Skip to content
BY 4.0 license Open Access Published by De Gruyter June 21, 2021

Material advancement in tissue-engineered nerve conduit

  • Wufei Dai , Yating Yang , Yumin Yang EMAIL logo and Wei Liu EMAIL logo
From the journal Nanotechnology Reviews

Abstract

Peripheral nerve injuries resulting from various traumatic events can cause mobility problems and sensory impairment, jeopardizing patients’ life quality and bringing serious economic burdens. Due to the shortcomings of autologous nerve grafts, such as limited tissue sources, unmatched size, and loss of innervation at the donor site, tissue-engineered nerve grafts using both natural and synthetic materials have been employed in the treatment of peripheral nerve defect and to promote nerve regeneration. Apart from traditional advantages such as good biocompatibility and controllable degradation, the development of fabrication technology and the advancement in material science have endowed tissue-engineered nerve conduits with upgraded properties such as biomimetic surface topography, extracellular matrix components, neurotrophic factors, and cell seeding, or a conduit with micropores on the surface for substance exchange and/or with fillers inside for microenvironment simulation. This article reviews recent progress in the biomaterials employed in fabricating tissue-engineered nerve conduits, in vitro characterization, and their applications in nerve repair in animal studies as well as in clinical trials.

1 Introduction

Peripheral nerve injury is common in traffic accidents, industrial accidents, and penetrating trauma, which can cause mobility problems, sensory impairment, and even paralysis and seriously affect patients’ life quality. Short defect of peripheral nerves is capable of self-regeneration by simple surgical suturing, whereas for a defect distance longer than 3 or 4 times of the diameter, other assistant treatments are essential [1,2,3].

As is shown in Figure 1, the basic functional unit of the peripheral nerve is the nerve bundle. The endoneurium consists of the endoneurial sheath, basal lamina, and collagen fibers. Single axons enwrapped by endoneurium are grouped into different bundles, surrounded by perineurium [4]. After severe injuries, disruption and necrosis of synapses and axons (Wallerian degeneration) may occur and gradually step into the stage of recovery [5]. Schwann cells are critical for nerve regeneration since they are able to transdifferentiate into repairing phenotype and modulate immune cells such as macrophages to facilitate nerve regeneration. They are also responsible for maintaining the structure of Büngner bands and basal lamina tubes [6,7]. Besides, certain growth factors and their receptors, such as the glial cell line-derived neurotrophic factor family, are also of great importance, and their expression could be upregulated temporarily to provide a favorable microenvironment [8,9].

Figure 1 
               The anatomy of peripheral nerves.
Figure 1

The anatomy of peripheral nerves.

For minor injuries, peripheral nerve tissue could be sutured directly or connected by fibrin sealant. As the first commercially available biological adhesive product launched in the 1970s, fibrin sealant has been widely used for the seamless repairment of peripheral nerves [10]. The satisfactory therapeutic effect of fibrin sealant has been reported in a 70-year-old patient with a 3 cm obturator nerve defect [11]. Studies showed no significant difference between surgical suturing and fibrin sealant in terms of total regenerated axons, fiber orientation, and the recovery of nerve conduction velocity. Apart from the recovery of anatomic integrity, fibrin sealant could also alleviate inflammation and fibrosis, reducing the risk of neuroma and scarring [12]. Moreover, as a user-friendly and time-saving device, fibrin sealant is highly suitable for emergency treatment or inexperienced operators. However, fibrin sealant cannot directly replace the nerve structure; thus, it does not apply to nerve injuries with a long defect, and there remain the risks of infection, necrosis, and seroma [13,14].

As for the lesions that are longer than 3 cm or four times the diameter (critical defect), end to end neurorrhaphy is of no help for the elongation rate and regeneration of nerve fibers [15], and nerve graft transplantation will be essential in order to avoid excessive tension [16]. The traditional gold-standard treatment for the critical defect in peripheral nerves is autologous nerve graft. Allogenic nerve graft is also commonly used in clinical practice. Both methods are disadvantaged in limited tissue sources, unmatched tissue size and structure, loss of innervation at the donor site, and immune rejection towards allogenic grafts [17]. As the most effective treatment, autogenous nerve tissues can provide supportive structure, guidance cues, neurotrophic factors, and Schwann cells without the risk of immune rejection, but complications such as secondary surgery and donor site dysfunction are of significant concern [18,19].

Decellularized allogenic nerve transplantation is an alternative method in which live Schwann cells are removed and only internal structure and crucial extracellular matrix components are maintained. With the maintenance of original structure, decellularized nerve grafts can perfectly guide the oriented growth of regenerating axons [20]. Soon after the implantation of short cell-free nerve allograft, axons, Schwann cells, and inflammatory cells from the host could migrate and gradually reconnect the distal nerve stump, realizing nerve repair and regeneration. Nevertheless, the regeneration potential of long decellularized nerve grafts is impaired because of the lack of Schwann cells [21]. The limited tissue source of human nerves is also a disadvantage. Thus, great efforts have been made to develop tissue-engineered nerve conduits. As the microenvironment factors could significantly modulate the growth of axons inside a nerve conduit, significant progress has been made in exploring new materials, modification of conduit structure and components, as well as nerve repair in animal models and human clinical trials. This article provides a comprehensive review of material advancement in tissue-engineered nerve conduit.

2 Materials for peripheral nerve regeneration

Materials with various properties in favor of nerve regeneration have been developed in the past decades, such as suitable mechanical strength, excellent biocompatibility, modulated degradation profile, and the capacity to encapsulate cells, growth factors, and bioactive proteins. Biomaterials for peripheral nerve repair can be generally divided into natural materials and synthetic materials in terms of their origins. Natural materials are represented by human cadaver, or animal-derived nerve grafts (autologous, allogenic, or xenogeneic nerve grafts), nonneural tissues (blood vessels, tendons, muscles, and outer membrane sheaths) [22,23,24,25,26,27], and biological macromolecules and their derivatives. However, immune rejection against allogenic and xenogeneic grafts is a major problem that restricts their application [28]. Biological macromolecules used as natural materials are primarily the extracellular matrix components with guaranteed biocompatibility [29]. Somehow, synthetic materials are more frequently used in in vivo studies due to their excellent mechanical properties and modulated degradability. In line with the original structure of nerve bundles, the tubular conduit has been considered the best physical format because it can minimize surgical trauma and store up neurotrophic factors released from the posttraumatic neural stem to promote nerve regeneration. Different manufacturing processes, including low-temperature deposition, electrospinning, leaching, 3D printing, lyophilization, and casting, have been employed to fabricate nerve conduit according to respective properties of different materials [30,31,32,33,34,35,36,37].

