Open access peer-reviewed chapter

Scaffolds for Peripheral Nerve Regeneration, the Importance of In Vitro and In Vivo Studies for the Development of Cell-Based Therapies and Biomaterials: State of the Art

Written By

Sílvia Santos Pedrosa, Ana Rita Caseiro, José Domingos Santos and Ana Colette Maurício

Submitted: 02 December 2016 Reviewed: 02 May 2017 Published: 13 December 2017

DOI: 10.5772/intechopen.69540

From the Edited Volume

Scaffolds in Tissue Engineering - Materials, Technologies and Clinical Applications

Edited by Francesco Baino

Chapter metrics overview

2,401 Chapter Downloads

View Full Metrics

Abstract

Human adult peripheral nerve injuries are a high incidence clinical problem that greatly affects patients’ quality of life. Although peripheral nervous system has intrinsic regenerative capacity, this occurs in an incomplete or poorly functional manner. When a nerve fiber loses its continuity with consequent damage of the basal lamina tubes, axon spontaneous regeneration is disorganized and mismatched. These phenomena translate in an inadequate nerve functional recovery and consequent musculoskeletal incapacity. Nerve grafts still remain the gold standard in peripheral injuries treatment. However, this approach contains its disadvantages such as the necessity of primary surgery to harvest the autografts, loss of a functional nerve, donor site morbidity and longer surgery procedures. Therefore, biomaterials and tissue engineering can provide efficient resources and alternatives to nerve injury repair not only by the development of biocompatible structures but also, introducing neurotrophic factors and cellular systems to stimulate optimum clinical outcome. In this chapter, a comprehensive state-of-the art picture of tissue-engineered nerve grafts scaffolds, their application in nerve regeneration along with latest advances in peripheral nerve repair and future perspectives will be discussed, including our own large experience in this field of knowledge.

Keywords

  • nerve regeneration
  • peripheral nerve
  • biomaterials
  • hydrogels
  • tube-guides
  • neurotrophic factors
  • cell-based therapies
  • functional assessment
  • mesenchymal stem cells
  • Schwann cells
  • tissue engineering
  • scaffolds

1. Introduction

Tissue engineering was originally defined by Skalak and Fox, in 1988 [1] as ‘the application of the principles and methods of engineering and life sciences towards the fundamental understanding of structure-function relationships in normal and pathological mammalian tissues and the development of biological substitutes to restore, maintain, or improve functions’. Later, Langer and Vacanti [2], in a widespread review paper, defined three main pillars of tissue-engineering principles, the (a) isolated cells and substitutes—cellular systems; (b) tissue-inducing substances—bioactive molecules; and (c) scaffolds, biomaterials and/or matrices.

The first strategy concerns cell-based therapies in which cells in a small volume or in cell sheets are transplanted into the body. Cellular system includes a wide range of cells and most significantly stem cells [37]. Stem cells are responsive undifferentiated cells with varying degrees of self-proliferation and differentiation plasticity [8]. Although the number of stem cells is higher before birth, in adults there are still several ‘niches’ with significant number of stem cells [9]. The second pillar focuses on the [5, 7, 10] bioactive molecules that can be signalling molecules, proteins and oligonucleotides that can enhance cell migration, cell growth and/or differentiation. These bioactive molecules are roughly divided into mitogens, growth factors and morphogens. Finally, the third pillar is the three-dimensional structure that provides shelter and structure for the cellular system [57, 11]. Usually, the biomaterials or scaffold mimics the environment and natural extracellular matrix (ECM) of the place of implantation and should be biocompatible such as their metabolites. Also, scaffolds can be used as drug-delivery system in the controlled release of bioactive molecules [9, 12, 13].

Biomaterials according to the American National Institute of Health describes ‘any substance or combination of substances, other than drugs, synthetic or natural in origin, which can be used for any period of time, which augments or replaces partially or totally any tissue, organ or function of the body, in order to maintain or improve the quality of life of the individual’. Earlier, the Williams Dictionary of Biomaterials [14] defined biomaterial as ‘any substance intended to interact with the biological system in order to replace living matter which has lost its function. It can serve as a vehicle or not, matrix, support, or for stimulating new tissue growth’. The selection of the most adequate material for a given application should fulfil several requirements of physical, mechanical, chemical and biological properties. The most important features of biomaterials must be [7] (i) biocompatibility, that is, the biomaterial itself must not cause any harm in the living system; (ii) biofunctionality, since the biomaterial must feature mechanical and physico-chemical properties adequate to the function and intended application; and (iii) sterilizability, while materials must be able to undergo sterilization procedures, especially the polymeric materials. Biocompatibility is considered the main feature of a biomaterial and represents the response of the living system to the introduction of a foreigner material. It can be defined as the ‘ability of a biomaterial to perform its desired function with respect to a medical therapy, without eliciting any undesirable local or systemic effects in the recipient or beneficiary of that therapy, but generating the most appropriate beneficial cellular or tissue response to that specific situation, and optimizing the clinically, relevant performance of that therapy’ [14]. However, biocompatible biomaterials are not useful if they are not biofunctional. Similarly, bioactive devices cannot be used if they are not biocompatible. The term biofunctionality can be simply explained as the suitability to the function [7]. Scaffold porosity is an important desirable feature in the majority of scaffolds, as it promotes cell seeding and cell-matrix interaction and leads to increased neovascularization. However, the exact pore size depends on application, the average pore diameter of 20–125 µm is adequate of skin tissue and >300 µm, in bone tissue [11, 15]. The macro- and micro-topography and other physico-chemical properties of the scaffolds influence cell attachment, migration, proliferation and differentiation and promote protein and other factors adsorption and therefore the success of the system [11, 16]. The mechanical properties and specially the degradation kinetics also is a key feature, especially for musculoskeletal and neuromuscular repair due to the slower repair rates.

In this chapter, a comprehensive state of the art of tissue engineering focused on peripheral nerve repair and the advances on materials and nerve grafts will be discussed, as well as our own large experience in this field of knowledge and the future perspectives.

Advertisement

2. Peripheral nerve regeneration

Peripheral nerve injury remains a major public health problem with an estimated incidence of 13–23 cases per 100,000 persons [17, 18]. These injuries may have a traumatic or an iatrogenic cause and usually are associated with pain, decrease of function and sensory sensibility with devastating effects on patients and family lives [3, 17, 19, 20].

Aegineta et al. [21] for the first time performed nerve repair and wound closure in wounded soldiers, as a military surgeon. Then, in 1873, Hunter [22] first described the epineural nerve repair procedure, still in use today. Sunderland portrayed the principles of nerve repair resourcing to microsurgical techniques, and Kurze and Smith were able to apply those principles in 1964, thanks to the advance in microscopy [2325].

Peripheral nerve injuries treatment understands the most challenging surgical procedures, and despite the major breakthroughs in this area, complete nerve recovery and nerve function in all clinical cases have not yet been achieved [17, 20, 26]. Despite the exquisite surgical techniques, poor recovery outcome results from nervous system intrinsic and extrinsic factors, such as the integrity of the surrounding tissues post lesion, type and level of the injury itself, the effect on the spinal cord and neurons, the compromising of end organs and with key importance the timing of the surgery [17, 2729]. Also, although peripheral nervous system has spontaneous regeneration ability, there is a very limited prospective of spontaneous recovery, mostly concerning the complete functional neuromuscular recovery [20].

2.1. Peripheral nerve anatomy

The peripheral nerve system is composed by neurons, Schwann cells (SCs), fibroblasts, macrophages and interconnected blood system [20, 30]. Motor and sensory neurons are polarized cells whose bodies reside in the spinal cord and with long cytoplasm called axon. Their terminations, called dendrites, target a site of innervation. The signal conduction originates in the axon hillock, in the cell body and projects itself in synapses with target end organs. Axons plasma membrane is partially enclosed by the SCs that produce myelin that encapsulates the axon and helps with signal transmission. Myelin is therefore an insulator that enhances the signal transmission efficiency down the axon. Then, there is a connective tissue net that surrounds the individual axons called the endoneurium. An arrangement of axons, designed fascicles, is surrounded by the perineurium, and groups of fascicles are separated by the epineurium. External to this layer is the blood supply derived from major arteries and the latter involved by the mesoneurium (Figure 1). The conservation of fascicles patterns and connective tissue is vital for optimal nerve repair and regeneration. Therefore, more commonly, the epineurium is sutured in end-to-end suture (called epineural end-to-end suture), and all nerve surgical interventions are strictly directed at these connective tissue layers. The most important feature is the fact that these sutures must be tension free; otherwise, they will compromise the nerve blood supply and the process of regeneration itself [20, 3032].

Figure 1.

Peripheral nerve anatomy. The figure was adapted from Ref. [30].

2.2. Nerve response to injury

Nerve injuries can be from a chronic or acute nature, and SCs are the major cause of chronic injuries [20, 30]. Acute injuries are mediated by axonal degeneration and occur in a sequence of events proximally and distally from the zone of trauma. The initial stages of degeneration occur proximally to the lesion as programmed cell death, called chromatolysis [33, 34], and distally to the lesion, through Wallerian degeneration of the distal axonal segment [35, 36]. Within 24–48 h of the injury, SCs degrade the myelin and phagocytes debris from distal axons [37, 38]. The proximal portion of the axon also degenerates up to the node of Ranvier, where the axonal regrowth occurs. Then macrophages recruitment occurs, originating growth factor release and fibroblast and SCs proliferation. The SCs form organized longitudinal structures inside the endoneurium—called bands of Büngner [30, 34]—that are critical to the axonal regeneration. At the distal site of lesion, the node of Ranvier, around 50–100 finger-like sprouts, starts to form a growth cone directed to the distal nerve stump [39]. Proteases are also released from the growth cone by the influence of several factors, clearing the way towards the target tissue. Nerve growth factor (NGF), brain-derived growth factor (BDNF) and other neurotrophic factors are upregulated by SCs, and their expression is increased which promotes the migration and proliferation of the SCs [20]. Axon existing actin allows axon elongation and which occurs in a 1–3 mm/day rate [36] until a receptor is reached. If no receptor or endoneurial tube is reached, axon continues to grow but in a disorganized manner, causing neuroma and clinically painful lesions [40]. In severe nerve injury, axon regeneration is further disorganized with additional scaring and pourer regeneration.

2.3. Nerve injury grading

Nerve injuries were firstly classified into neuropraxia, axonotmesis and neurotmesis by Seddon [41], after his World War II experience in treating nerve-injured soldiers. Neuropraxia is characterized by the segmentation of the myelin without disturbance of the axon, usually a consequence of a compression. Typically, it resolves itself, once the myelin is restored, within 12 weeks. Axonotmesis concerns axonal injury and occurs from a crush mechanism. In this case, connective tissue and nerve continuity are not affected but are followed by Wallerian degeneration. Axonal regeneration occurs at 1–3 mm/day rate [36], and depending on the distance of the lesion, an incomplete recovery may happen. Neurotmesis comprehends the anatomical and physiological section of the axons and connective tissues. Therefore, no spontaneous regeneration may occur and needs surgical reconstruction [30, 42]. Sunderland later expanded this classification by including five types of injury, based on histological knowledge that allowed further distinction between axonotmesis injuries [25, 43]. Sunderland grade Type I classification is equivalent to neurapraxia. The Type II, III and IV classifications differentiate axonotmesis injuries based on the commitment of the connective tissues. In Type II class injury, there is axonal damage without commitment of the endoneurium, and therefore, it is possible to achieve a full recovery. In Type III lesions, we find axon impairment affecting the endoneurium, and in Type IV besides endoneurium, there is perineurium damage. Sunderland grades III and IV may heal spontaneously, but there is attendant scaring and increasing axon and connective tissue damage that causes incomplete recovery. Type IV lesion usually implies surgical intervention and results in extensive scaring. Scars are associated with pain and nerve conduction impairment and may require reconstructive surgery. Type V Sunderland classification corresponds to neurotmesis [30, 42]. Finally, Mackinnon and Dellon [44] described a mixed type of lesion degree, a Type VI addition to the Sunderland classification. This classification represents probably the most common type of lesions, with several layers of injury and not necessarily traditional model as described by Sunderland. The recovery potential and also the treatment approach vary, according to the type of lesion, considering the three classifications mentioned earlier.

In Table 1, the major findings in nerve injury grading according to Seddon [41], Sunderland [25, 43] and Mackinnon and Dellon [44] addition are described.

Sunderland Seddon Characteristics Spontaneous recovery potential
Type I Neuropraxia Injury to myelin sheath only Full
Type II Axonotmesis Injuries involve the axon only Full
Type III Axonotmesis Injuries involve the axon and disrupt the endoneurium Usually slow or incomplete
Type IV Axonotmesis Injuries involve the axon and disrupt the endoneurium and perineurium Poor to none
Type V Neurotmesis Complete disruption of the nerve; with the epineurium None
Type VI* Mixed Combination of Types II, III and IV Variable, can be poor to none

Table 1.

Nerve injury classification according to Sunderland and Seddon.

Mackinnon and Dellon [44] addition to the Sunderland [25, 43] classification.


2.4. Diagnosis

Nerve injury may involve variable lengths of nerve impairment, and the degree of the lesion is affected by the type of lesion [45]. Also, the prognosis is dependent on the age of the patient, location of the lesion—distal fare better than proximal lesions—and also demographics [42]. In terms of nerve electrical signal and electrophysiology, the absence of electrical conduction may not indicate severe nerve damage, since conduction may be recovered after just 1 week. Clinical examination (including the functional evaluation) and surgical inspection still are the most accurate means to obtain diagnosis. However, non-invasive procedures such as nerve conduction studies (NCSs) and electromyograms (EMGs) have a diagnostic role in the delayed setting, when muscle fibrillation occurs [30, 42]. NCS assesses both motor and sensory functions through a voltage stimulator applied to the skin at different points of the nerve. A sensor detects response at the muscle (motor function) or nerve (sensory function). This is the initial screening test for the presence or absence of conduction signal. The EMG assesses only the motor function, and in this test, a needle is inserted in the muscle to assess the resting electrical activity and voluntary motor unit analysis [30]. In Figure 2, the representation of an algorithm of best treating approach selection according to the type of lesion, length and also complementary diagnostic results is shown. Sciatic Function Index (SFI) is one of the most widely used forms of functional assessment. It compares parameters from footprints and mathematically infers about sensory-motor gait function mediated by the sciatic nerve, without requiring terminal assessment [46, 47]. Sciatic Static Index (SSI) was first introduced by Bervar [48, 49] and is another way of assessing recovery of function after sciatic injury in animal models. It also uses the footprints in a static position and minimizes bias related to gait’s velocity. Also, the SSI improves the acquisition of footprints and is more repeatable and accurate than the SFI. Other motor performance index is measuring the extensor postural thrust (EPT) and nociceptive function using the withdrawal reflex latency (WRL). EPT is induced by lowering the affected hindlimb towards the platform of a digital balance supporting the animal by the thorax. During the test, the rat extends the hindlimb and the distal metatarsus and digits connect with digital platform balance [5054]. Nociceptive function using the withdrawal reflex latency was described by Masters and colleagues [55] and is based on the fact that rats without sciatic nerve injury withdraw their paws from the hotplate within 4.3 s or less, when this period of time is increased, it is a symptom of impaired nerve conduction [5153, 5661]. Nuclear magnetic resonance (RMN) imaging could become a valuable tool in nerve injury diagnosis since it allows fine, detailed evaluation nerve anatomy and pathology due to excellent image resolution [42]. Also, functional diffusion tensor imaging (DTI), a new diagnosis tool, has been showing peripheral nerve sheath tumours; however, its application on differentiating various grades of injuries remains to be tested [42].

Figure 2.

Peripheral nerve repair treatment diagram.

2.5. Timing of medical intervention

After peripheral nerve injury, repair events begin to take place. Primary repair events occur within the first couple of days. However, the rate of axon regeneration is very slow, as previously referred—it is 1–3 mm/day [36]—and no therapeutic methods have yet improved this regeneration rate [30]. However, it is consensual that early nerve repair results in improved functional outcomes, as described by Mackinnon and Dellon [44]. Furthermore, there is a set period of 12–18 months in which muscle re-enervation can occur before irreversible motor end-plate degeneration occurs, and the neurogenic atrophy takes place [30]. Slow axonal regeneration associated with muscle structural changes and increasingly degraded stromal environment contribute for an incomplete functional recovery. Muscle fibrosis and atrophy phenomena begin immediately after denervation and are called neurogenic atrophy. After 4 months, a plateau is reached, when 60–80% of muscle mass is lost, and although motor end plates increase, beyond a 12-month period, a functional muscle re-enervation is highly unlikely [30, 39, 62]. The time frame for sensory re-enervation is longer but not endless, and early repair also grants better results [30, 39]. In Figure 2, we present the peripheral nerve injuries repair algorithm which helps to understand the variables taken into account in the selection of best nerve repair strategy.

