Abstract
Traumatic peripheral nerve injury represents a major clinical and public health problem that often leads to significant functional impairment and permanent disability. Despite modern diagnostic procedures and advanced microsurgical techniques, functional recovery after peripheral nerve repair is often unsatisfactory. Therefore, there is an unmet need for new therapeutic or adjunctive strategies to promote the functional recovery in nerve injury patients. In contrast to the central nervous system, Schwann cells in the peripheral nervous system play a pivotal role in several aspects of nerve repair such as degeneration, remyelination, and axonal growth. Several non-surgical approaches, including pharmacological, electrical, cell-based, and laser therapies, have been employed to promote myelination and enhance functional recovery after peripheral nerve injury. This review will succinctly discuss the potential therapeutic strategies in the context of myelination following peripheral neurotrauma.
Keywords: peripheral nerve injury, therapeutic strategies, physical therapy, pharmacotherapy, myelination, epigenetics, limitations
Traumatic peripheral nerve injury (TPNI) represents a major clinical and public health problem that often leads to significant functional impairment and permanent disability.1 It is estimated that roughly 3% of all trauma patients have peripheral nerve injuries2 and more than 50,000 peripheral nerve repair procedures are performed annually in the United States alone.3 TPNIs are generally associated with motor vehicle collisions, penetrating injuries, lacerations, gun-shots, falls, burns, fractures, ischemia, traction and crush injuries,4 and have been studied in both civilian2,5–7 and military populations.8 TPNIs are also more common in children than previously identified and can result in major social and economic burdens.9 Despite progress in understanding the pathophysiology and the biological factors involved in of nerve injury and recovery, as well as the availability of modern diagnostic procedures and advanced microsurgical techniques, few treatment options exist to alter the course of recovery after TPNI.4,10 This review will discuss the current knowledge of TPNI and recovery in terms of pathophysiology of nerve injury, classification, regeneration, diagnostic consideration and therapeutic strategies. The main focus is to provide a better understanding of potential non-surgical therapeutic approaches in promoting myelination following peripheral neurotrauma.
Pathophysiology and classification of nerve injury
At the cellular level, TPNIs exist in varying severities, which relate directly to the classification of these injuries. The severity of injury is related to the extent of anatomic disruption to the constituents of the nerve. On an anatomic level, the nerves grossly consist of axons, myelin (in myelinated fibers), Schwann cells, blood vessels, fat and connective tissue.10–12 The outer covering of the nerve is a connective tissue epineurium, which is the site of most microsurgical techniques for repair. Within the nerve there are fascicles embedded in an internal connective tissue epineurium. Each individual fascicle is surrounded by perineurial connective tissue, and each fascicle contains many nerve fibers, which in turn, are encapsulated by longitudinally oriented endoneurial tubes. Nerves with fewer and larger fascicles may be more vulnerable to injury as a result of their inherent deficiency in connective epineurial tissue.11,12 Some fibers within any given fascicle contain myelin which is compact in distribution around internodes, in contrast to the less myelin-dense paranodal and myelin-deficient nodes of Ranvier, where sodium channels are concentrated allowing propagation of action potentials (saltatory conduction). In practice, injuries to connective tissue, myelin, and axons all comingle in most TPNI.
The two commonly used classifications of nerve injury are the Seddon and Sunderland Classifications.11,13 Seddon’s scheme, first published in 1943, divides nerve injuries into neurapraxia, axonotmesis, and neurotmesis in order of increasing severity. Neurapraxia, the mildest class of nerve injury, is characterized as a conduction block from a seemingly isolated injury to myelin, usually 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 injuries to other tissue types may also play a role. Nonetheless, neuropraxic injuries are believed to spare most, if not all axons and these injuries often result from compression or traction on the nerve. In axonotmesis, the hallmark is axonal disruption and resulting Wallerian degeneration, but some connective tissue structures are preserved. Axonotmesis is typical after peripheral nerve crush and stretch injuries. Neurotmesis, where the nerve is severed, is the most severe class of nerve injury and is often caused by substantial trauma to the nerve and adjoining structures.11–13
Sunderland’s classification breaks down nerve injury into five categories:12 the first degree of nerve injury corresponds to neurapraxia, here with the distinction that the distal segment of the nerve, which is outside of the zone of injury, can conduct normally. Here too, the axonal structure is believed to be preserved. The second degree of nerve injury consists of axon degeneration distally with preserved endoneurial tubes with a good chance of near normal recovery if the injuries are not too distant from the targets. The third degree of nerve injury consists of the destruction of endoneurial 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 fascicular and perineurial destruction with only epineurium spared. The fifth degree of nerve injury is of complete loss of nerve continuity and complete destruction of axon and surrounding connective tissue, which corresponds to neurotmesis in Seddon’s classification. In neurotmesis the prognosis of spontaneous recovery is poor, and nerve continuity is believed unlikely or even impossible without surgical intervention.
Regeneration
Recovery of nerve function after injury depends on several factors including the degree and location of the nerve injury and patient related factors. The degree of nerve injury itself depends on number and size of fascicles within the injured nerve (e.g. partial vs. complete severance of fascicles). Not all nerves have the same capacity for recovery, and therefore the type and location of nerve injured (e.g. proximal vs distal nerve segment, mixed nerve, etc.), are critical prognostic factors for regeneration.4,12,14,15 Finally, advanced patient age and the chronicity of the injury each negatively affect the capacity for regeneration with older patients and those with delayed presentations or intervention often faring most poorly. Regeneration and repair processes of injured nerve occur at multiple levels including the nerve cell body, the segment between the neuron and the injury site (proximal stump), the injury site itself, the segment between the injury site and the end organ (distal stump), and the end organ.4 One critical factor in regeneration is the process of Wallerian degeneration, which affects the segment of nerve distal to transected axons. This process starts within 48 hours and is usually complete by day 7–9 for motor axons and by day 11 for sensory nerves, with the difference in times ascribed to special consequences to dysfunction and early failure in neuromuscular transmission in motor fibers. Wallerian degeneration itself is an early means to distinguish neurapraxic from axonal injuries provided electrodiagnostic studies are performed after this period.
There are generally accepted three mechanisms of repair: remyelination, collateral sprouting, and axon regrowth.4,16 In neurapraxic lesions, Schwann cells must de-differentiate to a stage where cell division is possible to proliferate and make new myelin, and this stage may take three months. Beyond this time period, persistent deficits imply axonal damage. The partial axon lesions recover through the mode of collateral sprouting that can take up to six months. Terminal sprouts are chemotrophic outgrowths from the intact nerve endings which grow from the conical growth-cone which emanates often from myelin-free nodes of Ranvier within the intramuscular nerves.1 This process sacrifices single fiber motor-specificity by placing more muscle fibers under the control of remaining axons. Therefore, the motor unit size (the number of muscle fibers dependent on one nerve fiber), can increase fivefold through the process of collateral sprouting.14 In complete lesions, recovery depends on an axon regrowth rate of approximately 1 mm/day, with some variation in this rate related to patient factor (age) and injury factors (injury site, mechanism of injury, proximity of the injury to the nerve cell body, time and type of repair),14,15 with the fastest reported regeneration rates associated with proximal limb and younger patients.12,14
During the regeneration process, the gap caused by the retracted stumps of a fully transected nerve is bridged by new tissue termed as “nerve bridge” and the regrowing axons from the proximal stump follow blood vessels through newly formed nerve bridge to reach the distal stump and their target organs.17,18 However, there can be scar formation within the nerve bridge that can lead to misdirection and aberrant regeneration of the sprouting axons.4 Therefore proximal lesions and those that must regenerate through scar hold the worst prognosis owing to the time lost during which reversible target muscle denervation atrophy becomes irreversible (12–18 months) in axotomy state.4,19 In the most severe forms of axonotmeses and neurotmeses (Sunderland class 3–5), recovery is encumbered by increasing disorganization of fascicular structure and scar formation inside the nerve. This is perhaps related to the random and disorganized growth of regenerating axons towards the end-organ.1,12 This may predispose more severe injuries to incomplete recovery of muscle function. Endoneurial tubes, Schwann cells and muscle fibers may indeed be viable for 12–18 months after prolonged axotomy;1,4,19 however, motor recovery is rarely possible after this time period even with surgical repair because of reduced capacity of motor axons to regenerate.16 Sensory fibers, without their inherent dependency on a viable muscle end-organ, may enjoy a slightly longer period of viable recovery.19
Electrodiagnostic evaluation of TPNI
The nature of the cellular and structural injury in the nerve defies imaging in most scenarios, and electrodiagnostic testing (EDX) is currently the most sensitive and specific method to evaluate TPNI. However, EDXs such as nerve conduction study (NCS) and electromyography (EMG) depend critically on the time-dependent post injury sequelae to the nerve and end-organ denervation, for a sensitivity that evolves over time after the injury. NCS determines nerve conduction velocities to help evaluate axonal degeneration from demyelinating disorders and EMG evaluates the motor unit action potential (MUAP) of a motor unit. Unlike surgical intervention, which can give some immediate information about the nature of the nerve injury, EDX has the key benefit of offering information about the functional status of the nerve and distally innervated units and carries far less risk of damage to the nerve or perioperative complications. It is for these reasons EDX plays a crucial ongoing role in the evaluation of these patients. The main aim of EDX in TPNI is to classify the pathophysiology of nerve injury as to axonal or demyelinating in nature, as well as to identify the location, grade and prognosis of the injury. This information is crucial in deciding the plans for an intervention. Despite the importance of these studies, it is also crucial to recognize the limitations and time dependency of EDX data, which typically include information from distinct NCSs and EMG studies obtained in a single session.
Neurapraxia and axonotmesis are indistinguishable using EDX testing for the first few days after nerve injury even when information is combined for sensory and motor nerves. The distal segment of an injured nerve remains excitable during this time, and therefore EDX may not provide sufficient information to inform prognosis for approximately 11 days after injury. Testing, during this period can nonetheless aid in localization of the injury and grading severity with the severest lesions (whether neurapraxic or axonotmetic) failing to evidence MUAP recruitment. In neurapraxia, the distal compound muscle action potential (CMAP) is normal, but stimulation proximal to the lesion will not result in responses in the case of complete conduction block. EMG in these cases will show no MUAP (indicating a complete neurapraxic lesion) or reduced recruitment (indicating a partial neurapraxic lesion) – all without abnormal spontaneous activity. These changes usually resolve by 12 weeks, but delayed conduction may persist owing to thinner and shorter internodes associated with remyelination.4,20 In axonotmesis, distal CMAP may be decreased or absent, but only after 7–9 days for motor and 11 days for sensory fiber injury. A decrease in amplitude of CMAP is proportional to the degree of axonal loss. EMG examination will show reduced recruitment or absent MUAP in the case of neurotmesis. MUAP recruitment can also serve to indicate axonal continuity after Wallerian degeneration is complete. Abnormal spontaneous activity will begin approximately 2–3 weeks after injury however; the extent of this activity may not correlate with severity.20 There are some notable shortcomings of commonly used EDX techniques. EDX does not allow the critical distinction of subtypes of intermediate (Sunderland class 3–5) injuries.1 The absent MUAP recruitment can be erroneously ascribed to loss of axonal continuity in mixed neurapraxic and axonotmetic lesions when NCS-EMG is performed before remyelination takes place.