3 Natural materials

3.1 Silk fibroin (SF)

Natural silk is one of the natural macromolecular proteins, consisting of sericin and two silk fibroin monofilament fibers wrapped in the inner layer. Silk fibroin is a natural polymer composed of 20 amino acids, among which glycine, alanine, and serine are the main components [38]. As a major component of natural silk fiber, silk fibroin has been widely used in regenerative medicine in different forms for its self-assembly characteristics [39,40,41,42,43]. Silk fibroin has expanded the surface to promote cell adhesion, proliferation, migration, and nutrient diffusion [44,45]. Silk fibroin could improve the process of angiogenesis inside a conduit due to the stimulation of fibroblast proliferation and production of vascular endothelial growth factor, which was proven to benefit nerve regeneration [46]. As early as in 2006, Gu’s group demonstrated that silk fibroin was biocompatible with nerve cells and nerve tissues such as Schwann cells and hippocampal neurons [47,48]. They assembled a novel conduit with silk fibrin wall and oriented silk fibroin fibers inside to bridge over a sciatic nerve defect of 10 mm in vivo. The diameter and thickness of silk fibroin conduits were customizable with a stainless-steel casting mold fixed on the mold bottom. Furthermore, the lumen of the conduit was filled with 20 longitudinal-aligned silk fibroin fibers. As the silk fibroin material was covered with interconnected pores, this eggshell-like structure exhibited good mechanical stability and permeability. At six months post-operation, the silk fibroin conduit was replaced by nerve-like tissues without inflammatory reaction. And locomotor functional recovery of injured limbs was comparable to that of autologous nerve transplantation [49]. In addition to the conduit itself, the constructed microenvironment where normal peripheral nerve tissue could grow can promote nerve regeneration [50]. The same team also introduced a nature-mimicked microenvironment for nerve regeneration by coculturing dorsal root ganglia and Schwann cells in vitro and then seeded the cell mixture into a silk fibroin conduit. When bridging a 10 mm-long sciatic nerve defect in rats, they found that at the early stage (4 weeks), expression of nerve regeneration-related protein (N-cadherin and PMP22) in the silk fibroin-based conduit group was higher than that of control groups. At the late stage (12 weeks), the conduit produced a further improved outcome of nerve regeneration and functional recovery (walking track analysis), which was close to that of autologous transplantation [51]. Although native silk possesses good mechanical properties, when degummed, silk fibroin takes on impaired mechanical properties, and thus compromising the optimal properties for nerve conduit when using silk fibroin materials.

3.2 Collagen

As the main component of connective tissue and extracellular matrix, collagens are the most abundant proteins in mammals (30% of total protein mass), consisting of various glycoproteins synthesized by fibroblasts [52]. Moreover, collagen is characterized by a triple helix structure formed by combining three polypeptide chains, where several lysine can be detected for further modification [53]. Thus, collagen is one of the optimal biomaterial types that are widely used in regenerating and reconstructing both soft tissues and hard tissues in various forms [54,55,56,57]. In nerves, type IV procollagen produced by Schwann cells is the major structural component of both endoneurium and perineurium. Compared with synthetic polymers, biomaterials made of collagen tend to have better biocompatibility and less immunological rejection.

Keilhoff et al. employed collagen type I/III conduit to repair rat sciatic nerve defects with 4 mm length. Seven days after implantation in vitro, the scanning electron microscopy showed that Schwann cells maintained their typical shape. Abundant regenerated Schwann cells on the internal surface of collagen conduits and the formation of Büngner band were observed, and collagen conduits could be well-integrated with host tissues and fully revascularized in 5–7 days. During the revascular process, macrophages were also timely recruited to the reparative site to perform their immunological and regenerative roles. There was no resorption of the conduit after three weeks; thus, this conduit can be a candidate for tissue-engineered conduits [58]. The collagen conduits seem to be suitable for different lengths and sites of the various peripheral nerve defects. It was reported that collagen І/III conduits were of equal therapeutic efficiency to the golden standard in repairing 10 mm-long peroneal nerve defect in rat models. At 12 weeks post-operation, the whole conduits were resorbed, and nerve-like tissues could be observed. Compared to the control group (severed and no treatment), there was an increased number of small axons in the collagen conduit group, and the mean axonal loss per rat was significantly lower than that of the control group [59]. Besides, collagen is unstable under an aqueous environment and may become swollen and be dissolved gradually [44]. Thus, a combination of collagen with other materials is likely to improve its performance and expand its application of collagen conduit in nerve regeneration.

3.3 Chitin/chitosan

Chitin is the second most abundant polysaccharide on earth, only after cellulose. It exists widely in crustaceans, insects, or cell walls of fungi [60]. Chitosan is the byproduct of N-deacetylated chitin that can degrade into glucosamine and N-acetyl glucosamine in the lysosome [61,62]. As a natural material whose degradation rate can be regulated by deacetylation, chitosan is an ideal candidate for artificial nerve grafts with excellent biocompatibility, permeability, and plasticity [63,64]. Besides, chitosan is characterized by low production cost, easy access, and easy processing. Moreover, the intermediate product – chitooligosaccharides – is also able to promote axon regeneration by inhibiting neuronal apoptosis and facilitating cell adhesion [65,66].

Tissue fibrosis is partially responsible for the failed repair of injured peripheral nerves. One study has demonstrated that chitosan membranes could alleviate this process by reducing the infiltration of fibroblasts [67]. Some researchers began to study the role of chitosan in nerve regeneration using large animal models. Over a follow-up of 6 months, researchers employed chitosan conduit to repair sciatic nerve injuries (3 cm) in dog models. The dogs in the conduit group and autologous nerve graft group could walk and run freely at six months after surgery, and their posture tended to be normal. However, dogs in no treatment group suffered from skin ulcer and muscle atrophy and disability to use the repaired leg. And no rejection was observed in the chitosan group. Slower than normal nerves, the conduction velocity of the chitosan conduit group and the autologous group were comparable. Also, no edema, hematoma, inflammation, or abscess were found in the conduit group. The thickness of regenerated nerve in chitosan conduits was similar to that of the autologous group. In contrast, no regenerative nerve could be observed in the control group, indicating that the chitosan conduit could bridge the gap between the nerve stumps and stimulate nerve regeneration [68].