2.6. Nerve repair strategies

2.6.1. Direct nerve repair

Direct nerve repair with epineural end-to-end sutures using microsurgery techniques is still the gold-standard surgical treatment for severe neurotmesis injuries, but only in cases where well-vascularized tension-free coaptation can be achieved [30, 37, 6365]. The procedure involves rough fascicular matching between proximal and distal nerve ends and the alignment of nerve fascicles and epineural blood vessels [30, 66]. Other types of direct repair consist in fascicular repair or grouped fascicular repair. This requires intranerval dissection and direct matching and suturing of fascicular groups. Despite better fascicle alignment, this procedure is no better than epineural repair in functional outcomes and in fact is associated with more traumas and scaring [30, 63, 66]. Complementary assistance techniques of histologic staining using acetylcholine esterase and carbonic anhydrase, electrical stimulation during the procedure in awaken patients and visual observation of surface vessels, are visual orientations to the surgeon that grant the success of the procedure [63, 67]. Another possible approach is the use of tissue adhesives such as fibrin glue to supplement or replace sutures, creating a gel-like clot at the nerve ends. The advantages of this procedure are the efficiency and practicality, the reduced trauma and scaring due to a barrier effect. The major disadvantage of this technique is the inferior holding strength more subject to stress [63, 68].

2.6.2. Nerve grafts

When peripheral nerve injury originates a significant gap (>3 cm) between the nerve ends with excessive tension for direct epineural repair and reversed interposition, nerve grafts are required [30, 63]. Such gaps may occur in severe neurotmesis lesions or in axonotmesis stretch injuries in which long regions of the nerve may be damaged in the setting of a lesion-in-continuity [63, 69]. Nerve grafts are single, cable, trunk, interfascicular or vascularized portions of the nerve with similar diameter to the affected [30, 70, 71]. Nerve grafting may be from autologous or allograph origin. Xenografts have been described as viable alternatives but require extensive immunosuppression and prionic diseases transmission if they are from ruminants [72, 73]. Nerve autografts are considered the gold standard since they provide appropriate neurotrophic factors and viable SCs, both essential for axonal regeneration without immune compromise [63, 74]. For the choice of the autologous grafts, many factors must be taken into account, such as the size of the nerve gap, the location of proposed nerve repair and associated donor-site morbidity [63, 74]. Grafts are either sutured to the epineurium of single nerves or more commonly to the perineurium of individual fascicles, depending on nerve calibre, type and location [30, 63, 74]. The interfascicular nerve graft was described by Millesi et al. [75]. Vascularized nerve graft was designed by Taylor and Ham, whereby the donor nerve is transposed with its arterial and venous supply into the graft site [76]. Terzis and Kostopoulos [71] clinically demonstrated that medium-sized trunk grafts, which would normally undergo central necrosis, could be transferred as vascularized nerve grafts and survive. However, autografts sacrifice a functioning nerve, usually a sensory nerve, to substitute a more important injured motor nerve. Therefore, sensory loss and scarring at the donor site, where neuroma and pain phenomena, are expected [30, 63, 77]. Autologous nerve graft undergoes Wallerian degeneration and therefore just provides support and guidance for the ingrowing axon. Also, fascicle mismatch, scarring and fibrosis of the repair site is unavoidable and is caused by the injury, tissue handling and suture itself. [30, 63, 77]. An alternative to autologous nerve grafting is the use of nerve allografts. The advantages of allografts are no donor supply limitations or donor-site morbidity, accessibility and unlimited supply of neuronal tissue. However, there are significant costs and complexity with their use, such as immunosuppression [30, 63, 72, 73, 77]. Several techniques have been used to reduce allograft antigenicity, such as cold preservation, irradiation and lyophilization and certainly patients’ immunosuppressive therapy. However, it is proven that immune response is caused by SCs, and once their migration has occurred, approximately 24 months after nerve repair, systemic immunosuppression can be withdrawn [74, 78]. To avoid immunosuppression, nerve allografts are decellularized by a process of chemical detergent, enzyme degradation or irradiation, resulting in an acellular nerve scaffold [30, 63, 79]. Similarly, in tendon transfer, a distal function is treated at the expenses of a secondary function [30, 63, 79].

2.6.3. Nerve transfers and free-functioning muscle transfer

The definition of nerve transfer is the surgical coaptation of a healthy nerve donor to a denervated nerve [30, 63, 80]. The procedure was first described by Harris in 1921 [81], in the treatment of low median nerve injury suffered during World War I. The major disadvantage is finding an expendable donor nerve near the target muscle with a large enough motor fibre population [30, 63]. Free-functioning muscle transfer (FFMT) is another treatment approach, in severe injuries and especially in secondary reconstructions. The procedure entails the transfer of a healthy muscle and its neurovascular pedicle to a new location to assume a new function [71, 82]. Since it is a complex procedure, it is only considered as a secondary reconstructive surgery.

2.6.4. Nerve conduits

Nerve gap repair and nerve grafts have its complications. In the procedures described earlier, the nerve repair requires a second incision site for autograft harvest, donor-site morbidity, loss of a functional, usually sensory nerve, and long-surgery procedure [63, 65]. The described disadvantages triggered the development of nerve conduit or nerve guides to bring a new approach for nerve gap repair [40, 83]. Also, developments in tissue engineering and regenerative medicine, and research in artificial and natural biomaterials have enabled the development of the first nerve conduits [73]. Nerve tubulation approach to the repair of peripheral nerve gaps can be traced back to the nineteenth century. Gluck (Gluck, 1881) performed the first experiment of nerve tabulation, with a tube of decalcified bone to aid the approximation of transected nerve ends, in animals [78, 84, 85]. Later, Dahlin and Lundborg [86] developed the first synthetic tube, made of silicone. Their work was also pioneer in the characterization of the mechanism of regeneration within the lumen of the engineered tube [78, 86]. A nerve conduit is a tubular structure made of biological or synthetic materials designed to bridge the gap of a sectioned nerve. It is used when primary end-to-end direct repair is not possible, to protect the nerve from scar formation, to prevent fluid from leaking from the nerve stump and to guide the axon nerve cone into the distal nerve stump [63, 65, 78]. The fluid formed from the transected nerve ends is essentially made of fibrin, which forms a matrix or a hydrogel matrix between the nerve ends that is able to support cell migration. Cell migration within the fibrin matrix creates some linear bands—bands of Büngner—that steer the growth of the nerve cone [78, 86]. The mechanism through which neurite growth cone forms within the lumen of the conduit depends on the volumetric ratio [87, 88]. If the gap is too long or the diameter of the inner lumen is too large, the growth cables are too thin, and due to the fibrin matrix, the growth cone takes on an hourglass figure that affects axonal regeneration. Nectow et al. [89] also studied the effect of the defect size in the regenerative process through nerve conduits. Nerve conduits provide control environment to outgrowing axons, migration of SCs and neurotrophic stimulation by the distal stump crucial for optimal regeneration of nerve function [40, 90]. This approach is usually reserved for gap defects between 1.5 and 3 cm [91, 92]. As early as 1994, Brunelli et al. [91] defined four factors for an ideal nerve conduit material: (i) biocompatibility, (ii) easy preparation and tailoring, (iii) incorporation of neurotrophins and stimulating substances, and (iv) protection against scaring. Recently, Arslantunali et al. [93] defined the desirable features for a nerve conduit, as flexibility, biocompatibility, biodegradability, high porosity, neuroinductivity, neuroconductivity, easy handling and sufficient endurance. Nowadays, the second- and third-generation nerve conduits are becoming Food and Drug Administration (FDA) approved and reaching the marked, so, many more pre-clinical and clinical trials are demonstrating major breakthrough in this area. In Figure 3, we represent the major features of nerve conduits, and the modification researchers have made to introduce neurotrophic elements that enhance nerve regeneration and peripheral nerve repair.

Figure 3.

Different types of nerve conduits and their main features. Nerve conduits modification that functions as nerve regeneration enhancers.

2.6.5. Biological conduits

The use of non-neuronal tissue as conduits was first reported by Büngner (Büngner, 1891), when he successfully used a segment of human brachial artery to regenerate a gap in sciatic nerve [84]. The use of arterial grafts has been demonstrated [94, 95] but is associated with high morbidity. Also, lack of donor vessels makes this a less popular approach in nerve repair. However, it has been described that neurovascular injuries in the hand have benefited from the use of homolateral arteries in the repair [40, 84]. Frerichs et al. and Kim at al. [96, 97] have used acellular allogenic nerve grafts effectively in the regeneration nerve gaps in a rat sciatic model. A similar approach was approved by such Food and Drug Administration and is commercially available as Avance®, by AxoGen, Inc. (Alachua, FL, USA) (Table 2). Veins have also been a viable option for nerve repair. The risk of vein collapse led to their filling with nerve or muscle tissue. This supplementation has the added advantage of supplying neurotrophic factors and ECM (muscle fibres) and has been described to facilitate nerve regeneration across longer gaps by promoting SCs migration, cell proliferation and guidance of the axonal growth cone [84]. Another type of biological conduit is tendon autograft. Although with only theoretically and historic interest, ECM macrostructure and the presence of hyaluronic acid have been described to enhance regenerating in the nerve cone [84]. AxoGuard™ is the only FDA-approved device composed by small intestine submucosa (SIS) extracellular matrix (Table 2). Preliminary studies in rat sciatic model showed distally directed growth of the proximal nerve [98]. Later further in vivo studies revealed better EMG response for distal motor latency and amplitude [99].

Name Composition Structure Length (cm) Degradation Time (months) Manufacturer FDA approval
NeuroTube Polyglycolic acid Absorbable woven mesh tube 2–4 3 Synovis Micro companies 1999
Saluleridge nerve cuff Polyvinyl alcohol Flexible tubular membrane 6.35 No degradation Salumedica LLC 2000
NeuraGen Collagen Type I Fibrillar, semipermeable 2–3 3–4 Integra LifeSciences Co. 2001
NeuroFlex Collagen Type I Flexible, Semipermeable and tubular 2.5 4–8 Collagen Matrix, Inc. 2001
NeuroMatrix Collagen Type I Flexible, Semipermeable and tubular 2.5 4–8 Collagen Matrix, Inc. 2001
Surgesis nerve cuff Type I, III, IV and VI collagen Extracellular collagen matrix 5 Reabsorbable Cook Biotech 2003
Neurolac Poly-dl-lactide-caprolactone Tubular 3 16 Polyganics BV 2003/2005
NeuraWrap Collagen Type I Flexible, Semipermeable longitudinal slit 2–4 36–48 Integra LifeSciences Co. 2004
NeuroMend Collagen Type I Semipermeable wrap that curls and enrols 2.5–5 4–8 Collagen Matrix, Inc. 2006
SaluTunnel Polyvinyl alcohol Tubular 6.35 No degradation Salumedica LLC 2010
Avance Processed human nerve allograft Tubular Variable No data AxoGen, Inc. 2010
Cova ORTHO-NERVE Type I collagen Rollable membrane 2.5–6 3–4 Biom’Up S.A. 2012
AxoGuard Extracellular matrix derived from porcine intestine Semipermeable, and absorbable tube Variable No data AxoGen, Inc. 2013
Flexible Collagen Nerve Cuff Collagen Type I Flexible, Semipermeable and tubular 2.5 No data Collagen Matrix, Inc. 2014
Nerve cuff Type I, III, IV and VI collagen Extracellular collagen matrix 1–5 Reabsorbable Cook Biotech 2014
Neuragen 3D Type I collagen and glycosaminoglycan (chondroitin-6-sulphate) Flexible, pliable tube with collagen-glycosaminoglycan inner matrix 6.35 9–12 Integra LifeSciences Corporation 2014
Reaxon Plus Chitosan Flexible, pliable tube 3 3 MEDOVENT GmbH 2015
Nerbridge polyglycolic acid and Type I and III collagen flexible, semipermeable tubular membrane filled with porous collagen 3–5 3–4 TOYOBO CO., LTD. 2016

Table 2.

Commercially available and FDA-approved nerve conduits.

2.6.6. Manufactured conduits

Manufactured nerve conduits can be divided as first-, second- and third-generation conduits. The first-generation conduits are non-resorbable, synthetic tubes made of silicone or polytetrafluoroethylene (ePTFE, Gore-Tex®) [65]. These conduits require a second surgery in order to remove the non-resorbable material. The original idea was to provide support, structure to guide axonal regrowth and form a stable barrier against connective tissue infiltration [73, 100, 101]. Synthetic nerve conduits made of ePTFE were successfully applied in a 4-cm nerve gap, in human [102]. Second-generation nerve conduits are resorbable, biocompatible tubes and are FDA approved and commercially available through different materials. The main advantage of these conduits is their permeability and resorbability that spares patients of a second surgery procedure. Third-generation nerve conduits contrary to second-generation may incorporate controlled release/delivery of neurotrophic factors, electroconductive material, stem cells or SCs, extracellular matrix proteins, surface micropatterning or luminal fillers [73, 103]. Already, two third-generation products have been approved by the FDA, namely NeuraGen® 3D from Integra LifeSciences Corporation and Nerbridge from Toyobo Co., Ltd. NeuraGen® 3D (K130557, approved in 2014) is a bovine Type I collagen conduit with a porous inner hydrogel matrix of collagen and glycosaminoglycan (chondroitin-6-sulfate). Nerbridge™ (K152967, approved in 2016) is a flexible, resorbable and semipermeable tubular membrane matrix filled with porous collagen that provides a non-constricting encasement for injured peripheral nerves for protection of the neural environment.

2.7. Materials

2.7.1. Collagen

Denaturated collagen conduits are available from a wide number of manufacturers and are in fact the most exploited material in nerve conduits [73, 78]. Collagen is a structural protein ubiquitous in the human body, particularly in the peripheral nerve system. Also, collagen supports cell proliferation and tissue regeneration [65, 73, 104]. As nerve conduits collagen allows the establishment of topographical cues that guide axons to regrow [105, 106] and has shown excellent cell adhesive properties that encourage cell attachment and proliferation [106, 107]. The degradation time of the collagen conduits is relatively prolonged and takes up to 48 months which can cause nerve compression and fibrosis [84]. The first commercially available collagen nerve conduit was NeuraGen®, from Integra Lifesciences, Princeton, NJ, FDA approved in 2001 (Table 2). Several studies regarding the efficacy of collagen conduits in peripheral nerve injuries have been stated. Bushnell and colleagues [108] performed in 2008 a retrospective study of the utilization of collagen conduits in digital sensory nerve gaps of up to 20 mm and demonstrated a significant recovery rate of 89%. In a retrospective review, Wangensteen and Kalliaine [109] reported on a large number of sensory nerve gaps of 2.5–20 mm repaired with collagen conduits in multiple body regions and concluded that clinically successful outcomes were only observed in 43% of the cases. A prospective cohort study was performed by Lohmeyer and colleagues in 2009 [110] in digital and palmar nerve gaps of 6–18 mm, and results showed meaningful recovery in 75% of patients. Later, in 2011, Taras et al. [111] reported a 73% meaningful recovery in 5- to 15-mm isolated digital nerve lacerations repaired with collagen conduits. In a niche approach, collagen conduits have been used in children suffering from plexus brachialis injury during birth [112]. Also, a significant number of in vivo studies on collagen conduits showed good functional outcomes in nerve reconstructions in rat, cat, dog and primate models [113115]. Researchers and surgeons have, however, raised their concern about these conduits due to its high cost, conduit stiffness, lack of flexibility and poor enhancement of nerve regeneration [116, 117]. Also, collagen conduit application on major peripheral nerve injuries is limited to median and ulnar nerve repairs at the wrist and only observed as an alternative to the classic epineural suture repair [78]. In a recent study, Monaco and colleagues [118] investigated the effect of three different sterilization methods, dry heat, ethylene oxide and electron beam radiation, on the properties of cylindrical collagen scaffolds with longitudinally oriented pore channels, specifically designed for peripheral nerve regeneration. Ethylene oxide exposure demonstrated to be the most suitable method for the sterilization of the proposed scaffolds, since β-sterilization significantly augmented scaffold enzymatic degradation. Currently, there are 10 commercially available and/or FDA-approved collagen nerve conduits (Table 2).