Reinnervation changes first appear 2–3 months after axonotmesis and are typified by nominal increases in CMAP amplitude, however are probably better predicted based on MUAP configuration. Collateral sprouting results in longer duration polyphasic complexes and unstable motor units in the subacute phase followed by large amplitude and long duration motor unit potentials with persistently reduced recruitment. The very first sign of axon regrowth can be nascent units, which are small, polyphasic motor unit potentials.20
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 obtained once denervation has taken place with EDX yielding more definitive answers over time after TPNI.4 Studies performed within a week of injury allow localization of the injured segment in addition to some information about the grade or completeness of the injury. After one week, studies that focus on motor nerves can distinguish between axonotmesis and neurapraxia, in addition to the information about completeness of the injury. This same distinction is possible for sensory fibers at 11 days post-injury. Studies performed at 3–4 weeks allow all of the above distinctions in addition to assessment of abnormal spontaneous activity. Studies performed at 3–4 months allow the documentation of reinnervation.
Therapeutic strategies
Currently the treatment of choice for peripheral nerve injury that results in laceration is advanced microsurgical end-to-end repair with tensionless epineurial sutures or autologous nerve grafting where end-to-end anastomosis is not possible (Table 1).21–24 However, functional recovery after peripheral nerve repair is often unsatisfactory and it is apparent that microsurgical approaches fail to address the complex cellular and molecular events associated with TPNI. Therefore, there is an unmet need for new therapeutic or adjunctive strategies to promote functional recovery in TPNI patients. In contrast to the central nervous system (CNS), Schwann cells in the peripheral nervous system (PNS) produce a growth-permissive environment for the injured nerve and play a pivotal role in several aspects of nerve repair such as degeneration, remyelination, and axonal growth.7,16 Several treatment strategies have been employed to enhance the recovery process after TPNI, including pharmacological, electrical and cell based therapies.7,25–27 While each has shown some promise in treating patients, none has provided a single universally applicable cure for the consequences of TPNI and they are also not without significant pitfalls or potent side-effect profiles. Surgical interventions have been extensively reviewed by several authors;4,24,28,29 this section will highlight the usefulness of novel non-surgical therapeutic approaches in promoting myelination following peripheral neurotrauma (Table 1).
Strategies | Intervention | Timing of Intervention | Benefit | References | Limitations/Comments/Next step |
---|---|---|---|---|---|
Surgical: | End-to-end neurorrhaphy Nerve grafting Nerve transfer Conduit repair |
Variable (Days to months) | Yes | 4,21–24,28,29 | For selected nerve laceration injuries Slow rate of nerve regeneration Sub-optimal functional outcome Causes of poor outcome unknown Possible benefit from adjunct therapy |
Non-Surgical: Physical agents | Physical therapy Electrical stimulation Low-level laser therapy Therapeutic ultrasound |
Variable (Days to weeks) | Yes | 100–105,107–118,121,123,125,147–149,152–156,159,160 | Mainly for post-surgical rehabilitation Device-dependent Compliance issues Requires user-friendly portable devices Large clinical studies required |
Pharmacological agents | Steroid hormones Erythropoietin 4-Aminopyridine |
Immediate/Early | Yes | 30–34,38,44–46,51–53,56,57,60,65–68, 86–90 | Mostly pre-clinical findings Effective on crush and laceration injuries Improves motor function Improves myelination Improves nerve conduction Reduces muscle atrophy Proof-of-concept clinical studies required |
Miscellaneous | Cell-based therapy Epigenetic target |
Unknown | Unknown | 131,134,137–141,144,146,170,172–174,176–183 | Limited in vitro findings Proof-of-concept pre-clinical studies required Proof-of-concept clinical studies required |
For more information, please refer to the listed references.
Pharmacotherapy
At present, there is no clinically approved pharmacological agent available for the treatment of TPNI. However, several agents have reported to be potential candidates to improve nerve regeneration by promoting myelination in the peripheral nerves.
Steroid Hormones
Steroid hormones, such as estrogen and progesterone, have been investigated for use in TPNI,30–32 with both in vivo and in vitro evidence demonstrating the receptors for these hormones in the constituent cells of the peripheral nerve such as Schwann cells, DRG neurons, sensory and autonomic neurons.27 Both hormones are known to be neuroprotective and promote myelination, with estrogen (17β-Estradiol) promotes nerve recovery in both the central and peripheral nervous systems.33,34 In the PNS, estrogen exerts its effects through the modulation of steroid nuclear receptors, which in turn upregulate the PI3K/AKT/mTOR signaling pathway and thereby promote myelination.32 It is reported that PNS dorsal root ganglion neurons activate mTOR following a sciatic nerve injury and this activity enhances axonal growth capacity with increased expression of growth-associated protein, GAP-43.35 Activation of mTOR can also lead to downstream phosphorylation of S6 ribosomal protein and 4EBP-1 to initiate protein synthesis.36 Since GAP-43 plays a key role in axon sprouting and outgrowth in regenerating axons,37 it is possible that GAP-43 is involved in estrogen-induced upregulation of mTOR signaling and myelination. Progesterone may act in an independent manner following binding to its specific progesterone receptor, which in turn induces myelination via upregulation of Krox20 (also termed early growth response gene: Egr2),38 a transcription factor that regulates PNS myelination.39 This activation directly drives expression of myelin proteins like MPZ and PMP22, both of which are primary constituents of the myelin sheath.34,38 However, both estrogen and progesterone suffer from well-documented side-effect profiles which can differ from patient to patient depending on the dose and duration of drug treatments. The United States Food and Drug Administration mandated a “black box” warning on all estrogen products based on the results of the Women’s Health Initiative (WHI) because unopposed estrogen use can increase the risk of endometrial cancer in intact uteri, invasive breast cancer in post-menopausal women, and deep vein thrombosis (DVT).40 Additional risk factors for estrogen include hypertension, obesity, diabetes mellitus, tobacco use, and/or history of venous thromboembolism. Estrogen receptor signaling are also involved in prostate carcinogenesis.41 In WHI studies, an increased risk of DVT, pulmonary embolism, stroke, and myocardial infarction was observed in women ≥65 years with daily conjugated estrogens combined with medroxyprogesterone.
On the other hand, corticosteroids are sometimes used as a treatment option for nerve injury, including acute spinal cord injury, facial nerve paralysis, compressive neuropathy, and neuropathic pain,42–45 A recent clinical study also demonstrated an important role for oral corticosteroids in the recovery of motor and sensory function following iatrogenic nerve injuries in total hip and knee arthroplasty.46 However, several retrospective reviews have shown that long-term glucocorticoid use, even in low doses, is a significant independent predictor of diverse adverse effects, including bone loss or fracture, serious infections, gastrointestinal bleeding or ulcer, and cataracts.47–50 The risk incurred with glucocorticoid use is both dose- and duration-dependent. Although the generalized use of hormonal therapies has not been common in the treatment of TPNI, several pre-clinical studies have shown that topical steroids can improve functional recovery and morphometric indices of the injured peripheral nerve51–53 and this may have clinical implications after nerve transection because of its fewer or no adverse effects compared to chronic systemic administration. Therefore, the potential clinical usefulness of topical steroids to reduce post-injury nerve dysfunction warrants further investigations.
Erythropoietin
Erythropoietin (EPO), an endogenous hormone and FDA-approved drug for the treatment of anemia, has neuroprotective properties in both the central and peripheral nervous system.54–57 Although originally discovered as hematopoietic agent, EPO has been extensively studied in translational neuroscience and its role in neuroprotection/neuroregeneration has been elegantly reviewed by several authors.27,58–60 It is documented that EPO and its receptor (EPOR) are present in a wide variety of non-erythroid cells throughout the body and may impact many biological functions. In the nervous system, EPO is produced and secreted by the neurons of hippocampus, cortex, internal capsule, midbrain, and nervous system tumors.61–63 The EPOR is also expressed on the myelin sheath of radicular nerves in human PNS.64 We and others have shown the presence and upregulation of EPOR at the site of peripheral nerve injury in mice.65,66 EPO improves the sciatic function index of mice and rats after crush injury,57,65–68 and this functional improvement was observed with EPO administration prior to injury, immediately after injury, and after 1 week suggesting that the timing is not critical.57,65–68 Importantly, EPO mediated functional improvement can be demonstrated from single doses up to days after injuries in mice and seems to correlate with injury.65 Although the mechanisms by which EPO works are still poorly understood, it is thought to work by promoting the expression of myelin genes, MPZ and PMP22.69 We have shown that mice which received systemic EPO following nerve crush injury maintained more myelinated axons at the site of injury.57 In vitro EPO treatment also promoted myelin formation and protected myelin from the effect of nitric oxide (NO) exposure in co-cultures of Schwann cells and dorsal root ganglion cells.57 In addition to the fundamental myeloprotective role of EPO, these findings also demonstrated an anti-oxidative effect of EPO at the site of nerve injury which is consistent with the direct role of EPO against oxidative stress.59 Given EPO’s widespread and frequent use to treat anemia in humans and its favorable side-effect profile,70,71 early clinical trials have already begun investigating EPO’s therapeutic potential treating human peripheral nerve trauma associated with joint replacement surgery.46,72 It is also noteworthy that clinical use of EPO is not without problems, in particular the route of administration and the potent side-effect profile. Aside from blood hyper-viscosity from increased RBC production, hypertension and its related problems,73,74 the most common side effects of EPO therapy are headache and an influenza-like syndrome.75,76
4-aminopyridine
4-aminopyridine (4-AP), a broad-spectrum potassium (K+) channel blocker and FDA-approved drug for the symptomatic treatment of multiple sclerosis (MS),77,78 that has been shown to improve neuromuscular function in patients with other demyelinating disorders including myasthenia gravis,79 spinal cord injury,80 and Lambert-Eaton syndrome.81,82 The neurological benefits of 4-AP are believed to result from increases in action potential duration, calcium influx, neurotransmitter release, synaptic transmission, and direct effects on muscle.83–85 4-AP may enhance cell-membrane excitability and impulse conduction, and we observed that clinically relevant doses of 4-AP treatment beginning shortly after injury enhance global functional recovery, promote remyelination and improve nerve conduction velocity in an established mouse model of peripheral nerve crush injury.86 We also showed that 4-AP can distinguish or classify a crush injury from a transection injury by transient motor function recovery,86–88 an effect most likely related to its nerve conduction restoration properties because the time course of recovery was far too rapid to be explained by regenerative mechanism and it is demonstrable at time-points far earlier than those allowed by EDX (see above). Importantly, at the tissue level, 4-AP also appears to be myeloprotective and possibly axonoprotective.86,87 Early studies with 4-AP showed an increase in action potential duration and amplitude when measured on experimentally demyelinated mammalian peripheral nerve fibers.89,90 These findings highlight a possible diagnostic and therapeutic contribution to recovery and prognosis owed to excitatory molecules that may stabilize impulse conduction within hours to days after injury.91–93 Ongoing work on clinical translation of these findings may reveal clinical indications to TPNI, both in diagnosis and treatment.94 However, despite the beneficial effects, the clinical ability of 4-AP to restore function has been limited because of its narrow therapeutic window, the need for frequent dosing throughout the day, and significant adverse side effects such as anxiety, tremors and seizures.95–97 Clinical trials have shown that the efficacy of 4-AP is related to the total drug exposure whereas the toxicity is related to the peak serum levels.98,99 We have demonstrated that long-term treatment is beneficial with 4-AP following acute TPNI improving functional recovery, myelin thickness, as well as important parameter of EDX, such as NCV.86,87 Our findings with 4-AP convincingly demonstrated that it can be used either as local, transdermal or injectable form to exert its beneficial effect on TPNI recovery.86–88 Given the interplay between clinical efficacy, safety profile, and patient compliance, further developments of safe methods of administering 4-AP are a reasonable focus in the near term to allow treatment of the demyelinating component of TPNI, despite its promise as a regenerative medicine agent.