Some researchers managed to enhance the mechanical property and tensile strength of chitosan conduit by adding chitin powder to the chitosan solution [69].

In vivo implantation showed that the conduits fabricated by modified chitosan remained biocompatible to surrounding host tissues [70]. Chitosan could also be upgraded by combining with other bioactive substances or materials. Zhang et al. seeded mesenchymal stem cells (MSC) on erythropoietin (EPO)-loaded chitosan nerve conduit (EPO/Chi) by mixing Matrigel with MSCs, then injected the mixture into the conduits after their implantation. In vitro experiments showed improved cellular viability and migration of mesenchymal cells and Schwann cells as well as their neurotrophins secretion. According to silver staining, after 8 weeks, the number of axons in the injured nerve bundles increased significantly in Gel/MSC than in the hollow conduit group. In addition, the level of morphological restoration and regularity of nerve bundles also showed better results than the hollow conduit [71]. Undeniably, chitin and chitosan have been widely applied, but several drawbacks limit their scope of application, such as the risk of allergy to seafood and dependence on the low-pH environment.

4 Synthetic materials

4.1 Silicone

Silicone is characterized by nontoxic and bioinert characters, which does not react with any substances except for strong acid and alkaline and thus is nonabsorbable and non-deformable. Thanks to its nonporous structure, silicone cannot harbor bacteria internally and is easily sterilized [72]. Hollow or nonporous designs are the most primitive of all conduit designs. Francel et al. compared the performance of empty silicone conduits with the silicone conduits containing a short-inserted nerve segment, and with nerve autografts in repairing sciatic nerve defects (13–15 mm), the conduction velocity of silicone conduit with nerve segment group was comparable to that of the autologous group and showed signs of nerve regeneration by a large number of myelinated axons. Moreover, walking track analysis with sciatic function index (SFI) value calculation exhibited no significant difference between silicone conduit with nerve segment group and autologous group. On the contrary, in the hollow silicone conduit group, the chamber was blocked by the serum, and no functional recovery could be observed [73]. Thus, silicone conduits are often loaded with various growth factors, cells, and other materials to promote their performance [74,75,76].

Although readily available, silicone cannot replace tissue transplantation in patients for extended nerve defects as it can block the interaction between cells inside and microenvironmental factors outside. Besides, since silicone is nondegradable in vivo, neuroma formation and nerve dysfunction induced by foreign body reaction and compression of neo-tissues may cause local discomfort and lead to secondary operation to remove the implant.

4.2 Polytetrafluoroethylene (PTFE)

As another bioinert synthetic material that has been widely applied in nerve repair, polytetrafluoroethylene (PTFE) is of high surface tension and viscosity resistance. Because of the chemical inertness, its modification was difficult by surface coating and thus has been a scientific and technological challenge for decades [77]. When PTFE conduits were applied to repair a 10 mm-long nerve defect, Zetti et al. introduced a novel PTFE conduit with porous structure with various sizes by extrusion and mechanical stretching. Six months after the surgery, all the rats were reinnervated on the operated side, according to electromyographic studies. The process of axonal regeneration tended to be significantly active because histological tests can observe thinly myelinated fibers and medium-large clusters, thus confirming the feasibility of nerve regeneration with PTFE-based conduits [78]. However, there was no control group in this study, and thus the results were less reliable. Labroo et al. also reported a GDNF-embedded PTFE conduit for nerve repair using a GDNF reservoir between two PTFE conduits. They found that the drug release rate could be adjusted by the size and quantity of the pores. Afterward, this GDNF-embedded nerve conduit was used to reconnect sciatic nerve defect of 10 mm length in BL6 YFP mouse models. Compared with using PTFE conduits alone, at 10 weeks after surgery, the distal nerve in the GDNF conduit group revealed a significantly higher number of myelinated axons. Additionally, at 18 weeks postimplantation, the GDNF conduit groups had significant myelination with healthier-looking nerve surrounded by multiple fibrous layers [79]. Same as silicone conduit, PTFE conduits can hardly break down in vivo, which may trigger persistent rejection reaction and hinder the general repair process. Generally speaking, the development of nondegradable guide conduits has not been actively pursued due to the limitations of permanent material [80].

4.3 Polyglycolic acid (PGA)

Polyglycolic acid (PGA) is a rigid thermoplastic polymer with features of high crystallinity, high tensile modulus, and low solubility in organic solvents. PGA retains both hydrophilic and lipophilic within their repetitive units, allowing processing and modification into different forms without additives [81]. Because of the nonspecific fracture of the ester chain, PGA materials could lose their strength (1–2 months) and total mass (6–12 months) fast in vivo [82,83].

The sciatic nerve has been extensively studied because of easy access and evaluation, whereas some of the nerves are more difficult to reach functional regeneration after injury, such as the recurrent laryngeal nerve (RLN), and thus are rarely reported. Sentürk et al. used a PGA conduit to repair damaged RLN, and they found that the vocal cord mobility was proportionally higher in the PGA conduit group than in other groups (only nerve cuts and primary repair group). In fact, there was a positive relationship between axon number and vocal cord mobility. And the mean number of axons in the PGA conduit group was the highest among the three groups. Histological evaluation outcomes indicated that this conduit has the potential to repair peripheral nerve damage by creating an isolated environment, then provide a more accurate orientation of nerve fibers [84]. Nevertheless, it was observed that PGA conduit might cause significant foreign body reaction and inflammation likely caused by rapidly degraded products [84].