2.7.2. Fibrin

Fibrin is a fibrous, non-globular protein involved in the clotting of blood. It is a commonly used biomaterial as fibrin glue in nerve repair. Nonetheless, it can also be engineered into a hydrogel with aligned matrix or shaped into tubular conduits [84]. As fibrin glue, it is widely used in sutures, thanks to its semisolid structure that enhances haemostasis and integrity of the repair and also due to its angiogenesis, chemotaxis, leucocytosis and macrophage proliferation stimulation [119, 120]. When used in a conduit, fibrin has shown to promote axon regeneration and functional recovery in small gaps [116, 121, 122]. Rafijah and colleagues [123] have reported the use of collagen conduits filled with fibrin glue in 10-mm rat sciatic nerve defect. After 12 weeks, no inhibitory effect was observed on function, axonal regeneration and compound motor action potential compared to hollow collagen conduit.

2.7.3. N-fibroin

N-fibroin is a soluble protein, derived from silk with great potential due to their superior biocompatibility and low immunogenicity and mechanical stability upon degradation [73, 124]. Several studies have demonstrated the potential of N-fibroin in the regeneration of peripheral nerve injuries [124128]. Silk fibroin-based nerve guidance conduit with oriented filaments was produced by Yang and colleagues [129] and originated successful results in rat sciatic model. Researchers have also shown that silk-based conduits [130] and silk nanofibres [131] enhance cell-material interactions like cell adhesion, proliferation and differentiation. A new approach entails the development of conduits with multi-walled silk fibroin/silk sericin internal lumen, beneficial to nerve regeneration and outer sheath (the hollow poly(lactic-co-glycolic acid) conduits that provide strong mechanical protection. Engineered bionic conduit showed promising in vivo results [132]. Research has also reported the development of silk fibroin conduits loaded with nerve growth factors [133138]. Despite the promising results, no silk conduit has yet been FDA approved.

2.7.4. Chitosan

Chitosan is a natural polymer currently under investigation as nerve conduits due to its favourable biocompatibility, biodegradability and bioactivity [139]. Our group research tested the nerve-regenerative potential of chitosan membrane with N1E-115 cellular system in rat sciatic nerve crush injury. Results showed that freeze-dried chitosan Type III without N1E-115 cell addition was the only type of membrane that significantly improved post-traumatic axonal regrowth and functional recovery [52]. Recent study showed meaningful motor and sensory recovery in 30-mm defect in the nerve of the distal right forearm [140].

2.7.5. Polyglycolic acid

Polyglycolic acid (PGA) was the first FDA approved and commercially available nerve conduit—NeuroTube®, from Synovis Micro Companies Alliance, Birmingham, Ala. PGA is a common suture material and is more flexible and porous than silicone. PGA is degraded into lactic acid in 6–12 months [73, 90]. Therefore, critics claim that PGA may degrade faster than the regeneration process and resulting lactic acid may a have toxic effects [84, 141143]. Dellon and Mackinnon [144] were the first to report the use of PGA conduits as secondary reconstitution of digital nerve defects 3 cm or smaller in 15 clinical cases in monkeys. After 1 year, 86% meaningful recovery was reported. Later, in an attempt to compare PGA conduit results to conventional autograft repair, Weber et al. [145] conducted a randomized prospective multicentre trial in the reconstruction of long sensory nerve gaps up to 3 cm. After 1 year, 74% meaningful recovery was noted in the PGA group compared with 86% in the standard techniques group (P > 0.05). Further analysis showed that PGA conduits were equivalent or superior to traditional autografts in less than 4- and 9–30-mm gaps [145]. Other researchers also compared PGA conduits and vein grafts to repair digital nerve gaps up to 4 cm and equivalent or superior recovery was obtained [146, 147]. Further experiments have demonstrated the success of bioabsorbable PGA nerve conduits in the regeneration of nerve defects [148151]. The most recent FDA-approved PGA conduit is Nerbridge®, from Toyobo Co., Ltd., which is a flexible, semipermeable tubular membrane filled with porous Type I and III collagens (Table 2).

2.7.6. Poly (d, l lactide-co-ε-caprolactone)

Poly-d, l lactide-co-epsilon-caprolactone (PCL) consists in lactic acid and caprolactone monomers. Their nerve conduits are resolvable polyester with the advantage of being transparent and with less acidic degradation product that therefore causes less toxic reaction [60, 84, 116, 152]. Also, PCL conduit is easy to produce, has a low-cost processing and has a long degradation time up to 16 months. However, resulting conduits have higher rigidity and more difficult to handle in clinical settings [84]. Also, Duda et al. [153] reported a strong foreign body response in PCL conduits. Currently, Neurolac® [154156] is the only FDA-approved caprolactone conduit (Table 2). Several reports have been made in the application of PCL conduits in sciatic rat nerve repair [157162]. Bertleff et al. [154] performed a randomized prospective multicentre study where PCL conduits were comparable to either primary end-to-end repair or nerve autograft. However, latter research showed no meaningful recovery in digital nerves repair [155, 156]. Secer and colleagues [163] studied the use of PCL in the recovery of 455 patients with ulnar nerve injuries. PCL conduits filled with muscle tissue showed superior results in comparison to single PCL conduits in 10- and 15-mm gap in sciatic nerve rat model [36, 156, 164168].

2.7.7. Polyhydroxybutyrate

Polyhydroxybutyrate (PHB) is a polyester polymer, also used in sutures and wound dressings [116, 169]. PHB has a long degradation time, up to 24–30 months [116, 169]. It has been reported that PHB has a neuroprotective effect and can help axon regeneration [170, 171]. Recently, Axongen Pharmaceuticals has a pending approval order of a PHB conduit. A study in which PHB wrap implants were used in human patients showed promising results compared to epineural suture [172].

2.7.8. Polyvinyl alcohol

Polyvinyl alcohol (PVA) [36, 89, 93] is the only nondegradable synthetic nerve conduit approved by the FDA—SaluBridge and SaluTunnel, from SaluMedica LLC, Atlanta, GA, USA (Table 2).

2.7.9. Polyhedral oligomeric silsesquioxane

Polyhedral oligomeric silsesquioxane (POSS) can be described as the smallest particles of silica [116]. The combination of POSS and PCL has been employed in the fabrication of peripheral nerve conduits and in vivo clinical studies to show the potential translation into clinics [117].

2.8. Optimizing nerve regeneration

Although nerve conduits provide sufficient guidance for regeneration of nerve defects, the development of new generation of scaffolds is under way. Third-generation conduits include artificial conduits that may incorporate controlled release/delivery of neurotrophic factors, electroconductive material, stem cells, SCs, extracellular matrix proteins, surface micropatterning, luminal fillers and guidance structures [73, 103].

2.8.1. Conduits structure modulation

Surface micropatterning and the inclusion of extracellular matrix proteins are new approaches that can provide suitable nanostructure topography for adequate neural growth and simulate topographical dimensions that mimic native nerve extracellular matrix [103, 173]. Coating of peripheral nerve conduits can enhance nerve regeneration process and solve longer nerve gaps repair. Resource to extracellular matrix materials, such as fibronectin, laminin and collagen, give naturally hydrophobic scaffolds a hydrophilic surface that promotes cell adhesion [73, 116, 174]. Collagen Type I conduits coated with laminin and fibronectin have shown improved neural regeneration [175, 176]. With that intend, new peptides have been engineered to mimic the active binding domains of various extracellular matrix molecules [177, 178]. Coating with arginylglycylaspartic acid (RGD) sequences (the tripeptide Arg-Gly-Asp) also obtained very good results [117, 179182]. Structure pore design is a great strategy to promote nerve regeneration. Controlling the architecture of the conduit wall is possible to develop a microporous inner layer and macroporous outer layer and obtain a bidirectional permeability [86, 103]. The diameter of the pores plays a critical role in the efficiency of the scaffolds since it influences cell attachment, axon regeneration and diffusion of nutrients [183, 184]. Electrospinning is a frequently used technique in bioengineering to produce imprinted micropatterns and can be used as a luminal guidance strategy. The advantages associate with electrospun conduits are (i) highly flexible and porous materials; (ii) high surface area-to-volume ration, thereby great availability for protein absorption, stem cells migration and regeneration of axons; (iii) fibers can be preferentially aligned, resulting in increased SC alignment, proliferation and growth, and the promotion of guided axonal growth [116, 185, 186]. Uneven fibre distribution and nerve growth inhibition caused by fibre overlapping are some of the disadvantages associated with this type of conduits. However, association with wall guides helps to avoid this problem [105, 187, 188].

2.8.2. Luminal fillers

Biomaterials and other strategies as conduits luminal fillers aim to change its microenvironment in a favourable way. The goal of this strategy is to promote axon regeneration inside the conduit and the restoration of motor and sensory nerve function.

2.8.2.1. Cellular systems

Stem cell therapies have received increased attention in regenerative medicine [189201]. The use of cellular systems inside nerve conduits is intended to promote axon regeneration [202206]. Several cells have been used with this intention, SCs, bone marrow stem cells (BMSCs) including more specifically the mesenchymal stem cells (MSCs). SCs are the natural glia of the peripheral nervous system and have been used successfully with beneficial effects in nerve reconstruction [204, 207210]. However, there is limited availability, and it requires previous surgery. Stem cells have the opportunity to show their potential since they are able to secrete neurotrophic factors and provide a favourable microenvironment for neurogenesis and the proliferation of SCs in peripheral nerve repair [211] and also are able to differentiate into Schwann cell-like phenotype [200, 212, 213]. MSCs therefore seem the most attractive approach as cellular system in peripheral nerve regeneration. MSCs all share mesenchymal markers that differentiate them from other cells that are positive staining for CD10, CD13, CD29, CD44, CD90 and CD105, and negative expression of haematopoietic markers [214]. Their main feature is their ability to proliferate and self-renew in a sustained manner, high plasticity and low immunogenicity, and differentiate into multiple mesodermal cells, including neuron-like cells [215, 216]. Also, they are major histocompatibility complex (MHC) class II negative and also their MHC class I expression levels can be manipulated; therefore they do not require the use of immunosuppressive drugs [217, 218]. MSCs can be isolated from several origins, including skin, hair follicle, periosteum, amniotic fluid, umbilical cord blood adipose tissue and dental pulp [53, 56, 200, 215, 219221]. Bone marrow represents the most commonly used tissue source of adult MSCs. BMSCs have limited availability especially in adult life and there is donor-site morbidity associated with its harvesting. Therefore, alternative sources have been reached. MSCs exist in the connective tissue (Wharton’s jelly (WJ)) of human umbilical cord and can be harvested easily [214, 218, 222, 223]. Several researchers have demonstrated that umbilical cord–derived stem cells can be differentiated into neuronal phenotype [214, 218, 222, 223] and have demonstrated potential utility in neurodegenerative diseases [224, 225]. Our group research has vast experience in the application of mesenchymal stem cells in peripheral nerve regeneration [50, 52, 53, 5661]. In our most recent work [56], we report the therapeutic value of MSCs isolated from the Wharton jelly in nerve repair associated to different tube guides made of biodegradable and biocompatible biomaterials. Biomaterials like PVA, PVA loaded with functionalized carbon nanotubes (PVA-CNTs), PVA loaded with polypyrrole (PVA-PPy) and PLC associated to MSCs were tested in terms of cytocompatibility and in vivo in the rat sciatic nerve neurotmesis injury model. The functional recovery was assessed serially for gait biomechanical analysis, by EPT, SFI and SSI, and by WRL. Results showed that MSCs enhanced the recovery of sensory and motor function in neurotmesis injuries showing a thicker myelin sheath. Other authors have reported the application of bone marrow stem cells and Schwann-like cells in the regeneration of facial nerves in rats [219].

Other sources of stem cells have been used in peripheral nerve injuries, such as adipose-derived stem cells [203, 206, 212, 226231] and dental pulp stem cells (DPSCs) [232236]. Dental pulp stem cells have also demonstrated differentiation capacity towards multiple other mesodermal and endodermal lineages, under appropriate conditions: adipogenic, osteo/dentinogenic, chondrogenic, neurogenic, endothelial, myofibroblastic [237] and hepatocytes [238]. DPSCs can be isolated from both the perivascular and the sub-odontoblastic compartments (the inner surface of the tooth and the outer part of the pulp tissue), by separate digestion of the tooth and extracted pulp tissue. Both populations presented identical cell size, doubling times and karyotype stability, differing only in morphology with rounder cells in the sub-odontoblastic compartment versus spindle-shaped cells with long processes in the perivascular one [239]. Pre-clinical experiments have demonstrated that stem cells show promising results in differentiation into neuronal-like cells [226, 228, 232, 233, 240] and their secretion of growth factors [229, 241, 242]. Shi and colleagues [243] stated the potential of glia-derived neurotrophic factor expressing neural stem cells in the regeneration of facial nerve gap in rats.

2.8.2.2. Neurotrophic factors

Growth factors and cytokines have a complex enhancing effect in tissue regeneration which can be exploited with great potential in nerve regeneration [73]. To date, several neurotrophic factors have been identified: transforming growth factor beta superfamily (TGF-β), nerve growth factor, neurotrophins 3, 4 and 5 (NT-3/4/5), ciliary neurotrophic factor (CNTF), neuregulin-1 (NRG1), brain-derived neurotrophic factor, glial cell line–derived neurotrophic factor (GDNF) and vascular endothelial growth factor (VEGF). The controlled release of the neurotrophic factors in nerve conduits may take several approaches, namely topical application, subcutaneous injection, microosmotic pump and diffusion or affinity-based polymer microspheres [244251]. In another approach, Cui and colleagues [252] loaded the neurotrophic factors into the wall of the nerve conduit with controlled diffusion into the lumen. NGF has been reported to promote neurite outgrowth in vitro in co-culture of neurons and astrocytes [253] and in vivo–enhanced axonal regeneration in sciatic nerve in rat model [248, 254]. Several researchers have demonstrated BDNF efficacy in the regeneration of rat motor nerves [253, 255, 256]. The dosage of BDNS has also been studied [257]. A high dosage, set as 12–20 mg/day, has been stated to interact with p75 receptors and therefore inhibits axonal regeneration. In fact, single exogenous dosage of BDNF has shown better results than continuous long-term applications [258]. It is reported that muscle regeneration causes the overexpression of GDNF which increases the number of motor axons in the neuromuscular junctions in vivo [259266]. Also, GDNF is a potent protective factor against axotomy-induced motor and sensory neuron death [267270]. CNTF has the ability to improve and regenerate muscle function after nerve injury in vivo [271, 272]. It has also demonstrated to enable peripheral nerve regeneration by promoting axon elongation and sprouting from axon distal stump [273]. Six main isoforms of NRG-1 are described. Due to their importance in nerve regeneration isoforms I, II and III are the most studied in the last few years [274]. Nerve-regenerative effect of NRG-1 is highly dependent on the isoform and its dosage [275, 276]. Several studies indicate that NRG-1 isoforms are capable of stimulating SCs proliferation a remyelinization [275278]. Gambarrota and colleagues [279] recommended that initial high dosages of NRGA-1 stimulate SCs differentiation.

