schwann cells from human embryonic
other related disorders. Multiple Sclerosis and Related Disorders, 3(2), stem cell‐derived neurospheres. Stem Cell Reviews and Reports, 7(2),
163–175. https​://doi.org/10.1016/j.msard.2013.08.007 394–403. https​://doi.org/10.1007/s12015-010-9198-2
Van Diemen, H. A., Polman, C. H., Koetsier, J. C., Van Loenen, A. C.,
Nauta, J. J., & Bertelsmann, F. W. (1993). 4‐Aminopyridine in patients
with multiple sclerosis: Dosage and serum level related to efficacy S U P P O R T I N G I N FO R M AT I O N
and safety. Clinical Neuropharmacology, 16(3), 195–204. https​://doi.
org/10.1097/00002​826-19930​6000-00002​ Additional supporting information may be found online in the
van Lunteren, E., & Moyer, M. (2004). Combination of variable frequency Supporting Information section at the end of the article.
train stimulation and K+ channel blockade to augment skeletal
muscle force. IEEE Transactions on Neural Systems and Rehabilitation
Transparent Peer Review Report
Engineering, 12(2), 288–294.
Walsh, S. K., Gordon, T., Addas, B. M., Kemp, S. W., & Midha, R. (2010).
Skin‐derived precursor cells enhance peripheral nerve regeneration
How to cite this article: Modrak M, Talukder MAH,
following chronic denervation. Experimental Neurology, 223(1), 221–
228. https​://doi.org/10.1016/j.expne​urol.2009.05.025 Gurgenashvili K, Noble M, Elfar JC. Peripheral nerve injury
Walsh, S., & Midha, R. (2009). Use of stem cells to augment nerve in‐ and myelination: Potential therapeutic strategies. J Neuro Res.
jury repair. Neurosurgery, 65(Suppl. 4), A80–A86. https​://doi. 2019;00:1–16. https​://doi.org/10.1002/jnr.24538​
org/10.1227/01.NEU.00003​35651.93926.2F

You might also like