4.4 Polyurethane (PU)

As one of the most versatile polymers, polyurethane (PU) accounts for 6% of the world’s plastic use [85]. PU is produced by the polyaddition reaction of long-chain oligomer diol with diisocyanate. The typical microphase-separated structure of the hard segment (diisocyanate and chain extender) and soft segment (long-chain diol) or oligodiols endows perfect mechanical properties such as wear resistance, elasticity, and high tensile strength. Moreover, PU could be fabricated into various hardness and different forms by modulating its cross-link density and molecular structure, resulting in regulated mechanical properties and permeability [86]. In vivo studies also found that PU could promote human umbilical vein endothelial cells’ adhesion and resist hemolysis and clotting [87], making it a promising material for regenerative medicine. In 2015, Wu et al. reported that PU could significantly improve Schwann cells’ myelin-related gene expression and neurotrophin secretion, which suggested that PU could serve as a promising candidate for peripheral nerve regeneration [88]. Meanwhile, a single PU nerve conduit with a pore size of 1–5 mm and a porosity of 88% was investigated to repair a 10 mm nerve defect. When cultured in vitro, the glial cell could well grow on PU conduits, indicating good cytocompatibility of PU conduits. After in vivo implantation, no inflammatory or adverse tissue reaction was observed. Furthermore, the regenerated neurofilaments were found distributed regularly inside the PU conduit, similar to the pattern of the autologous group. At different time points postsurgery (2, 4, 8, 10, 14, and 20 weeks), functional nerve recovery was observed by walking track analysis, which showed that the PU conduit group exhibited a higher SFI value than that of PCL and silicone groups, revealing a better reparative capability [89].

4.5 Poly(l-lactide-co-caprolactone) (PLCL)

PLCL is an aliphatic polyester copolymer prepared by ring-open polymerization of poly(l-lactic acid) (PLLA) and poly(e-caprolactone) (PCL). The strength and degradation rate of PLCL can also be modified by changing the proportion of PLLA and PCL. The copolymer PLCL made up for the brittle behavior of PLLA and the low stiffness of PCL and improved the local acidic environment caused by PLLA degradation [90]. PLCL can be customized for desired degradation rate by changing the ratio of PCL/PLLA, and the more PLLA, the higher the degradation rate [91]. Moreover, when the content of PCL is increased, the shape memory effect of PLCL will also be improved [90].

As early as 2009, electrospun nanofibrous substrates of PLCL have been reported to promote mesenchymal stem cell differentiation into neuronal cells for nerve tissue engineering [92]. It was recently demonstrated that modified PLCL conduits could significantly improve SC migration, proliferation, and myelination, promote the differentiation of PC12 cells (exhibiting many neuronal properties), and upregulate the expression of neurite growth-related proteins in vitro [93]. In vivo studies further proved that the PLCL conduit could fully restore motor function (evaluated by triceps surae muscles weight ratio) and nerve conduction function (electrophysiological analysis) in 12 weeks, and the therapeutic effect was equal to that of the gold-standard treatment (autologous transplantation) [93]. Moreover, compared with the electrospun caprolactone group, PLCL conduit had a lower risk of neuroma formation or infection [94]. However, PLCL is highly hydrophobic and lacks cell recognition sites, and thus causes low biocompatibility [95]. Enhancement in the topological modification of PLCL to increase the cytocompatibility and cell adhesion remains a challenge for researchers in the future.

4.6 PLLA

Polymers of lactic acid (PLA) can be decomposed into glycolate and pyruvate and hydrolyzed into H2O and CO2 eventually in vivo. Owing to the asymmetric carbon atom in the lactic acid molecule, PLA is featured with optical activity and composed of D-type (PDLA, right-handed rotation) and L-type (PLLA, left-handed rotation) [96]. Both PLA and PLLA are widely applied in the field of tissue-engineered repair [97]. The amorphous areas of PLLA usually degrade faster, leading to the increase of crystal concentration of the rest materials that become resistant to degradation. Therefore, they may remain in the body for a long time accompanied by inflammatory cells [98].

As a plastic material that is easy to obtain and process [99], a single PLLA conduit was implanted in Sprague Dawley rats to repair a defect of 12 mm in the right sciatic nerve. Evaluation of long-term (8 months) recovery showed that the PLLA conduit could maintain the structural integrity and induce vascularization. The SFI value demonstrated no difference between the autologous graft group and the PLLA conduit group. Besides, the number of axons per unit area in the PLLA conduit was comparable to that in isograft controls, suggesting that the PLLA conduit might be used as a suitable conduit for nerve regeneration [100]. The same team reported similar treatment outcomes in another study where allogeneic Schwann cells were seeded onto the PLLA conduit to facilitate nerve repairing [101]. It could cause some problems such as incomplete degradation and decreasing pH values as a relatively slow-degradation material. Thus, PLLA implant may incur late-phase inflammation, cell necrosis, and delayed axon recovery in vivo [98].

4.7 Poly(lactic-co-glycolic acid) (PLGA)

PLGA is one of the most popular biodegradable copolymers, consisting of two different monomers – polylactic acid and polyglycolic acid and it can be synthesized by ring-opening copolymerization in the presence of catalysts [102]. Several factors can affect the degradation of PLGA copolymers, such as molecular weight, crystallinity, and the ratio of lactic acid to glycolic acid. Generally, PLGA degrades faster when the glycolic acid part increases (<50%) in the main chain [103,104]. As a biosafe material that can promote the formation of capsules and film, PLGA has been widely used in the pharmaceutical and tissue engineering industry [105]. Researchers found that PLGA membrane pre-coated with Nectin-like molecule 1 (NECL1) could mimic natural axons to improve the adhesion of Schwann cells. In vitro, they compared the single PLGA conduit with the PLGA conduit coated with NECL1 of different concentrations (PLGA/NECL1) and observed a significant increase in the gene expression of neurotrophic factors related to nerve regeneration in the PLGA/NECL1 group (the concentration of 50 ng/mL had the best performance). Moreover, MTT assay showed that the PLGA/NECL1 conduit could also better promote cell viability and proliferation. In vivo, they evaluated nerve conduction velocity and muscle weight ratio among single PLGA/NECL1 group, PLGA conduit group, and transected group, and they found that the PLGA/NECL1 group was better-performed over the rest two control groups (single PLGA and transected group). SFI results also indicated that the PLGA/NECL1 group had a faster recovery by 12 weeks. Thus, they concluded that PLGA conduit pre-coated with NECL1 could facilitate the regeneration of peripheral nerves [106].