Electrical stimulation
Electrical stimulation (ES) represents a promising non-pharmacological approach to accelerate and promote recovery following peripheral nerve injury.100–105 Studies in animals and humans have shown that ES promotes preferential reinnervation of both motor and sensory neurons, allowing for a faster recovery.100,106,107 In addition to promoting reinnervation, ES also aids in the remyelination process following peripheral nerve injury.108 ES is also reported to provide benefit on nerve injury-induced muscle atrophy and function.109–111 Like ES, treadmill running is reported to exert positive effects on nerve regeneration and functional recovery.112,113 Moreover, the combination of these two activity-dependent therapies, ES and treadmill running, has been shown to exert positive synergistic effects on nerve regeneration and muscle reinnervation.114,115 While post-operative direct low-frequency (20 Hz) ES of the proximal nerve stump for 1 hour (2- to 3- fold threshold current) is a standard regimen,116 experimental studies have demonstrated that the same ES parameters seven days prior to axotomy (conditioning ES, CES) can promote functional nerve regeneration with upregulation of regeneration-associated genes.117,118 Although the exact mechanisms by which ES and exercise enhance nerve regeneration are poorly defined, cyclic adenosine monophosphate (cAMP) and brain-derived neurotrophic factor (BDNF) are reported to play key roles.119–121 ES causes an increased influx of calcium into the neurons followed by an increase in intracellular cAMP levels,120,122 and the downstream signaling molecule of cAMP, protein kinase A, promotes the expression of regeneration-associated genes for axonal growth.120 Calcium-induced phosphorylation of extracellular signal regulated kinase, ERK, has been shown to cause increased expression of BDNF and myelination in the peripheral nervous system.123,124 Despite the reported benefits with ES, the clinical use of implantable electrical devices remains a challenge because of the nature of invasive procedures, the lack of proper controls, and patient dissatisfaction with discomfort.125
Cell-based therapy
Some of the severest injuries to peripheral nerves, such as avulsions, lacerations, and contusions may suffer from the additional loss of Schwann cells needed to mediate regeneration. Such injuries may fail to regenerate even at the accepted rate of 1 mm/day.16 With this slower rate of regeneration, end organ reinnervation may take months or years or may fail eventaully.16,19 In the chronic axotomy state, the denervated Schwann cells eventually lose their capacity to support growth of the neurons and the lack of healthy Schwann cells is an important issue in nerve regeneration, which spurs interest in cell-based therapy.16,19,126,127
Cell-based therapy is a promising branch of regenerative medicine and Schwann cell cultures have demonstrated favorable results in the experimental model of TPNI with regeneration and remyelination.128,129 However, the process of human Schwann cell collection involves invasive nerve biopsy and the culture also has limited in vitro expansion.130 In search for a suitable cell line, stem cells (SCs) have garnered substantial interests as candidate transplant cells because of their availability, rapid in vitro expansion, survival and integration within the host tissue.131 Based on the differentiation potential, there are three categories of SC therapy: totipotent, pluripotent, and multipotent stem cells. Pluripotent and multipotent stem cells have been the focus of most research to date.132
Embryonic stem cells (ESCs) are pluripotent SCs that can differentiate into all three germ layers and Schwann cells can be generated from ESCs with 60% efficiency.133 In an animal model, injection of ESCs translated to improved nerve repair and functional ability.134 ESCs are limited in their therapeutic potential as they are in short supply, owing to their source in the human embryo and they also carry a risk of teratoma formation.135 Human induced pluripotent stem cells (hPSCs) have shown some promise in the regeneration and protection of myelin, possibly by providing an exogenous source of self-renewing Schwann cells.136 Multipotent somatic stem cells from bone marrow, mesenchymal stem cells (MSCs), have also shown some promise with greater number of myelinated axons when used in combination of artificial conduits and acellular grafts,131,137,138 and they are also capable in myelinating cultured PC12 cells in vitro.139
In addition, adipose tissue and skin have been reported to provide easily accessible and less invasive potential sources of SCs. Adipose-derived stem cells (ADSCs) are a form of multipotent stem cells that can differentiate into a Schwann-like cell with similar functional properties.140 It is possible that ADSCs may exert their effects via the release of growth factors, such as nerve growth factor, vascular endothelial growth factor, and BDNF, as well as through the recruitment of endogenous Schwann cells.141,142 Although ADSCs are one of the more attractive SC therapies due to their availability in the adipose tissue, one restriction is their tendency to differentiate toward adipocytes, which can hinder nerve recovery.143 The skin and associated structures have a highly available supply of transplantable cells due to their ability to differentiate into Schwann-like cells.7,131 Skin-derived precursor cells (SKPCs) are neural crest-related precursor cells found in the dermis, which are capable of in vitro differentiation into neural crest cells, including those with the features of Schwann cells and peripheral neurons.144–146
While there is still much to learn about the role of SCs and their therapeutic potential in TPNI, there is now exciting in-vivo and in-vitro evidence indicating that they may be efficacious myelinating phenotypes and localization can be maintained. Most importantly, clinical studies exploring the feasibility of cell-based strategies as an adjunct therapy in chronic nerve injury will require careful investigation to determine the amount and method of cell delivery and the fate (cell survival and differentiation) of transplanted cells for safety, reliability, and maximum efficacy.
Photobiomodulation with laser therapy
Photobiomodulation (PBM) with low-level laser therapy (LLLT) has been extensively studied in many clinical practices including physical medicine and rehabilitation, stroke, degenerative or traumatic brain disorders, and nerve repair.147–149 Laser therapy is a noninvasive treatment modality that induces a photochemical reaction in the cell and increases the DNA and RNA synthesis in the cell nucleus, with subsequent cell proliferation and protein synthesis, including changes in nerve cell action potential.147,148 The precise mechanisms underlying PBM with LLLT and its therapeutic benefits are not fully understood. It is believed that the initial trigger of PBM is the absorption of light (photons) by cytochrome C oxidase in the mitochondrial respiratory pathway. The increased activity of cytochrome C oxidase in turn increases the production of adenosine triphosphate (ATP) and thus modulates the cell functions.150,151 Both clinical and experimental studies have reported the beneficial effects of LLLT in TPNIs. Animal and in vitro studies have shown that LLLT promotes the regeneration and functional recovery of the injured peripheral nerve, accelerates the myelination of regenerated nerves, increases the axonal diameter, and stimulates Schwann cell proliferation.152–158 Double-blind randomized studies have reported that post-operative LLLT can enhance the regenerative process of the peripheral nerve with increased number of myelinated axons159 and improve the motor nerve function with functional recovery.160 However, other studies did not observe any beneficial effects of LLLT.161,162 Although LLLT has no reported adverse effects, there is no standardization in treatment with LLLT and different irradiation parameters have been used in different models of PNI, and there is also a paucity in the clinical trials with LLLT. Therefore, future studies exploring the effects of different variables, such as wavelengths, dose, continuous or pulsed mode, application site, and type of radiation would verify the usefulness of LLLT in TPNI as an adjunct therapy.