2.8.2.3. Pharmacological agents

Until now, there is no pharmacological method that can effectively enhance nerve regeneration. However, as mentioned earlier, neurotrophic and growth factors have demonstrated potential in enhancing nerve repair and regeneration by reducing neuronal death and promoting axonal outgrowth. Recent advances in molecular biology have indicated that targeting specific steps in molecular pathways may allow for purposeful pharmacologic intervention, potentially leading to a better functional recovery after nerve injury [63, 280]. Major molecular pathways implicated in neuron survival and neurite outgrowth include PI3K (phosphatidylinositol-3 kinase)/Akt (protein kinase B)-signalling cascade, Ras-ERK (rat sarcoma-extracellular signal-regulated kinase) pathway, the cyclic adenosine monophosphate (cAMP)/protein kinase A (PKA) and Rho-ROK signalling [280]. PI3K/Akt cascade seems to provide trophic support for neurons, block apoptosis, facilitate signal transmission and mediate cell growth and differentiation in neurons. Also, it is reported that NGF is mediated by PI3K/Akt/mTOR pathway [281283]. Ras-ERK pathway is a key promoter of neurite outgrowth and also has been found to enhance axonal survival [284]. Rho–ROK pathway participates in neural growth and modulates neurite outgrowth [285]. Nectins and nectin-like molecules are cell-cell adhesion molecules that participate in cell communication. Nectin-like molecule 1 (NECL1) is restricted to the nervous system and is responsible for the synapses formation, axon bundles, myelinated axons and cerebellar morphogenesis [286, 287]. In a recent study, Xu and colleagues [288] developed a PLGA scaffold coated with NECL1 to enhance adhesion of rat SCs and applied in the treatment of transected sciatic nerve in rat. Results also revealed that the final outcome of both motor and sensory regeneration and reinnervation. Other molecules with different clinical applications have, however, demonstrated beneficial effect in nerve regeneration process: erythropoietin (EPO) [289], tacrolimus (FK506) [290], acetyl-L-carnitine (ALCAR) [291], N-acetylcysteine (NAC) [292], ibuprofen [293], melatonin [294] and transthyretin [295].

2.8.2.4. Channels

The use of longitudinal channels inside nerve conduits is the usual strategy to promote axon guidance towards distal stump. The artificial micro-tubular structure mimics the endoneural tubes and fascicles of a peripheral nerve anatomy and therefore enhances neuroregeneration [296298].

2.8.2.5. Hydrogels

As described earlier, conduits lumen can be filled with ECM components such as collagen and laminin which are involved in the process of regeneration by forming a substrate for neuron cell migration. Laminin-filled silicone conduits have demonstrated enhanced nerve regeneration [299, 300]. Collagen has also demonstrated to increase nerve regeneration [301]. BD Matrigel® and other lamini and collagen gels have been widely used as conduit fillings to incorporate or support cells and neurotrophic factors [302304].

2.8.2.6. Conductive conduits

Electrical stimulation as a therapeutic in nerve injuries has been widely discussed in the academic community [305313]. External electrical stimulation as peripheral nerve regeneration strategy has been demonstrated. Several conductive polymers such as polyaniline (PANI) and polypyrrole have been described with great potential for nerve regeneration due to their biocompatibility, tuneable conductivity, environmental stability and facility to produce. The great advantage of this material is the ability to continue to transmit the electrical signal in impaired nerves and physically stimulate cell growth and regeneration [314316].

Advertisement

2.9. Future perspectives

Gene therapy involves the introduction of a foreign gene into living cells with the intention to overcome a disease [317]. The most efficient way to deliver transgenes into cells is through a vector, such as herpes simplex, adenovirus, lentivirus and adeno-associated viral vectors [318, 319]. Gene can reprogramme cells to produce neurotrophic factors, cell adhesion or extracellular matrix molecules, and transcription factors. Therefore, in peripheral nerve injury, Schwann cells, fibroblasts and denervated muscle are potential targets for this breakthrough approach [63].

Bioactive glasses have been widely used as bone-filling materials and dental implants and have demonstrated its potential in soft-tissue regeneration. A comprehensive study on the application of bioactive glass in peripheral nerve regeneration has been conducted by Novajra and colleagues [320, 321]. Polymer-glass composite devices are made with bioactive glass powder and successfully applied in peripheral nerve repair. In other approach, this material can be used as fibres to produce nerve wraps, topographic patterns at conduit lumen or guidance channel to guide axonal growth. Their major advantage is the ions release from bioactive glasses, which have demonstrated angiogenic effects and the possibility of manipulation of glass composition; I order to include antibacterial ions, such as silver, gallium, zinc and copper, that can be useful in the prevention and treatment of infections resulting from the clinical intervention.

In conclusion, the vast field for improvement in peripheral nerve regeneration strategies has been well recognized. The ideal therapeutic alternative should be readily available, able to confine and direct axonal growth and be biofunctional, by the supplementation with bioactive molecules and/or cellular systems. Therefore, there is scope for improvement in the development of new and better alternatives of bioactive peripheral nerve repair complexes.

Advertisement

Acknowledgments

This research was supported by Programa Operacional Regional do Norte (ON.2—O Novo Norte), QREN, FEDER with the project ‘iBone Therapies: Terapias inovadoras para a regeneração óssea’, ref. NORTE-01-0247-FEDER-003262, and by the programme COMPETE—Programa Operacional Factores de Competitividade, Projects PEst-OE/AGR/UI0211/2011 and PEst-C/EME/UI0285/2013 funding from FCT. This research was also supported by Programa Operacional Competitividade e Internacionalização (P2020), Fundos Europeus Estruturais e de Investimento (FEEI) and FCT with the project ‘BioMate—A novel bio-manufacturing system to produce bioactive scaffolds for tissue engineering’ with reference PTDC/EMS-SIS/7032/2014 and by COMPETE 2020, from ANI—Projectos ID&T Empresas em Copromoção, Programas Operacionais POCI, by the project ‘insitu.Biomas—Reinvent biomanufacturing systems by using an usability approach for in situ clinic temporary implants fabrication’ with the reference POCI-01-0247-FEDER-017771. Ana Rita Caseiro (SFRH/BD/101174/2014) acknowledges FCT for financial support.

Advertisement

Conflicts of interest

The authors confirm that they have no conflicts of interest to declare for this publication.

Advertisement

Abbreviations

AktProtein kinase B
BDNFBrain-derived neurotrophic factor
BMSCsBone marrow mesenchymal stem cells
cAMPCyclic adenosine monophosphate
CNTFCiliary neurotrophic factor
DTIDiffusion tensor imaging
DPSCsDental pulp stem cells
ECMExtracellular matrix
EMGElectromyograms
ePTFEPolytetrafluoroethylene
FDAFood and Drug Administration
FFMTFree-functioning muscle transfer
GDNFGlial cell line-derived neurotrophic factor
MHCMajor histocompatibility complex
MSCsMesenchymal stem cells
NACN-acetylcysteine
NCSNerve conduction studies
NGFNerve growth factor
NRG1Neuregulin-1
NT-3/4/5Neurotrophins 3, 4 and 5
PANIPolyaniline
PCLPoly d, l lactide-co-epsilon-caprolactone
PGAPolyglycolic acid
PHBPolyhydroxybutyrate
PI3KPhosphatidylinositol-3 kinase
PKAProtein kinase A
POSSPolyhedral oligomeric silsesquioxane
PPyPolypyrrole
PVAPolyvinyl alcohol
PVA-CNTsPVA-functionalized carbon nanotubes
Ras-ERKRat sarcoma-extracellular signal-regulated kinase
RGDArginylglycylaspartic acid
SCSchwann cells
SFISciatic Functional Index
SISSmall intestine submucosa
SSIStatic Sciatic Index
TGF-βTransforming growth factor beta superfamily
VEGFVascular endothelial growth factor
WJWharton jelly
WRLWithdrawal reflex latency