However, when applied in vivo, the relatively low mechanical properties and non-bioactive nature are the apparent drawbacks of pure PLGA conduit [107]. Different modifications have been incorporated into PLGA to create polymer composites with desirable properties to overcome these disadvantages. Hou et al. tried to promote the repairing effect of PLGA conduit by combining the electrospinning PLGA membrane and freeze-dried oxidized antibacterial cellulose and collagen sponge. The ECM-like sponge was expected to facilitate tissue regeneration, while the external PLGA conduit could bridge the defect and provide mechanical support. The conduits were implanted in rats with a 10 mm defect in the sciatic nerve. Staining results demonstrated that the number of nerve fibers and blood cells/vessels in modified conduit groups was significantly higher than that of hollow conduit groups and exhibited uniform and orderly distribution. By 8 weeks postoperation, the positive expressions of proteins related to nerve regeneration (S-100 and NF-200) in modified conduit groups were close to those of autologous groups and much higher than those of hollow conduit groups [108].

4.8 Composite materials

As described, natural substances are good in biocompatibility but relatively weak in mechanical properties, whereas synthetic materials are the opposite. Composite materials have been developed and evolved to make the best use of their prominent features. It was believed that composite materials are a promising alternative to traditional nerve conduits in the future.

Composite materials were fabricated initially by simple mixture of various components. Nieto-Suarez et al. observed that the porous chitosan/gelatin conduit prepared through ice segregation-induced self-assembly (ISISA) was stable and highly swollen, suitable for fabricating tissue-engineered nerve conduits [109]. Some researchers used one material as the major and modified it with other materials. Ouyang et al. invented a seamless axially aligned nerve graft composed of collagen (25%) and PLGA nanofibers by a one-step modified electrospinning technique. With the application of insulating mandrel of different sizes, the fibers on both inner and outer walls were aligned along the longitudinal axis, and the inner diameter of the scaffold could be adjusted from 1 to 10 mm. After being seeded into the conduits in vitro, Schwann cells in conduits resembled a morphology similar to that of normal nerves, indicating good biocompatibility of the conduit. The gastrocnemius muscle cross-sectional and SFI values in both autograft and the uniaxially aligned seamless collagen/PLGA conduit groups were larger and higher than those of randomly oriented groups. HE staining analysis also demonstrated well-organized structures of regenerated axons, indicating a good reparative effect of uniaxially aligned seamless collagen/PLGA nanofibers [110].

Researchers started making new tissue-engineered nerve conduits with three or more bioactive materials combined using advanced fabrication technology based on a better understanding of nerve regeneration mechanisms. Wang et al. developed a novel PLGA-silk fibroin-collagen (PLGA-SF-COL) conduit with improved hydrophilicity and mechanical property. When cultured for 3 days on the conduits, Schwann cells revealed good cytocompatibility to the conduit as they attached well and exhibited the normal phenotype (bipolar or tripolar morphology). MTT assay also showed higher value in PLGA-SF-COL conduit than that in the pure PLGA conduit group. They concluded that PLGA-SF-COL conduits composed of 30% PLGA, 35% silk fibroin, and 35% collagen had the potential to facilitate nerve regeneration. However, they only conducted cytology experiments in vitro; studies in vivo need to be further validated [111] (Figure 2).

Figure 2 
                  Schematic illustration of peripheral nerve repair strategies via tissue-engineered conduit materials.
Figure 2

Schematic illustration of peripheral nerve repair strategies via tissue-engineered conduit materials.

4.9 Clinical trials

As research continues, several conduits have been applied in clinical trials and exhibited promising outcomes (Table 1 and Figure 3). When the nerve defect was minor, the microsurgical suture is the first choice in clinical treatment. As early as in 1982, Lundborg et al. reported a clinical randomized prospective study of repairing median and ulnar nerve defect with 3–5 mm length. The study followed 28 patients for up to five years to compare the repair outcome of tubular treatment (silicone conduits) with traditional microsurgical repair. After 5 years, they found no statistical differences in clinical or neurophysiological outcomes of tubular repair and the traditional microsurgical treatment. Moreover, cold intolerance was much better relieved in the tube group than in the microsurgical suturing group. Therefore, they concluded that tubular treatment could be an alternative for traditional microsurgical repair [112].

Table 1

Clinical studies of materials employed in periphery nerve repairs

Materials Nerves Length (mm) Outcomes Reference
Silicone Median and ulnar nerve 3–5 Comparable therapeutic effects (except for cold intolerance) to that of the traditional microsurgical treatment (direct suture) [112]
Collagen Brachial plexus nerve ≤20 MSC ≥ 0.6 in 4/5 cases [113]
PTFE Median and ulnar nerve 15–60 15–40 mm: functional recovery of both motor and sensory nerves in 78.6% cases (15 of 17 median nerves and 7 of 11 ulnar nerves); 41–60 mm: functional recovery in 13.3% cases (1 of 4 median nerves and 1 of 11 ulnar nerves) [114]
Inferior alveolar and lingual nerve 2–15 Functional recovery in 2/7 cases (2 inferior alveolar nerves <3 mm) [115]
Polyglycolic acid (PGA) Digital nerve 5–30 Excellent functional sensation recovery in 33% cases and good in 53% cases; pain sense recovery in over 77% cases [116]
Inferior alveolar and lingual nerve / Allodynia or dysesthesia resolved in 6/10 cases (3 inferior alveolar nerves and 3 lingual nerves); sensory functional recovery in all cases; reconnection with normal nerve-like structure in 9/10 cases [117]
PGA and collagen Frontalis branch of facial nerve 11 mm and 28 mm Partial muscle contraction and recovered distal latency in 1/2 cases (by 2 months); symmetrical eyebrow-lifting in 2/2 cases (by 5 months) [118]

“/”: The length was not mentioned in the original articles.

Figure 3 
                  Schematic illustration of clinical trials of peripheral nerve repair with different types of nerve conduits at different anatomic regions.
Figure 3

Schematic illustration of clinical trials of peripheral nerve repair with different types of nerve conduits at different anatomic regions.

The collagen-based conduit was first applied for the clinical repair of brachial plexus nerve injuries in 2006. They defined the motor scale composite (MSC) as a numeric measure for representing the brachial plexus palsy motor outcome. And MSC value ≥0.6 represents good functional recovery and ≥0.75 represents an excellent functional recovery. Finally, three patients (total of 5) exhibited an excellent recovery (MSC ≥0.75), and one reached a good recovery (MSC ≥0.6) at 2 years postoperation. In all five patients, applying collagen-based conduits alone could improve the MSC value by 69 and 78% at 1 and 2 years, respectively. Moreover, joint movement scores of four patients also improved to a range of motion of more than 56%. Overall, most patients achieved a good functional recovery with improved functions of feeding and dressing by themselves [113].