Epigenetics and small molecule alterations in myelination
There is growing evidence that the interplay of environmental risk factors and individual genetic susceptibility modulates disease presentation and therapeutic responsiveness.163–165 Epigenetics offers a promising link between genetic and environmental influences on phenotype development.163–167 Epigenetic modifications, which include DNA methylation, posttranslational modifications of histones, and non-coding RNAs, results in the heritable silencing of genes without a change in their coding sequence.167,168 Such modifications can be induced by stress, tissue damage, and diseases, and can affect both physiological and pathological processes.166,169–171 Epigenetic mechanisms thus play an essential role in transcriptional control of genes, maintenance of cellular identity, cell activation, and cellular repair processes during stress; with purported roles in many disease processes including autoimmune disease, multiple sclerosis, neuroinflammation, cancer, and normal aging.169–171
The ever-evolving field of epigenetics also has the potential to play an important role in peripheral nerve recovery and myelination.172–175 Myelination by Schwann cells is under strict transcriptional control176,177 involving sequential, feedforward cascades of promyelinating transcription factors where Sox 10 (SRY-related HMG-box-10) and Oct6 (octamer-binding transcription factor-6) synergistically induce the expression of Krox20 (also termed early growth response gene: Egr2). Krox 20 is considered a master regulator of PNS myelination because it activates many myelin genes, suppresses myelination inhibitors, and maintains the myelinated state.39 Krox20 together with NAB (NGFI-A/Egr-binding) protein regulates the transcription of myelin structural proteins and biosynthetic components of myelin lipid layer.178,179 In addition, several epigenetic and chromatin modifiers are involved in myelination of mammalian nervous system and are crucial for SC differentiation, myelin formation and myelin maintenance.180,181 For example, histone deacetylase (HDAC) remodels chromatin and condenses chromatic architecture and thus limits DNA access for transcription factors.182 Recently, an HDAC3-dependent pathway was identified as a potent inhibitor of peripheral myelogenesis.183 Both in vitro and in vivo studies demonstrate that addition of HDAC3 inhibitors or the ablation of HDAC3 in Schwann cells significantly increases the production of myelin, conduction velocity, CMAP amplitude, and enhances sensory and motor function.183 HDAC3 also antagonizes the myelogenic neuregulin-PI3K-AKT signaling axis. While these findings highlight the therapeutic potential of transient HDAC3 inhibition for improving peripheral myelin repair, it is unknown how different members of the HDAC family interact, their potential compensatory mechanisms, and how their expression and activity are regulated.182
It is evident that epigenetic changes can give rise to several significant disorders and we are just at the beginning of learning and understanding the contributions of these molecular genetic alterations to human diseases. While the reversible nature of epigenetic alterations is encouraging in the effort to find therapies that can reverse the molecular silencing early, it is unknown whether the epigenetic modifications are the cause or the result of disease progression. Future epigenetic studies in TPNIs will certainly enrich our knowledge and pave the way to use epigenetics as a tool to identify a disease biomarker and the potential therapeutic target.
Future directions and conclusions
In the past three decades, tremendous advances were made in TPNI diagnosis and management. However, the failure of well-defined microsurgical techniques to provide satisfactory functional recovery in the presence of complex cellular and molecular events with TPNI raises the question of using more robust therapeutic approaches or combination of new approaches with current strategies to promote functional recovery and quality of life in TPNI patients. Table 1 shows currently used and the potentially novel therapeutic strategies for TPNI management. Future treatments for TPNI will likely require the development of new pharmacologic adjuvant agents as well as uncovering the mechanistic details of those currently available. Key to the successful translation of treatments into widespread use in humans is well-executed clinical trials, which we and others have begun on several adjuvant forms of TPNI treatments. Critical gains in promoting myelin formation or myeloprotection, and allowing early diagnosis will advance treatment substantially.
Acknowledgments:
This work was supported by grants from the NIH (K08 AR060164–01A) and DOD (W81XWH-16-1-0725) in addition to institutional support from the University of Rochester and Pennsylvania State University Medical Centers.
Abbreviations
- TPNI
traumatic peripheral nerve injury
- EDX
electrodiagnostic testing
- NCS
nerve conduction study
- EMG
electromyography
- MUAP
motor unit action potential
- CMAP
compound muscle action potential
- CNS
central nervous system
- PNS
peripheral nervous system
- WHI
Women’s Health Initiative
- DVT
deep vein thrombosis
- TCF/LEF
T-cell factor/lymphoid-enhancer
- MPZ
myelin protein zero (MPZ)
- PMP22
peripheral myelin protein 22
- EPO
erythropoietin (EPO)
- EPOR
erythropoietin receptor
- 4-AP
4-aminopyridine
- ES
electrical stimulation
- cAMP
cyclic adenosine monophosphate
- BDNF
brain-derived neurotrophic factor
- ESCs
embryonic stem cells
- hPSCs
human pluripotent stem cells
- ADSCs
adipose-derived stem cells
- SKPCs
skin-derived precursor cells
- LLLT
low-level laser therapy
- PBM
Photobiomodulation
- Sox 10
SRY-related HMG-box-10
- Oct6
octamer-binding transcription factor-6
- Krox20
early growth response gene (Egr2)
- NAB
NGFI-A/Egr-binding protein
- HDAC
histone deacetylase
Footnotes
Ethical publication statement: we confirm that we have read the Journal’s position on issues involved in ethical publication and affirm that this report is consistent with those guidelines.
Conflict of interest: The authors have no conflict of interest to disclose.
References
- 1).Robinson LR. Traumatic injury to peripheral nerves. Muscle Nerve 2000;23(6):863–873. [DOI] [PubMed] [Google Scholar]
- 2).Taylor CA, Braza D, Rice JB, Dillingham T. The incidence of peripheral nerve injury in extremity trauma. Am J Phys Med Rehabil 2008;87(5):381–385. [DOI] [PubMed] [Google Scholar]
- 3).Evans GR. Peripheral nerve injury: a review and approach to tissue engineered constructs. Anat Rec 2001;263(4):396–404. [DOI] [PubMed] [Google Scholar]
- 4).Campbell WW. Evaluation and management of peripheral nerve injury. Clin Neurophysiol 2008;119(9):1951–1965. [DOI] [PubMed] [Google Scholar]
- 5).Robinson LR. traumatic injury to peripheral nerves. Suppl Clin Neurophysiol 2004;57:173–186. [DOI] [PubMed] [Google Scholar]
- 6).Noble J, Munro CA, Prasad VS, Midha R. Analysis of upper and lower extremity peripheral nerve injuries in a population of patients with multiple injuries. J Trauma 1998;45(1):116–122. [DOI] [PubMed] [Google Scholar]
- 7).Sullivan R, Dailey T, Duncan K, Abel N, Borlongan CV. Peripheral Nerve Injury: Stem Cell Therapy and Peripheral Nerve Transfer. Int J Mol Sci 2016;17(12). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8).Birch R, Misra P, Stewart MP, Eardley WG, Ramasamy A, Brown K, Shenoy R, Anand P, Clasper J, Dunn R, Etherington J. Nerve injuries sustained during warfare: part I--Epidemiology. J Bone Joint Surg Br 2012;94(4):523–528. [DOI] [PubMed] [Google Scholar]
- 9).Missios S, Bekelis K, Spinner RJ. Traumatic peripheral nerve injuries in children: epidemiology and socioeconomics. J Neurosurg Pediatr 2014;14(6):688–694. [DOI] [PubMed] [Google Scholar]
- 10).Menorca RM, Fussell TS, Elfar JC. Nerve physiology: mechanisms of injury and recovery. Hand Clin 2013;29(3):317–330. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11).SUNDERLAND S A classification of peripheral nerve injuries producing loss of function. Brain 1951;74(4):491–516. [DOI] [PubMed] [Google Scholar]
- 12).Sunderland S The anatomy and physiology of nerve injury. Muscle Nerve 1990;13(9):771–784. [DOI] [PubMed] [Google Scholar]
- 13).Kaya Y, Sarikcioglu L. Sir Herbert Seddon (1903–1977) and his classification scheme for peripheral nerve injury. Childs Nerv Syst 2015;31(2):177–180. [DOI] [PubMed] [Google Scholar]
- 14).Miller RG. AAEE minimonograph #28: injury to peripheral motor nerves. Muscle Nerve 1987;10(8):698–710. [DOI] [PubMed] [Google Scholar]
- 15).Post R, de Boer KS, Malessy MJ. Outcome following nerve repair of high isolated clean sharp injuries of the ulnar nerve. PLoS One 2012;7(10):e47928. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16).Sulaiman W, Gordon T. Neurobiology of peripheral nerve injury, regeneration, and functional recovery: from bench top research to bedside application. Ochsner J 2013;13(1):100–108. [PMC free article] [PubMed] [Google Scholar]