References

  1. 1. Skalak R, Fox CF. Tissue Engineering: Proceedings of a Workshop, Granlibakken, Lake Tahoe, CA; February 26-29, 1988; Liss; 1988, ISBN: 0845147064, 9780845147061
  2. 2. Langer R, Vacanti J. Tissue engineering. Science. 1993;260(5110):920-926
  3. 3. Bianco P, Robey PG. Stem cells in tissue engineering. Nature. 2001;414(6859):118-121
  4. 4. Tuan RS, Boland G, Tuli R. Adult mesenchymal stem cells and cell-based tissue engineering. Arthritis Research & Therapy. 2003;5(1):32-45
  5. 5. Nerem RM, Sambanis A. Tissue engineering: From biology to biological substitutes. Tissue Engineering. 1995;1(1):3-13
  6. 6. Muschler GF, Nakamoto C, Griffith LG. Engineering principles of clinical cell-based tissue engineering. The Journal of Bone and Joint Surgery America. 2004;86-A(7):1541-1558
  7. 7. Zavaglia CAC, Prado da Silva MH. Feature Article: Biomaterials. Reference Module in Materials Science and Materials Engineering. Elsevier; 2016, DOI: 10.1016/B978-0-12-803581-8.04109-6
  8. 8. Graziano A, d’Aquino R, Laino G, Papaccio G. Dental pulp stem cells: A promising tool for bone regeneration. Stem Cell Reviews. 2008;4(1):21-26
  9. 9. Demarco FF, Conde MC, Cavalcanti BN, Casagrande L, Sakai VT, Nor JE. Dental pulp tissue engineering. Brazilian Dental Journal. 2011;22(1):3-13
  10. 10. Lee K, Silva EA, Mooney DJ. Growth factor delivery-based tissue engineering: General approaches and a review of recent developments. Journal of the Royal Society, Interface. 2011;8(55):153-170
  11. 11. Henkel J, Hutmacher Dietmar W. Design and fabrication of scaffold-based tissue engineering. BioNanoMaterials. 2013;14(3-4):171-193, DOI:10.1515/bnm-2013-0021
  12. 12. Potdar PD, Jethmalani YD. Human dental pulp stem cells: Applications in future regenerative medicine. World Journal of Stem Cells. 2015;7(5):839-851
  13. 13. Barnes CP, Sell SA, Boland ED, Simpson DG, Bowlin GL. Nanofiber technology: Designing the next generation of tissue engineering scaffolds. Advanced Drug Delivery Reviews. 2007;59(14):1413-1433
  14. 14. Williams DF. The Williams Dictionary of Biomaterials: Liverpool University Press; 1999
  15. 15. Yannas IV. Emerging rules for inducing organ regeneration. Biomaterials. 2013;34(2):321-330
  16. 16. Ponche A, Bigerelle M, Anselme K. Relative influence of surface topography and surface chemistry on cell response to bone implant materials. Part 1: Physico-chemical effects. Proceedings of the Institution of Mechanical Engineers, Part H. 2010;224(12):1471-1486
  17. 17. Muheremu A, Ao Q. Past, present, and future of nerve conduits in the treatment of peripheral nerve injury. Biomedical Research International. 2015;2015:237507
  18. 18. Katirji B, Kaminski HJ, Ruff RL. Neuromuscular Disorders in Clinical Practice. Springer Science & Business Media. 2013, ISBN: 978-1-4614-6567-6
  19. 19. Isaacs J. Major peripheral nerve injuries. Hand Clinics. 2013;29(3):371-382
  20. 20. Palispis WA, Gupta R. Surgical repair in humans after traumatic nerve injury provides limited functional neural regeneration in adults. Experimental Neurology. 2017;290:106-114
  21. 21. Aegineta P. De re medica. Paulou Aiginetou iatrou aristoubiblia hepta. En arche hekastou ton biblion deiknytai ta en ekeino pereichomena. Pavli Aeginetae medici optimi. Libri septem. 1528
  22. 22. Millesi H. Microsurgery of peripheral nerves. The Hand. 1973;5(2):157-160
  23. 23. Kurze T. Microtechniques in neurological surgery. Clinical Neurosurgery. 1964;11:128-137
  24. 24. Smith JW. Microsurgery of peripheral nerves. Plastic and Reconstructive Surgery. 1964;33:317-329
  25. 25. Sunderland SS, Williams HB. Nerve injuries and their repair: A critical appraisal. Plastic and Reconstructive Surgery. 1992;89(6):1170
  26. 26. Rochkind S, Nevo Z. Recovery of peripheral nerve with massive loss defect by tissue engineered guiding regenerative gel. Biomedical Research International. 2014;2014:327578
  27. 27. He B, Zhu Z, Zhu Q, Zhou X, Zheng C, Li P, et al. Factors predicting sensory and motor recovery after the repair of upper limb peripheral nerve injuries. Neural Regeneration Research. 2014;9(6):661-672
  28. 28. He C-q, Zhang L-h, Liu X-f, Tang P-f. A 2-year follow-up survey of 523 cases with peripheral nerve injuries caused by the earthquake in Wenchuan, China. Neural Regeneration Research. 2015;10(2):252-259
  29. 29. Ma CH, Omura T, Cobos EJ, Latr, xE, moli, et al. Accelerating axonal growth promotes motor recovery after peripheral nerve injury in mice. The Journal of Clinical Investigation. 2011;121(11):4332-4347, DOI: 10.1172/JCI58675
  30. 30. Grinsell D, Keating CP. Peripheral nerve reconstruction after injury: A review of clinical and experimental therapies. Biomedical Research International. 2014;2014:698256
  31. 31. Yegiyants S, Dayicioglu D, Kardashian G, Panthaki ZJ. Traumatic peripheral nerve injury: A wartime review. Journal of Craniofacial Surgery. 2010;21(4):998-1001
  32. 32. Flores AJ, Lavernia CJ, Owens PW. Anatomy and physiology of peripheral nerve injury and repair. The American Journal of Orthopedics (Belle Mead, NJ). 2000;29(3):167-173
  33. 33. Menorca RM, Fussell TS, Elfar JC. Nerve physiology: Mechanisms of injury and recovery. Hand Clinics. 2013;29(3):317-330
  34. 34. Pfister BJ, Gordon T, Loverde JR, Kochar AS, Mackinnon SE, Cullen DK. Biomedical engineering strategies for peripheral nerve repair: Surgical applications, state of the art, and future challenges. Critical Reviews in Biomedical Engineering. 2011;39(2):81-124
  35. 35. Schwartz LM. Atrophy and programmed cell death of skeletal muscle. Cell Death and Differentiation. 2008;15(7):1163-1169
  36. 36. Deumens R, Bozkurt A, Meek MF, Marcus MA, Joosten EA, Weis J, et al. Repairing injured peripheral nerves: Bridging the gap. Progress in Neurobiology. 2010;92(3):245-276
  37. 37. Griffin JW, Hogan MV, Chhabra AB, Deal DN. Peripheral nerve repair and reconstruction. The Journal of Bone and Joint Surgery America. 2013;95(23):2144-2151
  38. 38. Fu SY, Gordon T. The cellular and molecular basis of peripheral nerve regeneration. Molecular Neurobiology. 1997;14(1-2):67-116
  39. 39. Lee SK, Wolfe SW. Peripheral nerve injury and repair. Journal of the American Academy of Orthopaedic Surgeons. 2000;8(4):243-252
  40. 40. Siemionow M, Brzezicki G. Chapter 8: Current techniques and concepts in peripheral nerve repair. International Review of Neurobiology. 2009;87:141-172
  41. 41. Seddon HJ. Three types of nerve injury. Brain. 1943;66(4):237-288
  42. 42. Chhabra A, Ahlawat S, Belzberg A, Andreseik G. Peripheral nerve injury grading simplified on MR neurography: As referenced to Seddon and Sunderland classifications. The Indian Journal of Radiology & Imaging. 2014;24(3):217-224
  43. 43. Sunderland S. A classification of peripheral nerve injuries producing loss of function. Brain. 1951;74(4):491-516
  44. 44. Mackinnon SE, Dellon AL. Surgery of the Peripheral Nerve. Thieme Medical Publishers; 1988, ISBN:1588905136
  45. 45. Glaus SW, Johnson PJ, Mackinnon SE. Clinical strategies to enhance nerve regeneration in composite tissue allotransplantation. Hand Clinics. 2011;27(4):495-509, ix
  46. 46. Leoni AS, Mazzer N, Guirro RR, Jatte FG, Chereguini PA, Monte-Raso VV. High voltage pulsed current stimulation of the sciatic nerve in rats: Analysis by the SFI. Acta Ortopedica Brasiliera. 2012;20(2):93-97
  47. 47. Merle M. SFI = sciatic functional index? Or some feebler imitation? Microsurgery. 1995;16(3):171-172
  48. 48. Bervar M. Video analysis of standing—an alternative footprint analysis to assess functional loss following injury to the rat sciatic nerve. Journal of Neuroscience Methods. 2000;102(2):109-116
  49. 49. Bervar M. An alternative video footprint analysis to assess functional loss following injury to the rat sciatic nerve. Acta Chirurgiae Plasticae. 2002;44(3):86-89
  50. 50. Amado S, Armada-da-Silva PAS, João F, Maurício AC, Luís AL, Simões MJ, et al. The sensitivity of two-dimensional hindlimb joint kinematics analysis in assessing functional recovery in rats after sciatic nerve crush. Behavioural Brain Research. 2011;225(2):562-573
  51. 51. Amado S, Rodrigues JM, Luis AL, Armada-da-Silva PA, Vieira M, Gartner A, et al. Effects of collagen membranes enriched with in vitro-differentiated N1E-115 cells on rat sciatic nerve regeneration after end-to-end repair. Journal of Neuroengineering and Rehabilitation. 2010;7:7
  52. 52. Amado S, Simões MJ, Armada da Silva PAS, Luís AL, Shirosaki Y, Lopes MA, et al. Use of hybrid chitosan membranes and N1E-115 cells for promoting nerve regeneration in an axonotmesis rat model. Biomaterials. 2008;29(33):4409-4419
  53. 53. Gärtner A, Pereira T, Armada-da-Silva PAS, Amorim I, Gomes R, Ribeiro J, et al. Use of poly(DL-lactide-ε-caprolactone) membranes and mesenchymal stem cells from the Wharton’s jelly of the umbilical cord for promoting nerve regeneration in axonotmesis: In vitro and in vivo analysis. Differentiation. 2012;84(5):355-365
  54. 54. Thalhammer JG, Vladimirova M, Bershadsky B, Strichartz GR. Neurologic evaluation of the rat during sciatic nerve block with lidocaine. Anesthesiology. 1995;82(4):1013-1025
  55. 55. Masters DB, Berde CB, Dutta SK, Griggs CT, Hu D, Kupsky W, et al. Prolonged regional nerve blockade by controlled release of local anesthetic from a biodegradable polymer matrix. Anesthesiology. 1993;79(2):340-346
  56. 56. Caseiro AR, Pereira T, Ribeiro J, Amorim I, Faria F, Bártolo PJ, et al. Neuro-muscular regeneration using scaffolds with mesenchymal stem cells (MSCs) isolated from human umbilical cord Wharton’s jelly: Functional and morphological analysis using rat sciatic nerve neurotmesis injury model. Procedia Engineering. 2015;110:106-113
  57. 57. Costa LM, Simões MJ, Maurício AC, Varejão ASP. Chapter 7 Methods and Protocols in Peripheral Nerve Regeneration Experimental Research: Part IV—Kinematic Gait Analysis to Quantify Peripheral Nerve Regeneration in the Rat. International Review of Neurobiology. Academic Press; 2009;87:127-139. DOI: 10.1016/S0074-7742(09)87007-4.
  58. 58. Gärtner A, Pereira T, Alves MG, Armada-da-Silva PAS, Amorim I, Gomes R, et al. Corrigendum to “use of poly(dl-lactide-ε-caprolactone) membranes and mesenchymal stem cells from the Wharton’s jelly of the umbilical cord for promoting nerve regeneration in axonotmesis: In vitro and in vivo analysis” [Differentiation 84 (2012) 355-365]. Differentiation. 2013;85(3):119
  59. 59. Luís AL, Amado S, Geuna S, Rodrigues JM, Simões MJ, Santos JD, et al. Long-term functional and morphological assessment of a standardized rat sciatic nerve crush injury with a non-serrated clamp. Journal of Neuroscience Methods. 2007;163(1):92-104
  60. 60. Luis AL, Rodrigues JM, Amado S, Veloso AP, Armada-Da-Silva PA, Raimondo S, et al. PLGA 90/10 and caprolactone biodegradable nerve guides for the reconstruction of the rat sciatic nerve. Microsurgery. 2007;27(2):125-137
  61. 61. Ribeiro J, Gartner A, Pereira T, Gomes R, Lopes MA, Gonçalves C, et al. Chapter four— Perspectives of employing mesenchymal stem cells from the Wharton’s jelly of the umbilical cord for peripheral nerve repair. In: Stefano Geuna IPPT, Bruno B, editors. International Review of Neurobiology. Academic Press; 2013;108:79-120. DOI: 10.1016/B978-0-12-410499-0.00004-6
  62. 62. Trehan SK, Model Z, Lee SK. Nerve repair and nerve grafting. Hand Clinics. 2016;32(2):119-125
  63. 63. Panagopoulos GN, Megaloikonomos PD, Mavrogenis AF. The present and future for peripheral nerve regeneration. Orthopedics. 2016;40(1):e141-e156, DOI: 10.3928/01477447-20161019-01
  64. 64. Gerth DJ, Tashiro J, Thaller SR. Clinical outcomes for conduits and scaffolds in peripheral nerve repair. World Journal of Clinical Cases. 2015;3(2):141-147
  65. 65. Konofaos P, Ver Halen JP. Nerve repair by means of tubulization: Past, present, future. Journal of Reconstructive Microsurgery. 2013;29(3):149-164
  66. 66. Lundborg G. A 25-year perspective of peripheral nerve surgery: Evolving neuroscientific concepts and clinical significance. Journal of Hand Surgery America. 2000;25(3):391-414
  67. 67. Isaacs J. Treatment of acute peripheral nerve injuries: Current concepts. Journal of Hand Surgery America. 2010;35(3):491-497; quiz 8
  68. 68. Isaacs JE, McDaniel CO, Owen JR, Wayne JS. Comparative analysis of biomechanical performance of available “nerve glues”. Journal of Hand Surgery America. 2008;33(6):893-899
  69. 69. Daroff RB, Jankovic J, Mazziotta JC, Pomeroy SL. Bradley’s Neurology in Clinical Practice. Elsevier Health Sciences; 2015, ISBN: 9780323287838
  70. 70. Colen KL, Choi M, Chiu DT. Nerve grafts and conduits. Plastic and Reconstructive Surgery. 2009;124(6 Suppl):e386–e394
  71. 71. Terzis JK, Kostopoulos VK. Vascularized nerve grafts and vascularized fascia for upper extremity nerve reconstruction. Hand (New York). 2010;5(1):19-30
  72. 72. Choi D, Raisman G. Immune rejection of a facial nerve xenograft does not prevent regeneration and the return of function: An experimental study. Neuroscience. 2003;121(2):501-507
  73. 73. Gaudin R, Knipfer C, Henningsen A, Smeets R, Heiland M, Hadlock T. Approaches to peripheral nerve repair: Generations of biomaterial conduits yielding to replacing autologous nerve grafts in craniomaxillofacial surgery. Biomedical Research International. 2016;2016:3856262
  74. 74. Ray WZ, Mackinnon SE. Management of nerve gaps: Autografts, allografts, nerve transfers, and end-to-side neurorrhaphy. Experimental Neurology. 2010;223(1):77-85
  75. 75. Millesi H, Meissl G, Berger A. The interfascicular nerve-grafting of the median and ulnar nerves. The Journal of Bone and Joint Surgery America. 1972;54(4):727-750
  76. 76. Taylor GI, Ham FJ. The free vascularized nerve graft. A further experimental and clinical application of microvascular techniques. Plastic and Reconstructive Surgery. 1976;57(4):413-426
  77. 77. Moore AM, Ray WZ, Chenard KE, Tung T, Mackinnon SE. Nerve allotransplantation as it pertains to composite tissue transplantation. Hand (New York). 2009;4(3):239-244
  78. 78. Safa B, Buncke G. Autograft substitutes: Conduits and processed nerve allografts. Hand Clinics. 2016;32(2):127-140
  79. 79. Karabekmez FE, Duymaz A, Moran SL. Early clinical outcomes with the use of decellularized nerve allograft for repair of sensory defects within the hand. Hand (New York). 2009;4(3):245-249
  80. 80. Lee SK, Wolfe SW. Nerve transfers for the upper extremity: New horizons in nerve reconstruction. Journal of the American Academy of Orthopedic Surgeons. 2012;20(8):506-517
  81. 81. Harris RI. The treatment of irreparable nerve injuries. Canadian Medical Association Journal. 1921;11(11):833-841
  82. 82. Carlsen BT, Bishop AT, Shin AY. Late reconstruction for brachial plexus injury. Neurosurgery Clinics of North America. 2009;20(1):51-64, vi
  83. 83. Kehoe S, Zhang XF, Boyd D. FDA approved guidance conduits and wraps for peripheral nerve injury: A review of materials and efficacy. Injury. 2012;43(5):553-572
  84. 84. Lin MY, Manzano G, Gupta R. Nerve allografts and conduits in peripheral nerve repair. Hand Clinics. 2013;29(3):331-348
  85. 85. FF IJpma, Van De Graaf RC, Meek MF. The early history of tubulation in nerve repair. Journal of Hand Surgery European Volume. 2008;33(5):581-586, DOI: 10.1177/1753193408091349
  86. 86. Dahlin LB, Lundborg G. Use of tubes in peripheral nerve repair. Neurosurgery Clinics of North America. 2001;12(2):341-352
  87. 87. Isaacs J, Mallu S, Yan W, Little B. Consequences of oversizing: Nerve-to-nerve tube diameter mismatch. Journal of Bone and Joint Surgery America. 2014;96(17):1461-1467
  88. 88. Moore AM, Kasukurthi R, Magill CK, Farhadi HF, Borschel GH, Mackinnon SE. Limitations of conduits in peripheral nerve repairs. Hand (New York). 2009;4(2):180-186
  89. 89. Nectow AR, Marra KG, Kaplan DL. Biomaterials for the development of peripheral nerve guidance conduits. Tissue Engineering Part B Reviews. 2012;18(1):40-50
  90. 90. Meek MF, Coert JH. US Food and Drug Administration/Conformit Europe-approved absorbable nerve conduits for clinical repair of peripheral and cranial nerves. Annals in Plastic Surgery. 2008;60(1):110-116
  91. 91. Brunelli GA, Vigasio A, Brunelli GR. Different conduits in peripheral nerve surgery. Microsurgery. 1994;15(3):176-178