Stanec et al. describe the effectiveness of expanded-polytetrafluoroethylene (e-PTFE) conduits in the clinical repair of nerves in the upper limbs. In the study, all the patients were evaluated and classified into 2 groups (minor defect and major defect). In the minor group (1.5–4 cm defect), most (78.6%) of the patients showed functional recovery of both motor and sensory nerves (15 of 17 reconstructed median nerves and 7 of 11 reconstructed ulnar nerves), whereas in the major group (4.1–6 cm defect), only a minority (13.3%) of the patients achieved functional recovery (1 of 4 reconstructed median nerves and 1 of 11 reconstructed ulnar nerves). These results concluded that long defect nerve injury remains a challenge to PTFE conduit, although it could be a measure to repair median or ulnar nerve defects shorter than 4 cm [114].

However, others reported that successful repair (sensation restoration) of inferior alveolar nerve and lingual nerve via Gore-Tex tubing (e-PTFE) was only observed in two of seven patients whose nerve injuries were shorter than 3 mm. One patient was fully recovered, and the other was close to expected recovery; the rest cases showed compromised sensational restoration after 3 years. It is known that inferior alveolar nerves tend to have a higher spontaneous recovery rate than lingual nerves. In this study, the only two cases succeeded in repairing are both inferior alveolar nerves shorter than 3 mm and thus the results could hardly indicate that Gore-Tex tubing was capable of repairing nerve defect [115].

As early as in 1985, researchers applied PGA conduits to reconstruct nerve defects with an averaged defect of 1.7 cm (from 0.5 to 3.0 cm) in 15 patients with a follow-up evaluation of sensibility for an average of 22.4 months. Consequently, 33% achieved excellent functional sensation and 53% achieved good functional sensation in their evaluation according to sensory relearning exercise. Also, over 77% of patients who lost their pain feeling before the surgery could achieve good repair, which revealed a satisfactory repair outcome [116].

Several composite materials have already been applied in clinical trials too. Seo et al. reported that PGA conduits coated with collagen (PGA-c) were applied to bridge lingual nerve or inferior alveolar nerve in a university hospital, and the outcomes were evaluated at 2 months to 8 years postsurgery. Allodynia or dysesthesia was resolved in 60% (6/10) and greatly reduced in 40% (4/10) of the patients. In almost all patients, functional sensory recovery can be observed. Thus, the results demonstrated that PGA-c conduits could improve the patients’ condition according to quantitative sensory testing results (brush stroke perception, mechanical touch threshold, sensitivity to cold/hot stimuli). And normal nerve-like structure could be observed in 9/10 cases, which indicated PGA-c conduits as a promising way for nerve regeneration [117]. Inada et al. reported that cylindrically woven PGA conduit filled with collagen was applied to repair peripheral motor nerve defects in two patients. Two months after the operation, partial contraction of the left frontalis muscle could be observed in one patient. Electrophysiological testing showed that the distal latency of both patients recovered and became almost symmetrical with the normal side. Five months later, both patients could manage to lift their eyebrows symmetrically with full recovery [118].

5 Conclusion

The recovery of injured periphery nerve highly relies on the structural integrity and functional restoration of nerve conduit. As is shown in Tables 1 and 2, advances in biomaterials have greatly contributed to the development of tissue-engineered nerve conduit and periphery nerve regeneration in the past decades. With upgraded biosafety and mechanical features, tissue-engineered nerve conduit has been evolved from a single hollow tube that merely connects both ends to the composite structure with multiple layers and a delicate topological surface for exchanging substances and regulating cell behaviors. Accumulated studies in vitro and in vivo have also confirmed the facilitating effects of newly developed materials in nerve regeneration, some of which have even been testified in clinical trials with satisfactory outcomes. In the future, tissue-engineered nerve conduits are expected to self-adjust to their in vivo complex microenvironments where the conduits are implanted [119]. Moreover, upgraded materials should actively participate in the repairing process by modulating different cells (Schwann cells, stem cells, and immune cells) and neurotrophic factors [120,121,122] and responding to physical stimulation as well as electrophysiological signals to accelerate nerve restoration from all aspects. With the further research of biomaterial science by combining fabrication and manufacturing technologies with chemical components and biological mechanisms, clinical application of tissue-engineered conduit will be a promising way of peripheral nerve regeneration.

Table 2

In vivo studies of materials employed in periphery nerve repairs

Materials Objects Nerves Length (mm) Outcomes Reference
Silk fibroin SD rats Sciatic nerve 10 Replaced by nerve-like tissues in 6 months [49]
SD rats Sciatic nerve 10 By 4 weeks (early stage): increased expression of nerve regeneration-related proteins [51]
By 12 weeks (late stage): densely distributed myelinated nerve fibers and muscles with large cross-sectional area
Collagen SD rats Sciatic nerve 4 Schwann cells regeneration, Bü-ngner band formation, and hematogenous macrophages recruitment in 5–7 days [58]
SD rats Peroneal nerve 30–40 Increased number of small axons and nerve-like tissues by 12 weeks [59]
Chitosan Dog Sciatic nerve 30 Comparable motor recovery (walk, run, posture), conduction velocity, and thickness of regenerated nerves to that of the autograft group [68]
SD rats Sciatic nerve 5 Significantly improved axon density, morphological restoration, and nerve bundle distribution in both the conduit + Gel/MSC group and the conduit group by 8 weeks [71]
Silicone Lewis rats Sciatic nerve 13–15 Increased number of myelinated axons; comparable conduction velocity and SFI value to that of the autograft group [73]
Polytetrafluoroethylene (PTFE) Wistar rats Sciatic nerve 10 The operated side was reinnervated in all cases by 6 months [78]
BL6 YFP mouse Sciatic nerve 10 Increased number of myelinated axons in the GDNF conduit group by 10 weeks; healthier-looking nerves and significant myelination in the FK506 group and the GDNF conduit group by 18 weeks [79]
Polyglycolic acid (PGA) Wistar rats Recurrent laryngeal nerve / Accurate orientation of nerve fibers, enhanced vocal cord mobility, and increased number of axons in the PGA conduit group [84]
Polyurethane (PU) SD rats Sciatic nerve 10 End-end connection of regenerated nerves, regularly distributed neurofilaments, and an obviously higher SFI value in the PU conduit group [89]
Poly(l-lactic-co-ε-caprolactone) SD rats Sciatic nerve 12 Improved SC migration, proliferation, myelination, and upregulated expression of neurite growth-related proteins; nerve conduction and motor function were restored in all cases by 12 weeks; comparable therapeutic effects to that of the autograft transplantation [93]
Poly(l-lactic acid) (PLLA) SD rats Sciatic nerve 10 Promoted vascularization and axon density [100]
Poly(lactic-co-glycolic acid) (PLGA) SD rats Tibio-peroneal bifurcation 12 Promoted cell proliferation and neurotrophic factor levels in the PLGA/NECL1 group; sciatic nerve stumps were reconnected in the PLGA/NECL1 groups by 12 weeks [106]
PLGA, oxidized bacterial cellulose and collagen SD rats Sciatic nerve 10 Increased number of myelinated nerve fibers and blood cells/vessels; comparable SFI value to that of the autograft group [108]
PLGA and collagen SD rats Sciatic nerve 15 Better SFI recovery and larger gastrocnemius muscle cross-sectional area in the uniaxially aligned collagen/PLGA conduit group than in the randomly oriented collagen/PLGA conduit group [110]