- 17).Parrinello S, Napoli I, Ribeiro S, Wingfield Digby P, Fedorova M, Parkinson DB, Doddrell RD, Nakayama M, Adams RH, Lloyd AC. EphB signaling directs peripheral nerve regeneration through Sox2-dependent Schwann cell sorting. Cell 2010;143(1):145–155. doi: 10.1016/j.cell.2010.08.039. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18).Cattin AL, Burden JJ, Van Emmenis L, Mackenzie FE, Hoving JJ, Garcia Calavia N, Guo Y, McLaughlin M, Rosenberg LH, Quereda V, Jamecna D1, Napoli I, Parrinello S, Enver T, Ruhrberg C, Lloyd AC. Macrophage-Induced Blood Vessels Guide Schwann Cell-Mediated Regeneration of Peripheral Nerves. Cell 2015;162(5):1127–1139. doi: 10.1016/j.cell.2015.07.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19).Fu SY, Gordon T. Contributing factors to poor functional recovery after delayed nerve repair: prolonged axotomy. J Neurosci 1995;15(5 Pt 2):3876–3885. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20).Preston DC, Shapiro BE. 2 - Anatomy and Neurophysiology. Electromyography and Neuromuscular Disorders (Third Edition). London: W.B. Saunders; 2013. p 8–18. [Google Scholar]
- 21).Isaacs J, Browne T. Overcoming short gaps in peripheral nerve repair: conduits and human acellular nerve allograft. Hand (N Y) 2014;9(2):131–137. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22).Millesi H Bridging defects: autologous nerve grafts. Acta Neurochir Suppl 2007;100:37–38. [DOI] [PubMed] [Google Scholar]
- 23).Muheremu A, Ao Q. Past, Present, and Future of Nerve Conduits in the Treatment of Peripheral Nerve Injury. Biomed Res Int 2015;2015:237507. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24).Siemionow M, Brzezicki G. Chapter 8: Current techniques and concepts in peripheral nerve repair. Int Rev Neurobiol 2009;87:141–172. [DOI] [PubMed] [Google Scholar]
- 25).Blits B, Boer GJ, Verhaagen J. Pharmacological, cell, and gene therapy strategies to promote spinal cord regeneration. Cell Transplant 2002;11(6):593–613. [PubMed] [Google Scholar]
- 26).Faroni A, Mobasseri SA, Kingham PJ, Reid AJ. Peripheral nerve regeneration: experimental strategies and future perspectives. Adv Drug Deliv Rev 2015;82–83:160–167. [DOI] [PubMed] [Google Scholar]
- 27).Magnaghi V, Procacci P, Tata AM. Chapter 15: Novel pharmacological approaches to Schwann cells as neuroprotective agents for peripheral nerve regeneration. Int Rev Neurobiol 2009;87:295–315. [DOI] [PubMed] [Google Scholar]
- 28).Dvali L, Mackinnon S. (2007). The role of microsurgery in nerve repair and nerve grafting. Hand Clin 2007;23(1), 73–81. doi: 10.1016/j.hcl.2007.02.003 [DOI] [PubMed] [Google Scholar]
- 29).Mackinnon SE. Future Perspectives in the Management of Nerve Injuries. J Reconstr Microsurg 2018;34(9):672–674. doi: 10.1055/s-0038-1639353. [DOI] [PubMed] [Google Scholar]
- 30).Nobakhti-Afshar A, Najafpour A, Mohammadi R, Zarei L. Assessment of Neuroprotective Effects of Local Administration of 17- Beta- Estradiol on Peripheral Nerve Regeneration in Ovariectomized Female Rats. Bull Emerg Trauma 2016;4(3):141–149. [PMC free article] [PubMed] [Google Scholar]
- 31).Koenig HL, Gong WH, Pelissier P. Role of progesterone in peripheral nerve repair. Rev Reprod 2000;5(3):189–199. [DOI] [PubMed] [Google Scholar]
- 32).Chen Y, Guo W, Xu L, Li W, Cheng M, Hu Y, Xu W. 17β-Estradiol Promotes Schwann Cell Proliferation and Differentiation, Accelerating Early Remyelination in a Mouse Peripheral Nerve Injury Model. Biomed Res Int 2016;2016:7891202. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33).Zhu TS, Glaser M. Neuroprotection and enhancement of remyelination by estradiol and dexamethasone in cocultures of rat DRG neurons and Schwann cells. Brain Res 2008;1206:20–32. [DOI] [PubMed] [Google Scholar]
- 34).Schumacher M, Guennoun R, Mercier G, Désarnaud F, Lacor P, Bénavides J, Ferzaz B, Robert F, Baulieu EE. Progesterone synthesis and myelin formation in peripheral nerves. Brain Res Brain Res Rev 2001;37(1–3):343–359. [DOI] [PubMed] [Google Scholar]
- 35).Abe N, Borson SH, Gambello MJ, Wang F, Cavalli V. Mammalian target of rapamycin (mTOR) activation increases axonal growth capacity of injured peripheral nerves. J Biol Chem 2010;285(36):28034–43. doi: 10.1074/jbc.M110.125336. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36).Park KK, Liu K, Hu Y, Kanter JL, He Z. PTEN/mTOR and axon regeneration. Exp Neurol 2010;223(1):45–50. doi: 10.1016/j.expneurol.2009.12.032. [DOI] [PubMed] [Google Scholar]
- 37).Aigner L, Arber S, Kapfhammer JP, Laux T, Schneider C, Botteri F, Brenner HR, Caroni P. Overexpression of the neural growth-associated protein GAP-43 induces nerve sprouting in the adult nervous system of transgenic mice. Cell 1995;83(2):269–78. [DOI] [PubMed] [Google Scholar]
- 38).Magnaghi V, Cavarretta I, Galbiati M, Martini L, Melcangi RC. Neuroactive steroids and peripheral myelin proteins. Brain Res Brain Res Rev 2001;37(1–3):360–371. [DOI] [PubMed] [Google Scholar]
- 39).Mirsky R, Woodhoo A, Parkinson DB, Arthur-Farraj P, Bhaskaran A, Jessen KR. Novel signals controlling embryonic Schwann cell development, myelination and dedifferentiation. J Peripher Nerv Syst 2008;13(2):122–35. doi: 10.1111/j.1529-8027.2008.00168.x. [DOI] [PubMed] [Google Scholar]
- 40).Miller VM, Harman SM. An update on hormone therapy in postmenopausal women: mini-review for the basic scientist. Am J Physiol Heart Circ Physiol 2017;313(5):H1013–H1021. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41).Bonkhoff H Estrogen receptor signaling in prostate cancer: Implications for carcinogenesis and tumor progression. Prostate 2018;78(1):2–10. [DOI] [PubMed] [Google Scholar]
- 42).Bracken MB. Steroids for acute spinal cord injury. Cochrane Database Syst Rev 2012;1:CD001046. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43).Eker HE, Cok OY, Aribogan A, Arslan G. Management of neuropathic pain with methylprednisolone at the site of nerve injury. Pain Med 2012;13(3):443–451. [DOI] [PubMed] [Google Scholar]
- 44).Gronseth GS, Paduga R, Neurology AAo. Evidence-based guideline update: steroids and antivirals for Bell palsy: report of the Guideline Development Subcommittee of the American Academy of Neurology. Neurology 2012;79(22):2209–2213. [DOI] [PubMed] [Google Scholar]
- 45).Stark H, Amirfeyz R. Cochrane corner: local corticosteroid injection for carpal tunnel syndrome. J Hand Surg Eur Vol 2013;38(8):911–914. [DOI] [PubMed] [Google Scholar]
- 46).Bernstein DT, Weiner BK, Tasciotti E, Mathis KB. Does the combination of erythropoietin and tapered oral corticosteroids improve recovery following iatrogenic nerve injury? Injury 2016;47(8):1819–1823. [DOI] [PubMed] [Google Scholar]
- 47).Curtis JR, Westfall AO, Allison J, Bijlsma JW, Freeman A, George V, Kovac SH, Spettell CM, Saag KG. Population-based assessment of adverse events associated with long-term glucocorticoid use. Arthritis Rheum 2006;55(3):420–426. [DOI] [PubMed] [Google Scholar]
- 48).Huscher D, Thiele K, Gromnica-Ihle E, Hein G, Demary W, Dreher R, Zink A, Buttgereit F. Dose-related patterns of glucocorticoid-induced side effects. Ann Rheum Dis 2009;68(7):1119–1124. [DOI] [PubMed] [Google Scholar]
- 49).McDougall R, Sibley J, Haga M, Russell A. Outcome in patients with rheumatoid arthritis receiving prednisone compared to matched controls. J Rheumatol 1994;21(7):1207–1213. [PubMed] [Google Scholar]
- 50).Saag KG, Koehnke R, Caldwell JR, Brasington R, Burmeister LF, Zimmerman B, et al. Low dose long-term corticosteroid therapy in rheumatoid arthritis: an analysis of serious adverse events. Am J Med 1994;96(2):115–23. [DOI] [PubMed] [Google Scholar]
- 51).Galloway EB, Jensen RL, Dailey AT, Thompson BG, Shelton C. Role of topical steroids in reducing dysfunction after nerve injury. Laryngoscope 2000;110(11):1907–1910. [DOI] [PubMed] [Google Scholar]
- 52).Mohammadi R, Azad-Tirgan M, Amini K. Dexamethasone topically accelerates peripheral nerve repair and target organ reinnervation: a transected sciatic nerve model in rat. Injury 2013;44(4):565–569. [DOI] [PubMed] [Google Scholar]
- 53).Suslu H, Altun M, Erdivanli B, Turan Suslu H. Comparison of the effects of local and systemic dexamethasone on the rat traumatic sciatic nerve model. Turk Neurosurg 2013;23(5):623–629. [DOI] [PubMed] [Google Scholar]
- 54).Dame C, Juul SE, Christensen RD. The biology of erythropoietin in the central nervous system and its neurotrophic and neuroprotective potential. Biol Neonate 2001;79(3–4):228–235. [DOI] [PubMed] [Google Scholar]
- 55).Morishita E, Masuda S, Nagao M, Yasuda Y, Sasaki R. Erythropoietin receptor is expressed in rat hippocampal and cerebral cortical neurons, and erythropoietin prevents in vitro glutamate-induced neuronal death. Neuroscience 1997;76(1):105–116. [DOI] [PubMed] [Google Scholar]
- 56).Sakanaka M, Wen TC, Matsuda S, Masuda S, Morishita E, Nagao M, Sasaki R. In vivo evidence that erythropoietin protects neurons from ischemic damage. Proc Natl Acad Sci U S A 1998;95(8):4635–4640. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57).Sundem L, Chris Tseng KC, Li H, Ketz J, Noble M, Elfar J. Erythropoietin Enhanced Recovery After Traumatic Nerve Injury: Myelination and Localized Effects. J Hand Surg Am 2016;41(10):999–1010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58).Maiese K Regeneration in the nervous system with erythropoietin. Front Biosci (Landmark Ed) 2016;21:561–596. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59).Maiese K, Chong ZZ, Shang YC, Wang S. Erythropoietin: new directions for the nervous system. Int J Mol Sci 2012;13(9):11102–11129. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60).Sargin D, Friedrichs H, El-Kordi A, Ehrenreich H. Erythropoietin as neuroprotective and neuroregenerative treatment strategy: comprehensive overview of 12 years of preclinical and clinical research. Best Pract Res Clin Anaesthesiol 2010;24(4):573–594. [DOI] [PubMed] [Google Scholar]
- 61).Lombardero M, Kovacs K, Scheithauer BW. Erythropoietin: a hormone with multiple functions. Pathobiology 2011;78(1):41–53. [DOI] [PubMed] [Google Scholar]