  92. 92. Ichihara S, Inada Y, Nakamura T. Artificial nerve tubes and their application for repair of peripheral nerve injury: An update of current concepts. Injury. 2008;39(Suppl 4):29-39
  93. 93. Arslantunali D, Dursun T, Yucel D, Hasirci N, Hasirci V. Peripheral nerve conduits: Technology update. Medical Devices (Auckland). 2014;7:405-424
  94. 94. Anderson PN, Turmaine M. Axonal regeneration through arterial grafts. Journal of Anatomy. 1986;147:73-82
  95. 95. Kosutic D, Krajnc I, Pejkovic B, Solman L. Autogenous digital artery graft for repair of digital nerve defects in emergency hand reconstruction: Two-year follow-up. Journal of Plastic, Reconstructive & Aesthetic Surgery. 2009;62(4):553
  96. 96. Frerichs O, Fansa H, Schicht C, Wolf G, Schneider W, Keilhoff G. Reconstruction of peripheral nerves using acellular nerve grafts with implanted cultured Schwann cells. Microsurgery. 2002;22(7):311-315
  97. 97. Kim BS, Yoo JJ, Atala A. Peripheral nerve regeneration using acellular nerve grafts. Journal of Biomedical Material Research A. 2004;68(2):201-209
  98. 98. Smith RM, Wiedl C, Chubb P, Greene CH. Role of small intestine submucosa (SIS) as a nerve conduit: Preliminary report. Journal of Investigative Surgery. 2004;17(6):339-344
  99. 99. Yi JS, Lee HJ, Lee HJ, Lee IW, Yang JH. Rat peripheral nerve regeneration using nerve guidance channel by porcine small intestinal submucosa. Journal of Korean Neurosurgery Society. 2013;53(2):65-71
  100. 100. Carriel V, Alaminos M, Garzon I, Campos A, Cornelissen M. Tissue engineering of the peripheral nervous system. Expert Review of Neurotherapeutics. 2014;14(3):301-318
  101. 101. Quigley AF, Bulluss KJ, Kyratzis IL, Gilmore K, Mysore T, Schirmer KS, et al. Engineering a multimodal nerve conduit for repair of injured peripheral nerve. Journal of Neural Engineering. 2013;10(1):016008
  102. 102. Stanec S, Stanec Z. Reconstruction of upper-extremity peripheral-nerve injuries with ePTFE conduits. Journal of Reconstructive Microsurgery. 1998;14(4):227-232
  103. 103. Zhang BG, Quigley AF, Myers DE, Wallace GG, Kapsa RM, Choong PF. Recent advances in nerve tissue engineering. International Journal of Artificial Organs. 2014;37(4):277-291
  104. 104. Kadler KE, Holmes DF, Trotter JA, Chapman JA. Collagen fibril formation. Biochemistry Journal. 1996;316 (Pt 1):1-11
  105. 105. Stang F, Fansa H, Wolf G, Reppin M, Keilhoff G. Structural parameters of collagen nerve grafts influence peripheral nerve regeneration. Biomaterials. 2005;26(16):3083-3091
  106. 106. Stang F, Fansa H, Wolf G, Keilhoff G. Collagen nerve conduits--assessment of biocompatibility and axonal regeneration. Bio-Medical Materials and Engineering. 2005;15(1-2):3-12
  107. 107. Sulong AF, Hassan NH, Hwei NM, Lokanathan Y, Naicker AS, Abdullah S, et al. Collagen-coated polylactic-glycolic acid (PLGA) seeded with neural-differentiated human mesenchymal stem cells as a potential nerve conduit. Advances in Clinical and Experimental Medicine. 2014;23(3):353-362
  108. 108. Bushnell BD, McWilliams AD, Whitener GB, Messer TM. Early clinical experience with collagen nerve tubes in digital nerve repair. Journal of Hand Surgery America. 2008;33(7):1081-1087
  109. 109. Wangensteen KJ, Kalliainen LK. Collagen tube conduits in peripheral nerve repair: A retrospective analysis. Hand (New York). 2010;5(3):273-277
  110. 110. Lohmeyer JA, Sommer B, Siemers F, Mailander P. Nerve injuries of the upper extremity-expected outcome and clinical examination. Plastic Surgical Nursing. 2009;29(2):88-93; quiz 4-5
  111. 111. Taras JS, Jacoby SM, Lincoski CJ. Reconstruction of digital nerves with collagen conduits. Journal of Hand Surgery America. 2011;36(9):1441-1446
  112. 112. Ashley WW, Jr., Weatherly T, Park TS. Collagen nerve guides for surgical repair of brachial plexus birth injury. Journal of Neurosurgery. 2006;105(6 Suppl):452-456
  113. 113. Archibald SJ, Shefner J, Krarup C, Madison RD. Monkey median nerve repaired by nerve graft or collagen nerve guide tube. Journal of Neuroscience. 1995;15(5 Pt 2):4109-4123
  114. 114. Bekler HI, Rosenwasser MP, Akilina Y, Bulut G. The use of an absorbable collagen cover (NeuraWrap) improves patency of interpositional vein grafts. Acta Orthopaedica et Traumatologica Turcica. 2010;44(2):157-161
  115. 115. Kitahara AK, Suzuki Y, Qi P, Nishimura Y, Suzuki K, Kiyotani T, et al. Facial nerve repair using a collagen conduit in cats. Scandinavian Journal of Plastic and Reconstructive Surgery, Hand Surgery. 1999;33(2):187-193
  116. 116. Dalamagkas K, Tsintou M, Seifalian A. Advances in peripheral nervous system regenerative therapeutic strategies: A biomaterials approach. Material Science and Engineering C: Materials for Biological Applications. 2016;65:425-432
  117. 117. Sedaghati T, Jell G, Seifalian A. Investigation of Schwann cell behaviour on RGD-functionalised bioabsorbable nanocomposite for peripheral nerve regeneration. Nature Biotechnology. 2014;31(3):203-213
  118. 118. Monaco G, Cholas R, Salvatore L, Madaghiele M, Sannino A. Sterilization of collagen scaffolds designed for peripheral nerve regeneration: Effect on microstructure, degradation and cellular colonization. Materials Science and Engineering: C. 2017;71:335-344
  119. 119. Hall H. Modified fibrin hydrogel matrices: Both, 3D-scaffolds and local and controlled release systems to stimulate angiogenesis. Current Pharmaceutical Design. 2007;13(35):3597-3607
  120. 120. Sameem M, Wood TJ, Bain JR. A systematic review on the use of fibrin glue for peripheral nerve repair. Plastic and Reconstructive Surgery. 2011;127(6):2381-2390
  121. 121. Kalbermatten DF, Pettersson J, Kingham PJ, Pierer G, Wiberg M, Terenghi G. New fibrin conduit for peripheral nerve repair. Journal of Reconstructive Microsurgery. 2009;25(1):27-33
  122. 122. Pettersson J, Kalbermatten D, McGrath A, Novikova LN. Biodegradable fibrin conduit promotes long-term regeneration after peripheral nerve injury in adult rats. Journal of Plastic, Reconstructive & Aesthetic Surgery. 2010;63(11):1893-1899
  123. 123. Rafijah G, Bowen AJ, Dolores C, Vitali R, Mozaffar T, Gupta R. The effects of adjuvant fibrin sealant on the surgical repair of segmental nerve defects in an animal model. Journal of Hand Surgery America. 2013;38(5):847-855
  124. 124. Huang W, Begum R, Barber T, Ibba V, Tee NC, Hussain M, et al. Regenerative potential of silk conduits in repair of peripheral nerve injury in adult rats. Biomaterials. 2012;33(1):59-71
  125. 125. Lu Y, Chi F, Zhao X, Shao Z, Cao Z. Experimental study on facial nerve regeneration by porous silk fibroin conduit. Zhonghua er bi yan hou tou jing wai ke za zhi= Chinese Journal of Otorhinolaryngology Head and Neck Surgery. 2006;41(8):603-606
  126. 126. Park SY, Ki CS, Park YH, Lee KG, Kang SW, Kweon HY, et al. Functional recovery guided by an electrospun silk fibroin conduit after sciatic nerve injury in rats. Journal of Tissue Engineering and Regenerative Medicine. 2015;9(1):66-76
  127. 127. Xie H, Yang W, Chen J, Zhang J, Lu X, Zhao X, et al. A silk sericin/silicone nerve guidance conduit promotes regeneration of a transected sciatic nerve. Advanced Healthcare Materials. 2015;4(15):2195-2205
  128. 128. Mottaghitalab F, Farokhi M, Zaminy A, Kokabi M, Soleimani M, Mirahmadi F, et al. A biosynthetic nerve guide conduit based on silk/SWNT/fibronectin nanocomposite for peripheral nerve regeneration. PLoS One. 2013;8(9):e74417
  129. 129. Yang Y, Ding F, Wu J, Hu W, Liu W, Liu J, et al. Development and evaluation of silk fibroin-based nerve grafts used for peripheral nerve regeneration. Biomaterials. 2007;28(36):5526-5535
  130. 130. Das S, Sharma M, Saharia D, Sarma KK, Sarma MG, Borthakur BB, et al. In vivo studies of silk based gold nano-composite conduits for functional peripheral nerve regeneration. Biomaterials. 2015;62:66-75
  131. 131. Cohen-Karni T, Jeong KJ, Tsui JH, Reznor G, Mustata M, Wanunu M, et al. Nanocomposite gold-silk nanofibers. Nano Letters. 2012;12(10):5403-5406
  132. 132. Rao J, Cheng Y, Liu Y, Ye Z, Zhan B, Quan D, et al. A multi-walled silk fibroin/silk sericin nerve conduit coated with poly(lactic-co-glycolic acid) sheath for peripheral nerve regeneration. Materials Science and Engineering: C. 2017;73:319-332
  133. 133. Dinis T, Elia R, Vidal G, Dermigny Q, Denoeud C, Kaplan D, et al. 3D multi-channel bi-functionalized silk electrospun conduits for peripheral nerve regeneration. Journal of the Mechanical Behavior of Biomedical Materials. 2015;41:43-55
  134. 134. Dinis TM, Vidal G, Jose RR, Vigneron P, Bresson D, Fitzpatrick V, et al. Complementary effects of two growth factors in multifunctionalized silk nanofibers for nerve reconstruction. PloS One. 2014;9(10):e109770
  135. 135. Lin Q, Cai Y, Li H. Experimental study on gradient of nerve growth factor immobilized conduits promoting peripheral nerve regeneration in rats. Zhongguo xiu fu chong jian wai ke za zhi= Zhongguo xiufu chongjian waike zazhi= Chinese Journal of Reparative and Reconstructive Surgery. 2014;28(2):167-172
  136. 136. Lin Y-C, Ramadan M, Hronik-Tupaj M, Kaplan DL, Philips BJ, Sivak W, et al. spatially controlled delivery of neurotrophic factors in silk fibroin–based nerve conduits for peripheral nerve repair. Annals of Plastic Surgery. 2011;67(2):147-155
  137. 137. Sivak WN, White JD, Bliley JM, Tien LW, Liao HT, Kaplan DL, et al. Delivery of chondroitinase ABC and glial cell line-derived neurotrophic factor from silk fibroin conduits enhances peripheral nerve regeneration. Journal of Tissue Engineering and Regenerative Medicine. 2014;11(3):733-742, DOI: 10.1002/term.1970
  138. 138. Zhang H, Wang K, Xing Y, Yu Q. Lysine-doped polypyrrole/spider silk protein/poly(l-lactic) acid containing nerve growth factor composite fibers for neural application. Materials Science and Engineering: C. 2015;56:564-573
  139. 139. Zheng L, Cui HF. Use of chitosan conduit combined with bone marrow mesenchymal stem cells for promoting peripheral nerve regeneration. The Journal of Material Science: Materials in Medicine. 2010;21(5):1713-1720
  140. 140. Gu J, Hu W, Deng A, Zhao Q, Lu S, Gu X. Surgical repair of a 30 mm long human median nerve defect in the distal forearm by implantation of a chitosan-PGA nerve guidance conduit. Journal of Tissue Engineering and Regenerative Medicine. 2012;6(2):163-168
  141. 141. Clavijo-Alvarez JA, Nguyen VT, Santiago LY, Doctor JS, Lee WP, Marra KG. Comparison of biodegradable conduits within aged rat sciatic nerve defects. Plastic and Reconstructive Surgery. 2007;119(6):1839-1851
  142. 142. Rampichova M, Kostakova E, Filova E, Prosecka E, Plencner M, Ocheretna L, et al. Non-woven PGA/PVA fibrous mesh as an appropriate scaffold for chondrocyte proliferation. Physiology Research. 2010;59(5):773-781
  143. 143. Schlosshauer B, Dreesmann L, Schaller HE, Sinis N. Synthetic nerve guide implants in humans: A comprehensive survey. Neurosurgery. 2006;59(4):740-747; discussion 7-8
  144. 144. Dellon AL, Mackinnon SE. An alternative to the classical nerve graft for the management of the short nerve gap. Plastic and Reconstructive Surgery. 1988;82(5):849-856
  145. 145. Weber RA, Breidenbach WC, Brown RE, Jabaley ME, Mass DP. A randomized prospective study of polyglycolic acid conduits for digital nerve reconstruction in humans. Plastic and Reconstructive Surgery. 2000;106(5):1036-1045; discussion 46-48
  146. 146. Battiston B, Geuna S, Ferrero M, Tos P. Nerve repair by means of tubulization: Literature review and personal clinical experience comparing biological and synthetic conduits for sensory nerve repair. Microsurgery. 2005;25(4):258-267
  147. 147. Rinker B, Liau JY. A Prospective randomized study comparing woven polyglycolic acid and autogenous vein conduits for reconstruction of digital nerve gaps. Journal of Hand Surgery. 2011;36(5):775-781
  148. 148. Crawley WA, Dellon AL. Inferior alveolar nerve reconstruction with a polyglycolic acid bioabsorbable nerve conduit. Plastic and Reconstructive Surgery. 1992;90(2):300-302
  149. 149. Dellon AL, Maloney CT, Jr. Salvage of sensation in a hallux-to-thumb transfer by nerve tube reconstruction. Journal of Hand Surgery America. 2006;31(9):1495-1498
  150. 150. Donoghoe N, Rosson GD, Dellon AL. Reconstruction of the human median nerve in the forearm with the Neurotube. Microsurgery. 2007;27(7):595-600
  151. 151. Rosson GD, Williams EH, Dellon AL. Motor nerve regeneration across a conduit. Microsurgery. 2009;29(2):107-114
  152. 152. Gu X, Ding F, Williams DF. Neural tissue engineering options for peripheral nerve regeneration. Biomaterials. 2014;35(24):6143-6156
  153. 153. Duda S, Dreyer L, Behrens P, Wienecke S, Chakradeo T, Glasmacher B, et al. Outer electrospun polycaprolactone shell induces massive foreign body reaction and impairs axonal regeneration through 3D multichannel chitosan nerve guides. Biomedical Research International. 2014;2014:835269
  154. 154. Bertleff MJ, Meek MF, Nicolai JP. A prospective clinical evaluation of biodegradable Neurolac nerve guides for sensory nerve repair in the hand. Journal of Hand Surgery America. 2005;30(3):513-518
  155. 155. Hernandez-Cortes P, Garrido J, Camara M, Ravassa FO. Failed digital nerve reconstruction by foreign body reaction to Neurolac nerve conduit. Microsurgery. 2010;30(5):414-416
  156. 156. Meek MF, Den Dunnen WF. Porosity of the wall of a Neurolac nerve conduit hampers nerve regeneration. Microsurgery. 2009;29(6):473-478
  157. 157. Den Dunnen W, Meek M. Sensory nerve function and auto-mutilation after reconstruction of various gap lengths with nerve guides and autologous nerve grafts. Biomaterials. 2001;22(10):1171-1176
  158. 158. Den Dunnen W, Meek M, Robinson P, Schakernraad J. Peripheral nerve regeneration through P (DLLA-ε-CL) nerve guides. Journal of Materials Science: Materials in Medicine. 1998;9(12):811-814
  159. 159. Den Dunnen W, Stokroos I, Blaauw E, Holwerda A, Pennings A, Robinson P, et al. Light‐microscopic and electron‐microscopic evaluation of short‐term nerve regeneration using a biodegradable poly (DL‐lactide‐ϵ‐caprolacton) nerve guide. Journal of Biomedical Materials Research Part A. 1996;31(1):105-115
  160. 160. Meek M, Den Dunnen W, Schakenraad J, Robinson P. Long‐term evaluation of functional nerve recovery after reconstruction with a thin‐walled biodegradable poly(dl‐lactide‐ε‐caprolactone) nerve guide, using walking track analysis and electrostimulation tests. Microsurgery. 1999;19(5):247-253
  161. 161. Meek MF. More than just sunshine with implantation of resorbable (p(DLLA-epsilon-CL)) biomaterials. Biomedical Materials and Engineering. 2007;17(5):329-334
  162. 162. Meek MF, Van Der Werff JF, Nicolai JPA, Gramsbergen A. Biodegradable p (DLLA‐ϵ‐CL) nerve guides versus autologous nerve grafts: Electromyographic and video analysis. Muscle & Nerve. 2001;24(6):753-759
  163. 163. Secer HI, Solmaz I, Anik I, Izci Y, Duz B, Daneyemez MK, et al. Surgical outcomes of the brachial plexus lesions caused by gunshot wounds in adults. Journal of Brachial Plexus and Peripheral Nerve Injury. 2009;4:11
  164. 164. Meek MF, den Dunnen WF, Robinson PH, Pennings AJ, Schakenraad JM. Evaluation of functional nerve recovery after reconstruction with a new biodegradable poly(dl-lactide-epsilon-caprolactone) nerve guide. International Journal of Artificial Organs. 1997;20(8):463-468
  165. 165. Meek MF, Dijkstra JR, Den Dunnen WF, Ijkema-Paassen J, Schakenraad JM, Gramsbergen A, et al. Functional assessment of sciatic nerve reconstruction: Biodegradable poly (DLLA-epsilon-CL) nerve guides versus autologous nerve grafts. Microsurgery. 1999;19(8):381-388
  166. 166. Meek MF, Robinson PH, Stokroos I, Blaauw EH, Kors G, den Dunnen WF. Electron microscopical evaluation of short-term nerve regeneration through a thin-walled biodegradable poly(DLLA-epsilon-CL) nerve guide filled with modified denatured muscle tissue. Biomaterials. 2001;22(10):1177-1185
  167. 167. Varejao AS, Cabrita AM, Geuna S, Patricio JA, Azevedo HR, Ferreira AJ, et al. Functional assessment of sciatic nerve recovery: Biodegradable poly (DLLA-epsilon-CL) nerve guide filled with fresh skeletal muscle. Microsurgery. 2003;23(4):346-353