“/”: The length was not mentioned in the original articles.


These authors contributed equally to this work.

tel: +86-21-2327-1699, fax: +86-21-5307-8128

Acknowledgements

This study was supported by National Key Research and Development Program of China (2018YFC1105800).

  1. Funding information: This study was supported by National Key Research and Development Program of China (2018YFC1105800).

  2. Author contributions: All authors have accepted responsibility for the entire content of this manuscript and approved its submission.

  3. Conflict of interest: The authors state no conflict of interest.

References

[1] Daly WT, Knight AM, Wang H, de Boer R, Giusti G, Dadsetan M, et al. Comparison and characterization of multiple biomaterial conduits for peripheral nerve repair. Biomaterials. 2013;34(34):8630–9.10.1016/j.biomaterials.2013.07.086Search in Google Scholar PubMed

[2] Amini S, Saudi A, Amirpour N, Jahromi M, Najafabadi SS, Kazemi M, et al. Application of electrospun polycaprolactone fibers embedding lignin nanoparticle for peripheral nerve regeneration: in vitro and in vivo study. Int J Biol Macromol. 2020;159:154–73.10.1016/j.ijbiomac.2020.05.073Search in Google Scholar PubMed

[3] Meyer C, Stenberg L, Gonzalez-Perez F, Wrobel S, Ronchi G, Udina E, et al. Chitosan-film enhanced chitosan nerve guides for long-distance regeneration of peripheral nerves. Biomaterials. 2016;76:33–51.10.1016/j.biomaterials.2015.10.040Search in Google Scholar PubMed

[4] Pauziene N, Pauza DH, Stropus R. Morphology of human intracardiac nerves: an electron microscope study. J Anat. 2000;197(Pt 3):437–59.10.1046/j.1469-7580.2000.19730437.xSearch in Google Scholar PubMed PubMed Central

[5] Kline RA, Dissanayake KN, Hurtado ML, Martínez NW, Ahl A, Mole AJ, et al. Altered mitochondrial bioenergetics are responsible for the delay in Wallerian degeneration observed in neonatal mice. Neurobiol Dis. 2019;130:104496.10.1016/j.nbd.2019.104496Search in Google Scholar PubMed PubMed Central

[6] Scheib J, Höke A. Advances in peripheral nerve regeneration. Nat Rev Neurol. 2013;9(12):668–76.10.1038/nrneurol.2013.227Search in Google Scholar PubMed

[7] Weinberg HJ, Spencer PS. The fate of Schwann cells isolated from axonal contact. J Neurocytol. 1978;7(5):555–69.10.1007/BF01260889Search in Google Scholar PubMed

[8] Li G, Xiao Q, Zhang L, Zhao Y, Yang Y. Nerve growth factor loaded heparin/chitosan scaffolds for accelerating peripheral nerve regeneration. Carbohydr Polym. 2017;171:39–49.10.1016/j.carbpol.2017.05.006Search in Google Scholar PubMed

[9] Zhang L, Yang W, Tao K, Song Y, Xie H, Wang J, et al. Sustained local release of NGF from a Chitosan-Sericin composite scaffold for treating chronic nerve compression. ACS Appl Mater Interfaces. 2017;9(4):3432–44.10.1021/acsami.6b14691Search in Google Scholar PubMed

[10] Ahmad E, Fatima MT, Hoque M, Owais M, Saleemuddin M. Fibrin matrices: the versatile therapeutic delivery systems. Int J Biol Macromol. 2015;81:121–36.10.1016/j.ijbiomac.2015.07.054Search in Google Scholar PubMed

[11] Scaletta G, Bizzarri N, Lauretti L, Scambia G, Fagotti A. Obturator nerve regeneration using a genito-femoral graft placed only by fibrin sealant (Tisseel®). Gynecol Oncol. 2019;153(3):703–4.10.1016/j.ygyno.2019.03.248Search in Google Scholar

[12] Koulaxouzidis G, Reim G, Witzel C. Fibrin glue repair leads to enhanced axonal elongation during early peripheral nerve regeneration in an in vivo mouse model. Neural Regen Res. 2015;10(7):1166–71.10.4103/1673-5374.156992Search in Google Scholar

[13] Barros LC Jr, Ferreira RS, Barraviera SR Jr, Stolf HO, Thomazini-Santos IA, Mendes-Giannini MJ, et al. A new fibrin sealant from Crotalus durissus terrificus venom: applications in medicine. J Toxicol Env Health B Crit Rev. 2009;12(8):553–71.10.1080/10937400903442514Search in Google Scholar

[14] Abbade LPF, Ferreira RS Jr, Dos Santos LD, Barraviera B. Chronic venous ulcers: a review on treatment with fibrin sealant and prognostic advances using proteomic strategies. J Venom Anim Toxins Incl Trop Dis. 2020;26:e20190101.10.1590/1678-9199-jvatitd-2019-0101Search in Google Scholar