- 62).Kondyli M, Gatzounis G, Kyritsis A, Varakis J, Assimakopoulou M. Immunohistochemical detection of phosphorylated JAK-2 and STAT-5 proteins and correlation with erythropoietin receptor (EpoR) expression status in human brain tumors. J Neurooncol 2010;100(2):157–164. [DOI] [PubMed] [Google Scholar]
- 63).Maiese K, Hou J, Chong ZZ, Shang YC. Erythropoietin, forkhead proteins, and oxidative injury: biomarkers and biology. ScientificWorldJournal 2009;9:1072–1104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64).Hassan K, Gross B, Simri W, Rubinchik I, Cohen H, Jacobi J, Shasha SM, Kristal B. The presence of erythropoietin receptors in the human peripheral nervous system. Clin Nephrol 2004;61(2):127–129. [DOI] [PubMed] [Google Scholar]
- 65).Elfar JC, Jacobson JA, Puzas JE, Rosier RN, Zuscik MJ. Erythropoietin accelerates functional recovery after peripheral nerve injury. J Bone Joint Surg Am 2008;90(8):1644–1653. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66).Toth C, Martinez JA, Liu WQ, Diggle J, Guo GF, Ramji N, Mi R, Hoke A, Zochodne DW. Local erythropoietin signaling enhances regeneration in peripheral axons. Neuroscience 2008;154(2):767–783. [DOI] [PubMed] [Google Scholar]
- 67).Geary MB, Li H, Zingman A, Ketz J, Zuscik M, De Mesy Bentley KL, Noble M, Elfar JC. Erythropoietin accelerates functional recovery after moderate sciatic nerve crush injury. Muscle Nerve 2017;56(1):143–151. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68).Grasso G, Meli F, Fodale V, Calapai G, Buemi M, Iacopino DG. Neuroprotective potential of erythropoietin and darbepoetin alfa in an experimental model of sciatic nerve injury. Laboratory investigation. J Neurosurg Spine 2007;7(6):645–651. [DOI] [PubMed] [Google Scholar]
- 69).Cervellini I, Annenkov A, Brenton T, Chernajovsky Y, Ghezzi P, Mengozzi M. Erythropoietin (EPO) increases myelin gene expression in CG4 oligodendrocyte cells through the classical EPO receptor. Mol Med 2013;19:223–229. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70).Itri LM. The use of epoetin alfa in chemotherapy patients: a consistent profile of efficacy and safety. Semin Oncol 2002;29(3 Suppl 8):81–87. [DOI] [PubMed] [Google Scholar]
- 71).Straus DJ. Epoetin alfa as a supportive measure in hematologic malignancies. Semin Hematol 2002;39(4 Suppl 3):25–31. [DOI] [PubMed] [Google Scholar]
- 72).Costa DD, Beghi E, Carignano P, Pagliacci C, Faccioli F, Pupillo E, Messina P, Gorio A, Redaelli T. Tolerability and efficacy of erythropoietin (EPO) treatment in traumatic spinal cord injury: a preliminary randomized comparative trial vs. methylprednisolone (MP). Neurol Sci 2015;36(9):1567–1574. [DOI] [PubMed] [Google Scholar]
- 73).Abraham PA, Macres MG. Blood pressure in hemodialysis patients during amelioration of anemia with erythropoietin. J Am Soc Nephrol 1991;2(4):927–936. [DOI] [PubMed] [Google Scholar]
- 74).Raine AE, Roger SD. Effects of erythropoietin on blood pressure. Am J Kidney Dis 1991;18(4 Suppl 1):76–83. [PubMed] [Google Scholar]
- 75).Bennett WM. Side effects of erythropoietin therapy. Am J Kidney Dis 1991;18(4 Suppl 1):84–86. [PubMed] [Google Scholar]
- 76).Eschbach JW, Abdulhadi MH, Browne JK, Delano BG, Downing MR, Egrie JC, Evans RW, Friedman EA, Graber SE, Haley NR. Recombinant human erythropoietin in anemic patients with end-stage renal disease. Results of a phase III multicenter clinical trial. Ann Intern Med 1989;111(12):992–1000. [DOI] [PubMed] [Google Scholar]
- 77).Egeberg MD, Oh CY, Bainbridge JL. Clinical overview of dalfampridine: an agent with a novel mechanism of action to help with gait disturbances. Clin Ther 2012;34(11):2185–2194. [DOI] [PubMed] [Google Scholar]
- 78).Jensen HB, Ravnborg M, Dalgas U, Stenager E. 4-Aminopyridine for symptomatic treatment of multiple sclerosis: a systematic review. Ther Adv Neurol Disord 2014;7(2):97–113. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79).Lundh H, Nilsson O, Rosén I. Effects of 4-aminopyridine in myasthenia gravis. J Neurol Neurosurg Psychiatry 1979;42(2):171–175. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80).Hansebout RR, Blight AR, Fawcett S, Reddy K. 4-Aminopyridine in chronic spinal cord injury: a controlled, double-blind, crossover study in eight patients. J Neurotrauma 1993;10(1):1–18. [DOI] [PubMed] [Google Scholar]
- 81).Sanders DB, Massey JM, Sanders LL, Edwards LJ. A randomized trial of 3,4-diaminopyridine in Lambert-Eaton myasthenic syndrome. Neurology 2000;54(3):603–607. [DOI] [PubMed] [Google Scholar]
- 82).Wirtz PW, Verschuuren JJ, van Dijk JG, de Kam ML, Schoemaker RC, van Hasselt JG, Titulaer MJ, Tjaden UR, den Hartigh J, van Gerven JM. Efficacy of 3,4-diaminopyridine and pyridostigmine in the treatment of Lambert-Eaton myasthenic syndrome: a randomized, double-blind, placebo-controlled, crossover study. Clin Pharmacol Ther 2009;86(1):44–48. [DOI] [PubMed] [Google Scholar]
- 83).Judge SI, Bever CT. Potassium channel blockers in multiple sclerosis: neuronal Kv channels and effects of symptomatic treatment. Pharmacol Ther 2006;111(1):224–259. [DOI] [PubMed] [Google Scholar]
- 84).Smith KJ, Felts PA, John GR. Effects of 4-aminopyridine on demyelinated axons, synapses and muscle tension. Brain 2000;123 (Pt 1):171–184. [DOI] [PubMed] [Google Scholar]
- 85).van Lunteren E, Moyer M. Combination of variable frequency train stimulation and K+ channel blockade to augment skeletal muscle force. IEEE Trans Neural Syst Rehabil Eng 2004;12(2):288–294. [DOI] [PubMed] [Google Scholar]
- 86).Tseng KC, Li H, Clark A, Sundem L, Zuscik M, Noble M, Elfar J. 4-Aminopyridine promotes functional recovery and remyelination in acute peripheral nerve injury. EMBO Mol Med 2016;8(12):1409–1420. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87).Clark AR, Hsu CG, Talukder MAH, Noble M, Elfar JC. Transdermal delivery of 4-aminopyridine accelerates motor functional recovery and improves nerve morphology following sciatic nerve crush injury in mice. Neural Regen Res 2019; 14(0):000–000. doi:10.4103/ [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88).Noble M, Tseng KC, Li H, Elfar JC. 4-Aminopyridine as a single agent diagnostic and treatment for severe nerve crush injury. Mil Med 2019;184(Suppl 1):379–385. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89).Bowe CM, Kocsis JD, Targ EF, Waxman SG. Physiological effects of 4-aminopyridine on demyelinated mammalian motor and sensory fibers. Ann Neurol 1987;22(2):264–268. [DOI] [PubMed] [Google Scholar]
- 90).Sherratt RM, Bostock H, Sears TA. Effects of 4-aminopyridine on normal and demyelinated mammalian nerve fibres. Nature 1980;283(5747):570–572. [DOI] [PubMed] [Google Scholar]
- 91).Lee DH, Claussen GC, Oh S. Clinical nerve conduction and needle electromyography studies. J Am Acad Orthop Surg 2004;12(4):276–287. [DOI] [PubMed] [Google Scholar]
- 92).Robinson LR. Role of neurophysiologic evaluation in diagnosis. J Am Acad Orthop Surg 2000;8(3):190–199. [DOI] [PubMed] [Google Scholar]
- 93).Shah A, Jebson PJ. Current treatment of radial nerve palsy following fracture of the humeral shaft. J Hand Surg Am 2008;33(8):1433–1434. [DOI] [PubMed] [Google Scholar]
- 94).NMMMGA Elfar J. A Clinical Trial of 4AP for the Treatment of Post-Prostatectomy Peripheral Nerve Injury in Humans (IND 128117 approved 2016). 2016.
- 95).Goodman AD, Stone RT. Enhancing neural transmission in multiple sclerosis (4-aminopyridine therapy). Neurotherapeutics 2013;10(1):106–110. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96).Blight AR, Toombs JP, Bauer MS, Widmer WR. The effects of 4-aminopyridine on neurological deficits in chronic cases of traumatic spinal cord injury in dogs: a phase I clinical trial. J Neurotrauma 1991;8(2):103–119. [DOI] [PubMed] [Google Scholar]
- 97).Goodman AD, Cohen JA, Cross A, Vollmer T, Rizzo M, Cohen R, Marinucci L, Blight AR. Fampridine-SR in multiple sclerosis: a randomized, double-blind, placebo-controlled, dose-ranging study. Mult Scler 2007;13(3):357–368. [DOI] [PubMed] [Google Scholar]
- 98).Bever CT, Young D, Anderson PA, Krumholz A, Conway K, Leslie J, Eddington N, Plaisance KI, Panitch HS, Dhib-Jalbut S. The effects of 4-aminopyridine in multiple sclerosis patients: results of a randomized, placebo-controlled, double-blind, concentration-controlled, crossover trial. Neurology 1994;44(6):1054–1059. [DOI] [PubMed] [Google Scholar]
- 99).Van Diemen HA, Polman CH, Koetsier JC, Van Loenen AC, Nauta JJ, Bertelsmann FW. 4-Aminopyridine in patients with multiple sclerosis: dosage and serum level related to efficacy and safety. Clin Neuropharmacol 1993;16(3):195–204. [DOI] [PubMed] [Google Scholar]
- 100).Al-Majed AA, Neumann CM, Brushart TM, Gordon T. Brief electrical stimulation promotes the speed and accuracy of motor axonal regeneration. J Neurosci 2000;20(7):2602–2608. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101).Haastert-Talini K, Schmitte R, Korte N, Klode D, Ratzka A, Grothe C. Electrical stimulation accelerates axonal and functional peripheral nerve regeneration across long gaps. J Neurotrauma 2011;28(4):661–674. [DOI] [PubMed] [Google Scholar]
- 102).Nix WA, Hopf HC. Electrical stimulation of regenerating nerve and its effect on motor recovery. Brain Res 1983;272(1):21–25. [DOI] [PubMed] [Google Scholar]
- 103).Pockett S, Gavin RM. Acceleration of peripheral nerve regeneration after crush injury in rat. Neurosci Lett 1985;59(2):221–224. [DOI] [PubMed] [Google Scholar]
- 104).MacEwan MR, Gamble P, Stephen M, Ray WZ. Therapeutic electrical stimulation of injured peripheral nerve tissue using implantable thin-film wireless nerve stimulators. J Neurosurg 2018:1–10. [DOI] [PubMed] [Google Scholar]