  168. 168. Varejao AS, Cabrita AM, Meek MF, Fornaro M, Geuna S, Giacobini-Robecchi MG. Morphology of nerve fiber regeneration along a biodegradable poly (DLLA-epsilon-CL) nerve guide filled with fresh skeletal muscle. Microsurgery. 2003;23(4):338-345
  169. 169. Chen GQ, Wu Q. The application of polyhydroxyalkanoates as tissue engineering materials. Biomaterials. 2005;26(33):6565-6578
  170. 170. Xiao XQ, Zhao Y, Chen GQ. The effect of 3-hydroxybutyrate and its derivatives on the growth of glial cells. Biomaterials. 2007;28(25):3608-3616
  171. 171. Young RC, Wiberg M, Terenghi G. Poly-3-hydroxybutyrate (PHB): A resorbable conduit for long-gap repair in peripheral nerves. British Journal of Plastic Surgery. 2002;55(3):235-240
  172. 172. Aberg M, Ljungberg C, Edin E, Millqvist H, Nordh E, Theorin A, et al. Clinical evaluation of a resorbable wrap-around implant as an alternative to nerve repair: A prospective, assessor-blinded, randomised clinical study of sensory, motor and functional recovery after peripheral nerve repair. Journal of Plastic, Reconstructive & Aesthetic Surgery. 2009;62(11):1503-1509
  173. 173. Subramanian A, Krishnan UM, Sethuraman S. Development of biomaterial scaffold for nerve tissue engineering: Biomaterial mediated neural regeneration. Journal of Biomedical Science. 2009;16:108
  174. 174. Neal RA, Tholpady SS, Foley PL, Swami N, Ogle RC, Botchwey EA. Alignment and composition of laminin-polycaprolactone nanofiber blends enhance peripheral nerve regeneration. Journal of Biomedical Materials Research Part A. 2012;100(2):406-423
  175. 175. Armstrong SJ, Wiberg M, Terenghi G, Kingham PJ. ECM molecules mediate both Schwann cell proliferation and activation to enhance neurite outgrowth. Tissue Engineering. 2007;13(12):2863-2870
  176. 176. Patel S, Kurpinski K, Quigley R, Gao H, Hsiao BS, Poo MM, et al. Bioactive nanofibers: Synergistic effects of nanotopography and chemical signaling on cell guidance. Nano Letters. 2007;7(7):2122-2128
  177. 177. Ahmed MR, Jayakumar R. Peripheral nerve regeneration in RGD peptide incorporated collagen tubes. Brain Research. 2003;993(1):208-216
  178. 178. Hersel U, Dahmen C, Kessler H. RGD modified polymers: Biomaterials for stimulated cell adhesion and beyond. Biomaterials. 2003;24(24):4385-4415
  179. 179. Ni HC, Lin ZY, Hsu SH, Chiu IM. The use of air plasma in surface modification of peripheral nerve conduits. Acta Biomaterialia. 2010;6(6):2066-2076
  180. 180. Ahmed MR, Basha SH, Gopinath D, Muthusamy R, Jayakumar R. Initial upregulation of growth factors and inflammatory mediators during nerve regeneration in the presence of cell adhesive peptide‐incorporated collagen tubes. Journal of the Peripheral Nervous System. 2005;10(1):17-30
  181. 181. Pierschbacher MD, Ruoslahti E. Cell attachment activity of fibronectin can be duplicated by small synthetic fragments of the molecule. Nature. 1984;309(5963):30
  182. 182. Pierschbacher MD, Ruoslahti E. Variants of the cell recognition site of fibronectin that retain attachment-promoting activity. Proceedings of the National Academy of Sciences. 1984;81(19):5985-5988
  183. 183. Vleggeert-Lankamp CL, de Ruiter GC, Wolfs JF, Pego AP, van den Berg RJ, Feirabend HK, et al. Pores in synthetic nerve conduits are beneficial to regeneration. Journal of Biomedical Materials Research Part A. 2007;80(4):965-982
  184. 184. O’Brien FJ, Harley BA, Yannas IV, Gibson LJ. The effect of pore size on cell adhesion in collagen-GAG scaffolds. Biomaterials. 2005;26(4):433-441
  185. 185. Chew SY, Mi R, Hoke A, Leong KW. Aligned protein-polymer composite fibers enhance nerve regeneration: A potential tissue-engineering platform. Advanced Functional Materials. 2007;17(8):1288-1296
  186. 186. Panseri S, Cunha C, Lowery J, Del Carro U, Taraballi F, Amadio S, et al. Electrospun micro- and nanofiber tubes for functional nervous regeneration in sciatic nerve transections. BMC Biotechnology. 2008;8:39
  187. 187. Clements IP, Kim YT, English AW, Lu X, Chung A, Bellamkonda RV. Thin-film enhanced nerve guidance channels for peripheral nerve repair. Biomaterials. 2009;30(23-24):3834-3846
  188. 188. Ngo TT, Waggoner PJ, Romero AA, Nelson KD, Eberhart RC, Smith GM. Poly(L-Lactide) microfilaments enhance peripheral nerve regeneration across extended nerve lesions. Journal of Neuroscience Research. 2003;72(2):227-238
  189. 189. Araki T, Nagarajan R, Milbrandt J. Identification of genes induced in peripheral nerve after injury expression profiling and novel gene discovery. Journal of Biological Chemistry. 2001;276(36):34131-34141
  190. 190. Chang L-W, Viader A, Varghese N, Payton JE, Milbrandt J, Nagarajan R. An integrated approach to characterize transcription factor and microRNA regulatory networks involved in Schwann cell response to peripheral nerve injury. BMC Genomics. 2013;14(1):84
  191. 191. Evans GR, Brandt K, Katz S, Chauvin P, Otto L, Bogle M, et al. Bioactive poly (l-lactic acid) conduits seeded with Schwann cells for peripheral nerve regeneration. Biomaterials. 2002;23(3):841-848
  192. 192. Gottlieb DI. Large-scale sources of neural stem cells. Annual Review of Neuroscience. 2002;25(1):381-407
  193. 193. Han SS, Fischer I. Neural stem cells and gene therapy: Prospects for repairing the injured spinal cord. Journal of American Medical Association. 2000;283(17):2300-2301
  194. 194. Lobsiger CS, Taylor V, Suter U. The early life of a Schwann cell. Biological Chemistry. 2002;383(2):245-253
  195. 195. Morrison SJ, White PM, Zock C, Anderson DJ. Prospective identification, isolation by flow cytometry, and in vivo self-renewal of multipotent mammalian neural crest stem cells. Cell. 1999;96(5):737-749
  196. 196. Morrissey TK, Kleitman N, Bunge RP. Isolation and functional characterization of Schwann cells derived from adult peripheral nerve. Journal of Neuroscience. 1991;11(8):2433-2442
  197. 197. Park KI, Liu S, Flax JD, Nissim S, Stieg PE, Snyder EY. Transplantation of neural progenitor and stem cells: Developmental insights may suggest new therapies for spinal cord and other CNS dysfunction. Journal of Neurotrauma. 1999;16(8):675-687
  198. 198. Rao MS, Anderson DJ. Immortalization and controlled in vitro differentiation of murine multipotent neural crest stem cells. Journal of Neurobiology. 1997;32(7):722-746
  199. 199. Rappa G, Kunke D, Holter J, Diep D, Meyer J, Baum C, et al. Efficient expansion and gene transduction of mouse neural stem/progenitor cells on recombinant fibronectin. Neuroscience. 2004;124(4):823-830
  200. 200. Tohill M, Mantovani C, Wiberg M, Terenghi G. Rat bone marrow mesenchymal stem cells express glial markers and stimulate nerve regeneration. Neuroscience Letters. 2004;362(3):200-203
  201. 201. Viader A, Chang L-W, Fahrner T, Nagarajan R, Milbrandt J. MicroRNAs modulate Schwann cell response to nerve injury by reinforcing transcriptional silencing of dedifferentiation-related genes. Journal of Neuroscience. 2011;31(48):17358-17369
  202. 202. Chang CJ, Hsu SH. The effects of low-intensity ultrasound on peripheral nerve regeneration in poly(DL-lactic acid-co-glycolic acid) conduits seeded with Schwann cells. Ultrasound in Medicine & Biology. 2004;30(8):1079-1084
  203. 203. di Summa PG, Kingham PJ, Raffoul W, Wiberg M, Terenghi G, Kalbermatten DF. Adipose-derived stem cells enhance peripheral nerve regeneration. Journal of Plastic, Reconstructive & Aesthetic Surgery. 2010;63(9):1544-1552
  204. 204. Hadlock T, Sundback C, Hunter D, Cheney M, Vacanti JP. A polymer foam conduit seeded with Schwann cells promotes guided peripheral nerve regeneration. Tissue Engineering. 2000;6(2):119-127
  205. 205. Kalbermatten DF, Kingham PJ, Mahay D, Mantovani C, Pettersson J, Raffoul W, et al. Fibrin matrix for suspension of regenerative cells in an artificial nerve conduit. Journal of Plastic, Reconstructive & Aesthetic Surgery. 2008;61(6):669-675
  206. 206. Santiago LY, Clavijo-Alvarez J, Brayfield C, Rubin JP, Marra KG. Delivery of adipose-derived precursor cells for peripheral nerve repair. Cell Transplantation. 2009;18(2):145-158
  207. 207. Madduri S, Gander B. Schwann cell delivery of neurotrophic factors for peripheral nerve regeneration. Journal of the Peripheral Nervous System. 2010;15(2):93-103
  208. 208. Schlosshauer B, Muller E, Schroder B, Planck H, Muller HW. Rat Schwann cells in bioresorbable nerve guides to promote and accelerate axonal regeneration. Brain Research. 2003;963(1-2):321-326
  209. 209. Euler de Souza Lucena E, Guzen FP, Lopes de Paiva Cavalcanti JR, Galvao Barboza CA, Silva do Nascimento Junior E, Cavalcante Jde S. Experimental considerations concerning the use of stem cells and tissue engineering for facial nerve regeneration: A systematic review. Journal of Oral and Maxillofacial Surgery. 2014;72(5):1001-1012
  210. 210. Hadlock T, Elisseeff J, Langer R, Vacanti J, Cheney M. A tissue-engineered conduit for peripheral nerve repair. Archives of Otolaryngology—Head and Neck Surgery. 1998;124(10):1081-1086
  211. 211. Wang J, Ding F, Gu Y, Liu J, Gu X. Bone marrow mesenchymal stem cells promote cell proliferation and neurotrophic function of Schwann cells in vitro and in vivo. Brain Research. 2009;1262:7-15
  212. 212. 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. Experimental Neurology. 2007;207(2):267-274
  213. 213. 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. European Journal of Neuroscience. 2001;14(11):1771-1776
  214. 214. Wang HS, Hung SC, Peng ST, Huang CC, Wei HM, Guo YJ, et al. Mesenchymal stem cells in the Wharton’s jelly of the human umbilical cord. Stem Cells. 2004;22(7):1330-1337
  215. 215. Park HW, Lim MJ, Jung H, Lee SP, Paik KS, Chang MS. Human mesenchymal stem cell-derived Schwann cell-like cells exhibit neurotrophic effects, via distinct growth factor production, in a model of spinal cord injury. Glia. 2010;58(9):1118-1132
  216. 216. Joshi CV, Enver T. Plasticity revisited. Current Opinion in Cell Biology. 2002;14(6):749-755
  217. 217. Thuret S, Moon LD, Gage FH. Therapeutic interventions after spinal cord injury. Nature Review Neuroscience. 2006;7(8):628-643
  218. 218. Sarugaser R, Lickorish D, Baksh D, Hosseini MM, Davies JE. Human umbilical cord perivascular (HUCPV) cells: A source of mesenchymal progenitors. Stem Cells. 2005;23(2):220-229
  219. 219. Costa HJZR, Bento RF, Salomone R, Azzi-Nogueira D, Zanatta DB, Costa MP, et al. Mesenchymal bone marrow stem cells within polyglycolic acid tube observed in vivo after six weeks enhance facial nerve regeneration. Brain Research. 2013;1510:10-21
  220. 220. Colter DC, Class R, DiGirolamo CM, Prockop DJ. Rapid expansion of recycling stem cells in cultures of plastic-adherent cells from human bone marrow. Proceedings of the National Academy of Sciences United States of America. 2000;97(7):3213-3218
  221. 221. Gnecchi M, Melo LG. Bone marrow-derived mesenchymal stem cells: Isolation, expansion, characterization, viral transduction, and production of conditioned medium. Methods in Molecular Biology. 2009;482:281-294
  222. 222. Fu YS, Cheng YC, Lin MY, Cheng H, Chu PM, Chou SC, et al. Conversion of human umbilical cord mesenchymal stem cells in Wharton’s jelly to dopaminergic neurons in vitro: Potential therapeutic application for Parkinsonism. Stem Cells. 2006;24(1):115-124
  223. 223. Mitchell KE, Weiss ML, Mitchell BM, Martin P, Davis D, Morales L, et al. Matrix cells from Wharton’s jelly form neurons and glia. Stem Cells. 2003;21(1):50-60
  224. 224. Weiss ML, Medicetty S, Bledsoe AR, Rachakatla RS, Choi M, Merchav S, et al. Human umbilical cord matrix stem cells: Preliminary characterization and effect of transplantation in a rodent model of Parkinson’s disease. Stem Cells. 2006;24(3):781-792
  225. 225. Weiss ML, Mitchell KE, Hix JE, Medicetty S, El-Zarkouny SZ, Grieger D, et al. Transplantation of porcine umbilical cord matrix cells into the rat brain. Experimental Neurology. 2003;182(2):288-299
  226. 226. Fujimura J, Ogawa R, Mizuno H, Fukunaga Y, Suzuki H. Neural differentiation of adipose-derived stem cells isolated from GFP transgenic mice. Biochemical and Biophysical Research Communications. 2005;333(1):116-121
  227. 227. Liu BS, Yang YC, Shen CC. Regenerative effect of adipose tissue-derived stem cells transplantation using nerve conduit therapy on sciatic nerve injury in rats. Journal of Tissue Engineering and Regenerative Medicine. 2014;8(5):337-350
  228. 228. Xu Y, Liu Z, Liu L, Zhao C, Xiong F, Zhou C, et al. Neurospheres from rat adipose-derived stem cells could be induced into functional Schwann cell-like cells in vitro. BMC Neuroscience. 2008;9:21
  229. 229. Zavan B, Vindigni V, Gardin C, D’Avella D, Della Puppa A, Abatangelo G, et al. Neural potential of adipose stem cells. Discovery Medicine. 2010;10(50):37-43
  230. 230. di Summa PG, Kingham PJ, Campisi CC, Raffoul W, Kalbermatten DF. Collagen (NeuraGen®) nerve conduits and stem cells for peripheral nerve gap repair. Neuroscience Letters. 2014;572:26-31
  231. 231. Georgiou M, Golding JP, Loughlin AJ, Kingham PJ, Phillips JB. Engineered neural tissue with aligned, differentiated adipose-derived stem cells promotes peripheral nerve regeneration across a critical sized defect in rat sciatic nerve. Biomaterials. 2015;37:242-251
  232. 232. Arthur A, Rychkov G, Shi S, Koblar SA, Gronthos S. Adult human dental pulp stem cells differentiate toward functionally active neurons under appropriate environmental cues. Stem Cells. 2008;26(7):1787-1795
  233. 233. Huang AH, Snyder BR, Cheng PH, Chan AW. Putative dental pulp-derived stem/stromal cells promote proliferation and differentiation of endogenous neural cells in the hippocampus of mice. Stem Cells. 2008;26(10):2654-2663
  234. 234. Kim BC, Jun SM, Kim SY, Kwon YD, Choe SC, Kim EC, et al. Engineering three dimensional micro nerve tissue using postnatal stem cells from human dental apical papilla. Biotechnology and Bioengineering. 2017;114(4):903-914
  235. 235. Yamamoto T, Osako Y, Ito M, Murakami M, Hayashi Y, Horibe H, et al. Trophic effects of dental pulp stem cells on schwann cells in peripheral nerve regeneration. Cell Transplantation. 2016;25(1):183-193
  236. 236. Sasaki R, Aoki S, Yamato M, Uchiyama H, Wada K, Ogiuchi H, et al. PLGA artificial nerve conduits with dental pulp cells promote facial nerve regeneration. Journal of Tissue Engineering and Regenerative Medicine. 2011;5(10):823-830
  237. 237. Karbanová J, Soukup T, Suchánek J, Pytlík R, Corbeil D, Mokrý J. Characterization of dental pulp stem cells from impacted third molars cultured in low serum-containing medium. Cells Tissues Organs. 2011;193(6):344
  238. 238. Atari M, Barajas M, Hernández-Alfaro F, Gil C, Fabregat M, Ferrés Padró E, et al. Isolation of pluripotent stem cells from human third molar dental pulp. Histology and Histopathology. 2011;26(7):1057
  239. 239. Suchánek J, Soukup T, Ivancaková R, Karbanová J, Hubková V, Pytlík R, et al. Human dental pulp stem cells—isolation and long term cultivation. Acta Medica (Hradec Králové)/Universitas Carolina, Facultas Medica Hradec Králové. 2007;50(3):195-201
  240. 240. McLeod M, Hong M, Mukhida K, Sadi D, Ulalia R, Mendez I. Erythropoietin and GDNF enhance ventral mesencephalic fiber outgrowth and capillary proliferation following neural transplantation in a rodent model of Parkinson’s disease. European Journal of Neuroscience. 2006;24(2):361-370
  241. 241. Nosrat IV, Smith CA, Mullally P, Olson L, Nosrat CA. Dental pulp cells provide neurotrophic support for dopaminergic neurons and differentiate into neurons in vitro; implications for tissue engineering and repair in the nervous system. European Journal of Neuroscience. 2004;19(9):2388-2398
  242. 242. Rehman J, Traktuev D, Li J, Merfeld-Clauss S, Temm-Grove CJ, Bovenkerk JE, et al. Secretion of angiogenic and antiapoptotic factors by human adipose stromal cells. Circulation. 2004;109(10):1292-1298
  243. 243. Shi Y, Zhou L, Tian J, Wang Y. Transplantation of neural stem cells overexpressing glia-derived neurotrophic factor promotes facial nerve regeneration. Acta Oto-Laryngologica. 2009;129(8):906-914
  244. 244. Allen C, Eisenberg A, Mrsic J, Maysinger D. PCL-b-PEO micelles as a delivery vehicle for FK506: Assessment of a functional recovery of crushed peripheral nerve. Drug Delivery. 2000;7(3):139-145