- 105).Mendez A, Hopkins A, Biron VL, Seikaly H, Zhu LF, Côté DWJ. Brief electrical stimulation and synkinesis after facial nerve crush injury: a randomized prospective animal study. J Otolaryngol Head Neck Surg 2018;47(1):20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106).Brushart TM, Jari R, Verge V, Rohde C, Gordon T. Electrical stimulation restores the specificity of sensory axon regeneration. Exp Neurol 2005;194(1):221–229. [DOI] [PubMed] [Google Scholar]
- 107).Gordon T Electrical Stimulation to Enhance Axon Regeneration After Peripheral Nerve Injuries in Animal Models and Humans. Neurotherapeutics 2016;13(2):295–310. doi: 10.1007/s13311-015-0415-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108).Huang J, Lu L, Zhang J, Hu X, Zhang Y, Liang W, Wu S, Luo Z. Electrical stimulation to conductive scaffold promotes axonal regeneration and remyelination in a rat model of large nerve defect. PLoS One 2012;7(6):e39526. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109).Dow DE, Dennis RG, Faulkner JA. Electrical stimulation attenuates denervation and age-related atrophy in extensor digitorum longus muscles of old rats. J Gerontol A Biol Sci Med Sci 2005;60(4):416–424. [DOI] [PubMed] [Google Scholar]
- 110).Eberstein A, Eberstein S. Electrical stimulation of denervated muscle: is it worthwhile? Med Sci Sports Exerc 1996;28(12):1463–1469. [DOI] [PubMed] [Google Scholar]
- 111).KOSMAN AJ, WOOD EC, OSBORNE SL. Effect of electrical stimulation upon atrophy of partially denervated skeletal muscle of the rat. Am J Physiol 1948;154(3):451–454. [DOI] [PubMed] [Google Scholar]
- 112).Armada-da-Silva PA, Pereira C, Amado S, Veloso AP. Role of physical exercise for improving posttraumatic nerve regeneration. Int Rev Neurobiol 2013;109:125–149. doi: 10.1016/B978-0-12-420045-6.00006-7. [DOI] [PubMed] [Google Scholar]
- 113).Marqueste T, Alliez JR, Alluin O, Jammes Y, Decherchi P. Neuromuscular rehabilitation by treadmill running or electrical stimulation after peripheral nerve injury and repair. J Appl Physiol (1985). 2004;96(5):1988–1995. [DOI] [PubMed] [Google Scholar]
- 114).Asensio-Pinilla E, Udina E, Jaramillo J, Navarro X. Electrical stimulation combined with exercise increase axonal regeneration after peripheral nerve injury. Exp Neurol 2009;219(1):258–265. doi: 10.1016/j.expneurol.2009.05.034. [DOI] [PubMed] [Google Scholar]
- 115).Gordon T, English AW. Strategies to promote peripheral nerve regeneration: electrical stimulation and/or exercise. Eur J Neurosci 2016;43(3):336–350. doi: 10.1111/ejn.13005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116).Gordon T, Sulaiman OA, Ladak A. Chapter 24: Electrical stimulation for improving nerve regeneration: where do we stand? Int Rev Neurobiol 2009;87:433–444. doi: 10.1016/S0074-7742(09)87024-4. [DOI] [PubMed] [Google Scholar]
- 117).Senger JLB, Verge VMK, Macandili HSJ, Olson JL, Chan KM, Webber CA. Electrical stimulation as a conditioning strategy for promoting and accelerating peripheral nerve regeneration. Exp Neurol 2018;302:75–84. doi: 10.1016/j.expneurol.2017.12.013. [DOI] [PubMed] [Google Scholar]
- 118).Senger JL, Chan KM, Macandili H, Chan AWM, Verge VMK, Jones KE, Webber CA. Conditioning electrical stimulation promotes functional nerve regeneration. Exp Neurol 2019;315:60–71. doi: 10.1016/j.expneurol.2019.02.001. [DOI] [PubMed] [Google Scholar]
- 119).Al-Majed AA, Brushart TM, Gordon T. Electrical stimulation accelerates and increases expression of BDNF and trkB mRNA in regenerating rat femoral motoneurons. Eur J Neurosci 2000;12(12):4381–4390. [PubMed] [Google Scholar]
- 120).Gordon T The role of neurotrophic factors in nerve regeneration. Neurosurg Focus 2009;26(2):E3. doi: 10.3171/FOC.2009.26.2.E3. [DOI] [PubMed] [Google Scholar]
- 121).Sabatier MJ, Redmon N, Schwartz G, English AW. Treadmill training promotes axon regeneration in injured peripheral nerves. Exp Neurol 2008;211(2):489–493. doi: 10.1016/j.expneurol.2008.02.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122).Wenjin W, Wenchao L, Hao Z, Feng L, Yan W, Wodong S, Xianqun F, Wenlong D. Electrical stimulation promotes BDNF expression in spinal cord neurons through Ca(2+)- and Erk-dependent signaling pathways. Cell Mol Neurobiol 2011;31(3):459–467. doi: 10.1007/s10571-010-9639-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123).Wan LD, Xia R, Ding WL. Electrical stimulation enhanced remyelination of injured sciatic nerves by increasing neurotrophins. Neuroscience 2010;169(3):1029–1038. [DOI] [PubMed] [Google Scholar]
- 124).Wan L, Xia R, Ding W. Short-term low-frequency electrical stimulation enhanced remyelination of injured peripheral nerves by inducing the promyelination effect of brain-derived neurotrophic factor on Schwann cell polarization. J Neurosci Res 2010;88(12):2578–2587. [DOI] [PubMed] [Google Scholar]
- 125).Williams HB. A clinical pilot study to assess functional return following continuous muscle stimulation after nerve injury and repair in the upper extremity using a completely implantable electrical system. Microsurgery 1996;17(11):597–605. [DOI] [PubMed] [Google Scholar]
- 126).You S, Petrov T, Chung PH, Gordon T. The expression of the low affinity nerve growth factor receptor in long-term denervated Schwann cells. Glia 1997; 20(2), 87–100. [DOI] [PubMed] [Google Scholar]
- 127).Li H, Terenghi G, Hall SM. Effects of delayed re-innervation on the expression of c-erbB receptors by chronically denervated rat Schwann cells in vivo. Glia 1997; 20(4), 333–347. [DOI] [PubMed] [Google Scholar]
- 128).Borlongan CV. Remyelinating the transected peripheral nerve by fabricated Schwann cells derived from bone marrow. Exp Neurol 2010;225(2):243–245. [DOI] [PubMed] [Google Scholar]
- 129).Guénard V, Kleitman N, Morrissey TK, Bunge RP, Aebischer P. Syngeneic Schwann cells derived from adult nerves seeded in semipermeable guidance channels enhance peripheral nerve regeneration. J Neurosci 1992;12(9):3310–3320. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130).Guest JD, Rao A, Olson L, Bunge MB, Bunge RP. The ability of human Schwann cell grafts to promote regeneration in the transected nude rat spinal cord. Exp Neurol 1997;148(2):502–522. [DOI] [PubMed] [Google Scholar]
- 131).Walsh S, Midha R. Use of stem cells to augment nerve injury repair. Neurosurgery 2009;65(4 Suppl):A80–86. [DOI] [PubMed] [Google Scholar]
- 132).Jiang L, Jones S, Jia X. Stem Cell Transplantation for Peripheral Nerve Regeneration: Current Options and Opportunities. Int J Mol Sci 2017;18(1). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133).Ziegler L, Grigoryan S, Yang IH, Thakor NV, Goldstein RS. Efficient generation of schwann cells from human embryonic stem cell-derived neurospheres. Stem Cell Rev 2011;7(2):394–403. [DOI] [PubMed] [Google Scholar]
- 134).Cui L, Jiang J, Wei L, Zhou X, Fraser JL, Snider BJ, Yu SP. Transplantation of embryonic stem cells improves nerve repair and functional recovery after severe sciatic nerve axotomy in rats. Stem Cells 2008;26(5):1356–1365. [DOI] [PubMed] [Google Scholar]
- 135).Rippon HJ, Bishop AE. Embryonic stem cells. Cell Prolif 2004;37(1):23–34. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136).Ren Z, Wang Y, Peng J, Zhao Q, Lu S. Role of stem cells in the regeneration and repair of peripheral nerves. Rev Neurosci 2012;23(2):135–143. [DOI] [PubMed] [Google Scholar]
- 137).Chen CJ, Ou YC, Liao SL, Chen WY, Chen SY, Wu CW, Wang CC, Wang WY, Huang YS, Hsu SH. Transplantation of bone marrow stromal cells for peripheral nerve repair. Exp Neurol 2007;204(1):443–453. [DOI] [PubMed] [Google Scholar]
- 138).Dezawa M, Takahashi I, Esaki M, Takano M, Sawada H. Sciatic nerve regeneration in rats induced by transplantation of in vitro differentiated bone-marrow stromal cells. Eur J Neurosci 2001;14(11):1771–1776. [DOI] [PubMed] [Google Scholar]
- 139).Keilhoff G, Stang F, Goihl A, Wolf G, Fansa H. Transdifferentiated mesenchymal stem cells as alternative therapy in supporting nerve regeneration and myelination. Cell Mol Neurobiol 2006;26(7–8):1235–1252. [DOI] [PubMed] [Google Scholar]
- 140).Kingham PJ, Kalbermatten DF, Mahay D, Armstrong SJ, Wiberg M, Terenghi G. Adipose-derived stem cells differentiate into a Schwann cell phenotype and promote neurite outgrowth in vitro. Exp Neurol 2007;207(2):267–274. [DOI] [PubMed] [Google Scholar]
- 141).Sowa Y, Imura T, Numajiri T, Nishino K, Fushiki S. Adipose-derived stem cells produce factors enhancing peripheral nerve regeneration: influence of age and anatomic site of origin. Stem Cells Dev 2012;21(11):1852–1862. [DOI] [PubMed] [Google Scholar]
- 142).Sowa Y, Kishida T, Imura T, Numajiri T, Nishino K, Tabata Y, Mazda O. Adipose-Derived Stem Cells Promote Peripheral Nerve Regeneration In Vivo without Differentiation into Schwann-Like Lineage. Plast Reconstr Surg 2016;137(2):318e–330e. [DOI] [PubMed] [Google Scholar]
- 143).Faroni A, Smith RJ, Lu L, Reid AJ. Human Schwann-like cells derived from adipose-derived mesenchymal stem cells rapidly de-differentiate in the absence of stimulating medium. Eur J Neurosci 2016;43(3):417–430. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144).McKenzie IA, Biernaskie J, Toma JG, Midha R, Miller FD. Skin-derived precursors generate myelinating Schwann cells for the injured and dysmyelinated nervous system. J Neurosci 2006;26(24):6651–6660. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145).Toma JG, McKenzie IA, Bagli D, Miller FD. Isolation and characterization of multipotent skin-derived precursors from human skin. Stem Cells 2005;23(6):727–737. [DOI] [PubMed] [Google Scholar]
- 146).Walsh SK, Gordon T, Addas BM, Kemp SW, Midha R. Skin-derived precursor cells enhance peripheral nerve regeneration following chronic denervation. Exp Neurol 2010;223(1):221–228. doi: 10.1016/j.expneurol.2009.05.025. [DOI] [PubMed] [Google Scholar]
- 147).de Oliveira RF, de Andrade Salgado DM, Trevelin LT, Lopes RM, da Cunha SR, Aranha AC, de Paula Eduardo C, de Freitas PM. Benefits of laser phototherapy on nerve repair. Lasers Med Sci 2015;30(4):1395–1406. [DOI] [PubMed] [Google Scholar]
- 148).Hashmi JT, Huang YY, Osmani BZ, Sharma SK, Naeser MA, Hamblin MR. Role of low-level laser therapy in neurorehabilitation. PM R 2010;2(12 Suppl 2):S292–305. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149).Schindl A, Schindl M, Pernerstorfer-Schön H, Schindl L. Low-intensity laser therapy: a review. J Investig Med 2000;48(5):312–326. [PubMed] [Google Scholar]
- 150).Karu TI. Mitochondrial signaling in mammalian cells activated by red and near-IR radiation. Photochem Photobiol 2008;84(5):1091–1099. doi: 10.1111/j.1751-1097.2008.00394.x. [DOI] [PubMed] [Google Scholar]
- 151).Passarella S, Karu T. Absorption of monochromatic and narrow band radiation in the visible and near IR by both mitochondrial and non-mitochondrial photoacceptors results in photobiomodulation. J Photochem Photobiol B 2014;140:344–58. doi: 10.1016/j.jphotobiol.2014.07.021. [DOI] [PubMed] [Google Scholar]
- 152).Anders JJ, Geuna S, Rochkind S. Phototherapy promotes regeneration and functional recovery of injured peripheral nerve. Neurol Res 2004;26(2):233–239. [DOI] [PubMed] [Google Scholar]
- 153).Barbosa RI, Marcolino AM, de Jesus Guirro RR, Mazzer N, Barbieri CH, de Cássia Registro Fonseca M. Comparative effects of wavelengths of low-power laser in regeneration of sciatic nerve in rats following crushing lesion. Lasers Med Sci 2010;25(3):423–430. [DOI] [PubMed] [Google Scholar]
- 154).Gigo-Benato D, Geuna S, de Castro Rodrigues A, Tos P, Fornaro M, Boux E, Battiston B, Giacobini-Robecchi MG. Low-power laser biostimulation enhances nerve repair after end-to-side neurorrhaphy: a double-blind randomized study in the rat median nerve model. Lasers Med Sci 2004;19(1):57–65. [DOI] [PubMed] [Google Scholar]
- 155).Mohammed IF, Al-Mustawfi N, Kaka LN. Promotion of regenerative processes in injured peripheral nerve induced by low-level laser therapy. Photomed Laser Surg 2007;25(2):107–111. [DOI] [PubMed] [Google Scholar]
- 156).Moges H, Wu X, McCoy J, Vasconcelos OM, Bryant H, Grunberg NE, Anders JJ. Effect of 810 nm light on nerve regeneration after autograft repair of severely injured rat median nerve. Lasers Surg Med 2011;43(9):901–906. [DOI] [PubMed] [Google Scholar]
- 157).Shen CC, Yang YC, Liu BS. Large-area irradiated low-level laser effect in a biodegradable nerve guide conduit on neural regeneration of peripheral nerve injury in rats. Injury 2011;42(8):803–13. doi: 10.1016/j.injury.2011.02.005. [DOI] [PubMed] [Google Scholar]
- 158).Yazdani SO, Golestaneh AF, Shafiee A, Hafizi M, Omrani HA, Soleimani M. Effects of low level laser therapy on proliferation and neurotrophic factor gene expression of human schwann cells in vitro. J Photochem Photobiol B 2012;107:9–13. [DOI] [PubMed] [Google Scholar]
- 159).Rochkind S, Leider-Trejo L, Nissan M, Shamir MH, Kharenko O, Alon M. Efficacy of 780-nm laser phototherapy on peripheral nerve regeneration after neurotube reconstruction procedure (double-blind randomized study). Photomed Laser Surg 2007;25(3):137–143. [DOI] [PubMed] [Google Scholar]
- 160).Rochkind S, Drory V, Alon M, Nissan M, Ouaknine GE. Laser phototherapy (780 nm), a new modality in treatment of long-term incomplete peripheral nerve injury: a randomized double-blind placebo-controlled study. Photomed Laser Surg 2007;25(5):436–442. [DOI] [PubMed] [Google Scholar]
- 161).Bagis S, Comelekoglu U, Coskun B, Milcan A, Buyukakilli B, Sahin G, Ozisik S, Erdogan C. No effect of GA-AS (904 nm) laser irradiation on the intact skin of the injured rat sciatic nerve. Lasers Med Sci 2003;18(2):83–88. [DOI] [PubMed] [Google Scholar]
- 162).Cömelekoğlu U, Bagiş S, Büyükakilli B, Sahin G, Erdoğan C, Kanik A. Acute electrophysiological effect of pulsed gallium-arsenide low-energy laser irradiation on isolated frog sciatic nerve. Lasers Med Sci 2002;17(1):62–67. [DOI] [PubMed] [Google Scholar]
- 163).Grolleau-Julius A, Ray D, Yung RL. The role of epigenetics in aging and autoimmunity. Clin Rev Allergy Immunol 2010;39(1):42–50. doi: 10.1007/s12016-009-8169-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 164).Hedrich CM, Tsokos GC. Epigenetic mechanisms in systemic lupus erythematosus and other autoimmune diseases. Trends Mol Med 2011;17(12):714–724. doi: 10.1016/j.molmed.2011.07.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 165).Huynh JL, Casaccia P. Epigenetic mechanisms in multiple sclerosis: implications for pathogenesis and treatment. Lancet Neurol 2013;12(2):195–206. doi: 10.1016/S1474-4422(12)70309-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 166).Picascia A, Grimaldi V, Pignalosa O, De Pascale MR, Schiano C, Napoli C. Epigenetic control of autoimmune diseases: from bench to bedside. Clin Immunol 2015;157(1):1–15. doi: 10.1016/j.clim.2014.12.013. [DOI] [PubMed] [Google Scholar]
- 167).Egger G, Liang G, Aparicio A, Jones PA. Epigenetics in human disease and prospects for epigenetic therapy. Nature 2004;429(6990):457–463. [DOI] [PubMed] [Google Scholar]
- 168).Berger SL, Kouzarides T, Shiekhattar R, Shilatifard A. An operational definition of epigenetics. Genes Dev 2009;23(7):781–783. doi: 10.1101/gad.1787609. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 169).Iridoy Zulet M, Pulido Fontes L, Ayuso Blanco T, Lacruz Bescos F, Mendioroz Iriarte M. Epigenetic changes in neurology: DNA methylation in multiple sclerosis. Neurologia 2017;32(7):463–468. doi: 10.1016/j.nrl.2015.03.011. [DOI] [PubMed] [Google Scholar]
- 170).Penas C, Navarro X. Epigenetic Modifications Associated to Neuroinflammation and Neuropathic Pain After Neural Trauma. Front Cell Neurosci 2018;12:158. doi: 10.3389/fncel.2018.00158. eCollection 2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 171).van den Elsen PJ, van Eggermond MC, Puentes F, van der Valk P, Baker D, Amor S. The epigenetics of multiple sclerosis and other related disorders. Mult Scler Relat Disord 2014;3(2):163–175. doi: 10.1016/j.msard.2013.08.007. [DOI] [PubMed] [Google Scholar]
- 172).Arthur-Farraj PJ, Morgan CC, Adamowicz M, Gomez-Sanchez JA, Fazal SV, Beucher A, Razzaghi B, Mirsky R, Jessen KR, Aitman TJ. Changes in the coding and non-coding transcriptome and DNA methylome that define the schwann cell repair phenotype after nerve injury. Cell Rep 2017;20(11):2719–2734. doi: 10.1016/j.celrep.2017.08.064. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 173).Oh YM, Mahar M, Ewan EE, Leahy KM, Zhao G, Cavalli V. Epigenetic regulator UHRF1 inactivates REST and growth suppressor gene expression via DNA methylation to promote axon regeneration. Proc Natl Acad Sci USA 2018;115(52):E12417–E12426. doi: 10.1073/pnas.1812518115. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 174).Pereira JA, Lebrun-Julien F, Suter U. Molecular mechanisms regulating myelination in the peripheral nervous system. Trends Neurosci 2012;35(2):123–34. doi: 10.1016/j.tins.2011.11.006. [DOI] [PubMed] [Google Scholar]
- 175).Salzer JL. Schwann cell myelination. Cold Spring Harb Perspect Biol 2015;7(8):a020529. doi: 10.1101/cshperspect.a020529. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 176).Jagalur NB, Ghazvini M, Mandemakers W, Driegen S, Maas A, Jones EA, Jaegle M, Grosveld F, Svaren J, Meijer D. Functional dissection of the Oct6 Schwann cell enhancer reveals an essential role for dimeric Sox10 binding. J Neurosci 2011;31(23):8585–94. doi: 10.1523/JNEUROSCI.0659-11.2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 177).Svaren J, Meijer D. The molecular machinery of myelin gene transcription in Schwann cells. Glia 2008;56(14):1541–1551. doi: 10.1002/glia.20767. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 178).Le N, Nagarajan R, Wang JY, Svaren J, LaPash C, Araki T, Schmidt RE, Milbrandt J. Nab proteins are essential for peripheral nervous system myelination. Nat Neurosci 2005;8(7):932–40. [DOI] [PubMed] [Google Scholar]
- 179).Topilko P, Schneider-Maunoury S, Levi G, Baron-Van Evercooren A, Chennoufi AB, Seitanidou T, Babinet C, Charnay P. Krox-20 controls myelination in the peripheral nervous system. Nature 1994;371(6500):796–9. [DOI] [PubMed] [Google Scholar]
- 180).Ma KH, Svaren J. Epigenomic reprogramming in peripheral nerve injury. Neural Regen Res 2016;11(12):1930–1931. doi: 10.4103/1673-5374.197133. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 181).Stolt CC, Wegner M. Schwann cells and their transcriptional network: Evolution of key regulators of peripheral myelination. Brain Res 2016;1641(Pt A):101–110. doi: 10.1016/j.brainres.2015.09.025. [DOI] [PubMed] [Google Scholar]
- 182).Jacob C, Lebrun-Julien F, Suter U. How histone deacetylases control myelination. Mol Neurobiol 2011;44(3):303–312. doi: 10.1007/s12035-011-8198-9. Epub 2011 Aug 23. [DOI] [PubMed] [Google Scholar]
- 183).He X, Zhang L, Queme LF, Liu X, Lu A, Waclaw RR, Dong X, Zhou W, Kidd G, Yoon SO, Buonanno A, Rubin JB, Xin M, Nave KA, Trapp BD, Jankowski MP, Lu QR. A histone deacetylase 3-dependent pathway delimits peripheral myelin growth and functional regeneration. Nat Med 2018;24(3):338–351. doi: 10.1038/nm.4483. [DOI] [PMC free article] [PubMed] [Google Scholar]