  245. 245. Fine EG, Decosterd I, Papaloïzos M, Zurn AD, Aebischer P. GDNF and NGF released by synthetic guidance channels support sciatic nerve regeneration across a long gap. European Journal of Neuroscience. 2002;15(4):589-601
  246. 246. Kanje M, Lundborg G, Edström A. A new method for studies of the effects of locally applied drugs on peripheral nerve regeneration in vivo. Brain Research. 1988;439(1):116-121
  247. 247. Laird J, Mason G, Thomas K, Hargreaves R, Hill R. Acidic fibroblast growth factor stimulates motor and sensory axon regeneration after sciatic nerve crush in the rat. Neuroscience. 1995;65(1):209-216
  248. 248. Lee AC, Vivian MY, Lowe JB, Brenner MJ, Hunter DA, Mackinnon SE, et al. Controlled release of nerve growth factor enhances sciatic nerve regeneration. Experimental Neurology. 2003;184(1):295-303
  249. 249. Marquardt LM, Sakiyama-Elbert SE. Engineering peripheral nerve repair. Current Opinion in Biotechnology. 2013;24(5):887-892
  250. 250. Newman JP, Verity AN, Hawatmeh S, Fee WE, Terris DJ. Ciliary neurotrophic factor enhances peripheral nerve regeneration. Archives of Otolaryngology–Head & Neck Surgery. 1996;122(4):399-403
  251. 251. Santos X, Rodrigo J, Hontanilla B, Bilbao G. Evaluation of peripheral nerve regeneration by nerve growth factor locally administered with a novel system. Journal of Neuroscience Methods. 1998;85(1):119-127
  252. 252. Cui Q. Actions of neurotrophic factors and their signaling pathways in neuronal survival and axonal regeneration. Molecular Neurobiology. 2006;33(2):155-179
  253. 253. Sendtner M, Holtmann B, Hughes RA. The response of motoneurons to neurotrophins. Neurochemical Research. 1996;21(7):831-841
  254. 254. Jones MG, Munson JB, Thompson SW. A role for nerve growth factor in sympathetic sprouting in rat dorsal root ganglia. Pain. 1999;79(1):21-29
  255. 255. Koliatsos VE, Clatterbuck RE, Winslow JW, Cayouette MH. Evidence that brain-derived neurotrophic factor is a trophic factor for motor neurons in vivo. Neuron. 1993;10(3):359-367
  256. 256. Novikov L, Novikova L, Kellerth J-O. Brain-derived neurotrophic factor promotes axonal regeneration and long-term survival of adult rat spinal motoneurons in vivo. Neuroscience. 1997;79(3):765-774
  257. 257. Boyd J, Gordon T. A dose‐dependent facilitation and inhibition of peripheral nerve regeneration by brain‐derived neurotrophic factor. European Journal of Neuroscience. 2002;15(4):613-626
  258. 258. Chai H, Wu W, So K-F, Prevette D, Oppenheim R. Long-term effects of a single dose of brain-derived neurotrophic factor on motoneuron survival following spinal root avulsion in the adult rat. Neuroscience Letters. 1999;274(3):147-150
  259. 259. Bozkurt A, Lassner F, O’Dey D, Deumens R, Bocker A, Schwendt T, et al. The role of microstructured and interconnected pore channels in a collagen-based nerve guide on axonal regeneration in peripheral nerves. Biomaterials. 2012;33(5):1363-1375
  260. 260. Cobianchi S, Casals-Diaz L, Jaramillo J, Navarro X. Differential effects of activity dependent treatments on axonal regeneration and neuropathic pain after peripheral nerve injury. Experimental Neurology. 2013;240:157-167
  261. 261. Cunha C, Panseri S, Antonini S. Emerging nanotechnology approaches in tissue engineering for peripheral nerve regeneration. Nanomedicine: Nanotechnology, Biology and Medicine. 2011;7(1):50-59
  262. 262. Dubový P. Wallerian degeneration and peripheral nerve conditions for both axonal regeneration and neuropathic pain induction. Annals of Anatomy-Anatomischer Anzeiger. 2011;193(4):267-275
  263. 263. Ramburrun P, Kumar P, Choonara YE, Bijukumar D, du Toit LC, Pillay V. A review of bioactive release from nerve conduits as a neurotherapeutic strategy for neuronal growth in peripheral nerve injury. BioMed Research International. 2014;2014:132350. DOI:10.1155/2014/132350
  264. 264. Strauch RJ, Strauch B. Nerve conduits: An update on tubular nerve repair and reconstruction. Journal of Hand Surgery. 2013;38(6):1252-1255
  265. 265. Wu P, Spinner RJ, Gu Y, Yaszemski MJ, Windebank AJ, Wang H. Delayed repair of the peripheral nerve: A novel model in the rat sciatic nerve. Journal of Neuroscience Methods. 2013;214(1):37-44
  266. 266. Cho MS, Rinker BD, Weber RV, Chao JD, Ingari JV, Brooks D, et al. Functional outcome following nerve repair in the upper extremity using processed nerve allograft. Journal of Hand Surgery America. 2012;37(11):2340-2349
  267. 267. Henderson CE, Phillips HS, Pollock RA, Davies AM, Lemeulle C, Armanini M, et al. GDNF: A potent survival factor for motoneurons present in peripheral nerve and muscle. Science. 1994;266(5187):1062-1064
  268. 268. Johnson EO, Charchanti A, Soucacos PN. Nerve repair: Experimental and clinical evaluation of neurotrophic factors in peripheral nerve regeneration. Injury. 2008;39(Suppl 3):S37–S42
  269. 269. Oppenheim RW, Houenou LJ, Johnson JE, Lin LF, Li L, Lo AC, et al. Developing motor neurons rescued from programmed and axotomy-induced cell death by GDNF. Nature. 1995;373(6512):344-346
  270. 270. Wu W, Li L, Yick LW, Chai H, Xie Y, Yang Y, et al. GDNF and BDNF alter the expression of neuronal NOS, c-Jun, and p75 and prevent motoneuron death following spinal root avulsion in adult rats. Journal of Neurotrauma. 2003;20(6):603-612
  271. 271. Sendtner M, Dittrich F, Hughes RA, Thoenen H. Actions of CNTF and neurotrophins on degenerating motoneurons: Preclinical studies and clinical implications. Journal of Neurology Science. 1994;124(Suppl):77-83
  272. 272. Helgren ME, Squinto SP, Davis HL, Parry DJ, Boulton TG, Heck CS, et al. Trophic effect of ciliary neurotrophic factor on denervated skeletal muscle. Cell. 1994;76(3):493-504
  273. 273. Siegel SG, Patton B, English AW. Ciliary neurotrophic factor is required for motoneuron sprouting. Experimental Neurology. 2000;166(2):205-212
  274. 274. Fricker FR, Lago N, Balarajah S, Tsantoulas C, Tanna S, Zhu N, et al. Axonally derived neuregulin-1 is required for remyelination and regeneration after nerve injury in adulthood. Journal of Neuroscience. 2011;31(9):3225-3233
  275. 275. Syed N, Reddy K, Yang DP, Taveggia C, Salzer JL, Maurel P, et al. Soluble neuregulin-1 has bifunctional, concentration-dependent effects on Schwann cell myelination. Journal of Neuroscience. 2010;30(17):6122-6131
  276. 276. Stassart RM, Fledrich R, Velanac V, Brinkmann BG, Schwab MH, Meijer D, et al. A role for Schwann cell-derived neuregulin-1 in remyelination. Nature Neuroscience. 2013;16(1):48-54
  277. 277. Michailov GV, Sereda MW, Brinkmann BG, Fischer TM, Haug B, Birchmeier C, et al. Axonal neuregulin-1 regulates myelin sheath thickness. Science. 2004;304(5671):700-703
  278. 278. Taveggia C, Zanazzi G, Petrylak A, Yano H, Rosenbluth J, Einheber S, et al. Neuregulin-1 type III determines the ensheathment fate of axons. Neuron. 2005;47(5):681-694
  279. 279. Gambarotta G, Fregnan F, Gnavi S, Perroteau I. Neuregulin 1 role in Schwann cell regulation and potential applications to promote peripheral nerve regeneration. International Review of Neurobiology. 2013;108:223-256
  280. 280. Chan KM, Gordon T, Zochodne DW, Power HA. Improving peripheral nerve regeneration: From molecular mechanisms to potential therapeutic targets. Experimental Neurology. 2014;261:826-835
  281. 281. Liu L, Sun T, Xin F, Cui W, Guo J, Hu J. Nerve growth factor protects against alcohol-induced neurotoxicity in PC12 cells via PI3K/Akt/mTOR pathway. Alcohol and Alcoholism. 2017;52(1):12-18
  282. 282. Wu W, Liu Q, Liu Y, Yu Z, Wang Y. Dixdc1 targets CyclinD1 and p21 via PI3K pathway activation to promote Schwann cell proliferation after sciatic nerve crush. Biochemical and Biophysical Research Community. 2016;478(2):956-963
  283. 283. Wu W, Liu Y, Wang Y. Sam68 promotes Schwann cell proliferation by enhancing the PI3K/Akt pathway and acts on regeneration after sciatic nerve crush. Biochemical and Biophysical Research Community. 2016;473(4):1045-1051
  284. 284. Liu A, Prenger MS, Norton DD, Mei L, Kusiak JW, Bai G. Nerve growth factor uses Ras/ERK and phosphatidylinositol 3-kinase cascades to up-regulate the N-methyl-d-aspartate receptor 1 promoter. Journal of Biological Chemistry. 2001;276(48):45372-45379
  285. 285. Li J, O’Connor KL, Greeley GH, Jr., Blackshear PJ, Townsend CM, Jr., Evers BM. Myristoylated alanine-rich C kinase substrate-mediated neurotensin release via protein kinase C-delta downstream of the Rho/ROK pathway. Journal of Biological Chemistry. 2005;280(9):8351-8357
  286. 286. Ogita H, Takai Y. Nectins and nectin-like molecules: Roles in cell adhesion, polarization, movement, and proliferation. IUBMB Life. 2006;58(5-6):334-343
  287. 287. Takai Y, Irie K, Shimizu K, Sakisaka T, Ikeda W. Nectins and nectin-like molecules: Roles in cell adhesion, migration, and polarization. Cancer Science. 2003;94(8):655-667
  288. 288. Xu F, Zhang K, Lv P, Lu R, Zheng L, Zhao J. NECL1 coated PLGA as favorable conduits for repair of injured peripheral nerve. Materials Science and Engineering: C. 2017;70, Part 2:1132-1140
  289. 289. Elfar JC, Jacobson JA, Puzas JE, Rosier RN, Zuscik MJ. Erythropoietin accelerates functional recovery after peripheral nerve injury. The Journal of Bone and Joint Surgery. 2008;90(8):1644-1653
  290. 290. Yan Y, Sun HH, Hunter DA, Mackinnon SE, Johnson PJ. Efficacy of short-term FK506 administration on accelerating nerve regeneration. Neurorehabilitation and Neural Repair. 2012;26(6):570-580
  291. 291. Kostopoulos VK, Davis CL, Terzis JK. Effects of acetylo-L-carnitine in end-to-side neurorrhaphy: A pilot study. Microsurgery. 2009;29(6):456-463
  292. 292. Reid AJ, Shawcross SG, Hamilton AE, Wiberg M, Terenghi G. N-Acetylcysteine alters apoptotic gene expression in axotomised primary sensory afferent subpopulations. Neuroscience Research. 2009;65(2):148-155
  293. 293. Mohammadi R, Hirsaee M-A, Amini K. Improvement of functional recovery of transected peripheral nerve by means of artery grafts filled with diclofenac. International Journal of Surgery. 2013;11(3):259-264
  294. 294. Odaci E, Kaplan S. Chapter 16 Melatonin and Nerve Regeneration. International Review of Neurobiology. Vol. 87. Academic Press; 2009. pp. 317-335, ISBN: 978-0-12-375084-6
  295. 295. Fleming CE, Saraiva MJ, Sousa MM. Transthyretin enhances nerve regeneration. Journal of Neurochemistry. 2007;103(2):831-839
  296. 296. Sabri F, Gerth D, Tamula GR, Phung TC, Lynch KJ, Boughter JD, Jr. Novel technique for repair of severed peripheral nerves in rats using polyurea crosslinked silica aerogel scaffold. Journal of Investigative Surgery. 2014;27(5):294-303
  297. 297. Farjah GH, Heshmatian B, Karimipour M, Saberi A. Using eggshell membrane as nerve guide channels in peripheral nerve regeneration. Iranian Journal of Basic Medical Sciences. 2013;16(8):901-905
  298. 298. Bellamkonda RV. Peripheral nerve regeneration: An opinion on channels, scaffolds and anisotropy. Biomaterials. 2006;27(19):3515-3518
  299. 299. Madison RD, Da Silva CF, Dikkes P. Entubulation repair with protein additives increases the maximum nerve gap distance successfully bridged with tubular prostheses. Brain Research. 1988;447(2):325-334
  300. 300. Madison RD, da Silva C, Dikkes P, Sidman RL, Chiu TH. Peripheral nerve regeneration with entubulation repair: Comparison of biodegradable nerve guides versus polyethylene tubes and the effects of a laminin-containing gel. Experimental Neurology. 1987;95(2):378-390
  301. 301. Verdu E, Labrador RO, Rodriguez FJ, Ceballos D, Fores J, Navarro X. Alignment of collagen and laminin-containing gels improve nerve regeneration within silicone tubes. Restorative Neurology and Neuroscience. 2002;20(5):169-179
  302. 302. Rodriguez FJ, Verdu E, Ceballos D, Navarro X. Nerve guides seeded with autologous Schwann cells improve nerve regeneration. Experimental Neurology. 2000;161(2):571-584
  303. 303. Sinis N, Schaller HE, Becker ST, Schlosshauer B, Doser M, Roesner H, et al. Long nerve gaps limit the regenerative potential of bioartificial nerve conduits filled with Schwann cells. Restorative Neurology and Neuroscience. 2007;25(2):131-141
  304. 304. Sinis N, Schaller HE, Schulte-Eversum C, Schlosshauer B, Doser M, Dietz K, et al. Nerve regeneration across a 2-cm gap in the rat median nerve using a resorbable nerve conduit filled with Schwann cells. Journal of Neurosurgery. 2005;103(6):1067-1076
  305. 305. Al-Majed AA, Neumann CM, Brushart TM, Gordon T. Brief electrical stimulation promotes the speed and accuracy of motor axonal regeneration. Journal of Neuroscience. 2000;20(7):2602-2608
  306. 306. Brushart TM, Hoffman PN, Royall RM, Murinson BB, Witzel C, Gordon T. Electrical stimulation promotes motoneuron regeneration without increasing its speed or conditioning the neuron. Journal of Neuroscience. 2002;22(15):6631-6638
  307. 307. Gordon T, Brushart TM, Chan KM. Augmenting nerve regeneration with electrical stimulation. Neurology Research. 2008;30(10):1012-1022
  308. 308. Huang J, Hu X, Lu L, Ye Z, Wang Y, Luo Z. Electrical stimulation accelerates motor functional recovery in autograft-repaired 10 mm femoral nerve gap in rats. Journal of Neurotrauma. 2009;26(10):1805-1813
  309. 309. Lal D, Hetzler LT, Sharma N, Wurster RD, Marzo SJ, Jones KJ, et al. Electrical stimulation facilitates rat facial nerve recovery from a crush injury. Otolaryngology—Head and Neck Surgery. 2008;139(1):68-73
  310. 310. Sharma N, Coughlin L, Porter RG, Tanzer L, Wurster RD, Marzo SJ, et al. Effects of electrical stimulation and gonadal steroids on rat facial nerve regenerative properties. Restorative Neurology and Neuroscience. 2009;27(6):633-644
  311. 311. Sinis N, Horn F, Genchev B, Skouras E, Merkel D, Angelova SK, et al. Electrical stimulation of paralyzed vibrissal muscles reduces endplate reinnervation and does not promote motor recovery after facial nerve repair in rats. Annals of Anatomy. 2009;191(4):356-370
  312. 312. Sisken BF, Walker J, Orgel M. Prospects on clinical applications of electrical stimulation for nerve regeneration. Journal of Cellular Biochemistry. 1993;51(4):404-409
  313. 313. Skouras E, Merkel D, Grosheva M, Angelova SK, Schiffer G, Thelen U, et al. Manual stimulation, but not acute electrical stimulation prior to reconstructive surgery, improves functional recovery after facial nerve injury in rats. Restorative Neurology and Neuroscience. 2009;27(3):237-251
  314. 314. Hardy JG, Lee JY, Schmidt CE. Biomimetic conducting polymer-based tissue scaffolds. Current Opinion in Biotechnology. 2013;24(5):847-854
  315. 315. Lee JY, Bashur CA, Milroy CA, Forciniti L, Goldstein AS, Schmidt CE. Nerve growth factor-immobilized electrically conducting fibrous scaffolds for potential use in neural engineering applications. IEEE Transactions on Nanobioscience. 2012;11(1):15-21
  316. 316. Wang H, Yi SI, Pu X, Yu C. Simultaneously improving electrical conductivity and thermopower of polyaniline composites by utilizing carbon nanotubes as high mobility conduits. ACS Applied Materials & Interfaces. 2015;7(18):9589-9597
  317. 317. de Winter F, Hoyng S, Tannemaat M, Eggers R, Mason M, Malessy M, et al. Gene therapy approaches to enhance regeneration of the injured peripheral nerve. European Journal of Pharmacology. 2013;719(1-3):145-152
  318. 318. Mason MR, Tannemaat MR, Malessy MJ, Verhaagen J. Gene therapy for the peripheral nervous system: A strategy to repair the injured nerve? Current Gene Therapy. 2011;11(2):75-89
  319. 319. Hastie E, Samulski RJ. Adeno-associated virus at 50: A golden anniversary of discovery, research, and gene therapy success—A personal perspective. Human Gene Therapy. 2015;26(5):257-265
  320. 320. Boccaccini AR, Brauer DS, Hupa L. Bioactive Glasses: Fundamentals, Technology and Applications. Royal Society of Chemistry. 2016, ISVN: 978-1-78262-976-4
  321. 321. Baino F, Novajra G, Miguez-Pacheco V, Boccaccini AR, Vitale-Brovarone C. Bioactive glasses: Special applications outside the skeletal system. Journal of Non-Crystalline Solids. 2016;432, Part A:15-30

Written By

Sílvia Santos Pedrosa, Ana Rita Caseiro, José Domingos Santos and Ana Colette Maurício

Submitted: 02 December 2016 Reviewed: 02 May 2017 Published: 13 December 2017