[15] Dahlin LB, Lundborg G. Use of tubes in peripheral nerve repair. Neurosurg Clin N Am. 2001;12(2):341–52.10.1016/S1042-3680(18)30059-7Search in Google Scholar

[16] Howarth HM, Orozco E, Lovering RM, Shah SB. A comparative assessment of lengthening followed by end-to-end repair and isograft repair of chronically injured peripheral nerves. Exp Neurol. 2020;331:113328.10.1016/j.expneurol.2020.113328Search in Google Scholar PubMed

[17] Koudehi MF, Fooladi AA, Mansoori K, Jamalpoor Z, Amiri A, Nourani MR. Preparation and evaluation of novel nano-bioglass/gelatin conduit for peripheral nerve regeneration. J Mater Sci Mater Med. 2014;25(2):363–73.10.1007/s10856-013-5076-1Search in Google Scholar PubMed

[18] Arda MS, Koçman EA, Özkara E, Söztutar E, Özatik O, Köse A, et al. Can a small intestine segment be an alternative biological conduit for peripheral nerve regeneration? Balk Med J. 2017;34(3):246–54.10.4274/balkanmedj.2015.1601Search in Google Scholar PubMed PubMed Central

[19] Du J, Chen H, Qing L, Yang X, Jia X. Biomimetic neural scaffolds: a crucial step towards optimal peripheral nerve regeneration. Biomater Sci. 2018;6(6):1299–311.10.1039/C8BM00260FSearch in Google Scholar

[20] Nectow AR, Marra KG, Kaplan DL. Biomaterials for the development of peripheral nerve guidance conduits. Tissue Eng Part B Rev. 2012;18(1):40–50.10.1089/ten.teb.2011.0240Search in Google Scholar

[21] Lundborg G, Kanje M. Bioartificial nerve grafts. A prototype. Scand J Plast Reconstr Surg Hand Surg. 1996;30(2):105–10.10.3109/02844319609056391Search in Google Scholar PubMed

[22] Sabongi RG, Fernandes M, Dos Santos JB. Peripheral nerve regeneration with conduits: use of vein tubes. Neural Regen Res. 2015;10(4):529–33.10.4103/1673-5374.155428Search in Google Scholar PubMed PubMed Central

[23] Wang J, Zhu YQ, Wang Y, Xu HG, Xu WJ, Wang YX, et al. A novel tissue engineered nerve graft constructed with autologous vein and nerve microtissue repairs a long-segment sciatic nerve defect. Neural Regen Res. 2021;16(1):143–9.10.4103/1673-5374.286977Search in Google Scholar PubMed PubMed Central

[24] Arino H, Brandt J, Dahlin LB. Implantation of Schwann cells in rat tendon autografts as a model for peripheral nerve repair: long term effects on functional recovery. Scand J Plast Reconstr Surg Hand Surg. 2008;42(6):281–5.10.1080/02844310802393966Search in Google Scholar PubMed

[25] Yang XN, Jin YQ, Bi H, Wei W, Cheng J, Liu ZY, et al. Peripheral nerve repair with epimysium conduit. Biomaterials. 2013;34(22):5606–16.10.1016/j.biomaterials.2013.04.018Search in Google Scholar PubMed

[26] Takeuchi H, Sakamoto A, Ikeguchi R, Ota S, Oda H, Yurie H, et al. Doxorubicin-immersed skeletal muscle grafts promote peripheral nerve regeneration across a 10 mm defect in the rat sciatic nerve. J Reconstr Microsurg. 2020;36(1):41–52.10.1055/s-0039-1694740Search in Google Scholar PubMed

[27] Bozkurt A, Dunda SE, Mon O’Dey D, Brook GA, Suschek CV, Pallua N. Epineurial sheath tube (EST) technique: an experimental peripheral nerve repair model. Neurol Res. 2011;33(10):1010–5.10.1179/1743132811Y.0000000029Search in Google Scholar PubMed

[28] Santos Roballo KC, Dhungana S, Jiang Z, Oakey J, Bushman JS. Localized delivery of immunosuppressive regulatory T cells to peripheral nerve allografts promotes regeneration of branched segmental defects. Biomaterials. 2019;209:1–9.10.1016/j.biomaterials.2019.04.015Search in Google Scholar PubMed PubMed Central

[29] Naghilou A, Pöttschacher L, Millesi F, Mann A, Supper P, Semmler L, et al. Correlating the secondary protein structure of natural spider silk with its guiding properties for Schwann cells. Mater Sci Eng C Mater Biol Appl. 2020;116:111219.10.1016/j.msec.2020.111219Search in Google Scholar PubMed

[30] Liu W, Wang D, Huang J, Wei Y, Xiong J, Zhu W, et al. Low-temperature deposition manufacturing: a novel and promising rapid prototyping technology for the fabrication of tissue-engineered scaffold. Mater Sci Eng C Mater Biol Appl. 2017;70(Pt 2):976–82.10.1016/j.msec.2016.04.014Search in Google Scholar PubMed

[31] Liu Z, Yushan M, Alike Y, Liu Y, Wu S, Ma C, et al. Preparation of multiwall carbon nanotubes embedded electroconductive multi-microchannel scaffolds for neuron growth under electrical stimulation. Biomed Res Int. 2020;2020:4794982.10.1155/2020/4794982Search in Google Scholar PubMed PubMed Central

[32] Peng S, Jin G, Li L, Li K, Srinivasan M, Ramakrishna S, et al. Multi-functional electrospun nanofibres for advances in tissue regeneration, energy conversion & storage, and water treatment. Chem Soc Rev. 2016;45(5):1225–41.10.1039/C5CS00777ASearch in Google Scholar PubMed

[33] Hinüber C, Chwalek K, Pan-Montojo FJ, Nitschke M, Vogel R, Brünig H, et al. Hierarchically structured nerve guidance channels based on poly-3-hydroxybutyrate enhance oriented axonal outgrowth. Acta Biomater. 2014;10(5):2086–95.10.1016/j.actbio.2013.12.053Search in Google Scholar PubMed

[34] Diez-Ahedo R, Mendibil X, Márquez-Posadas MC, Quintana I, González F, Rodríguez FJ, et al. UV-casting on methacrylated PCL for the production of a peripheral nerve implant containing an array of porous aligned microchannels. Polym (Basel). 2020;12(4):971.10.3390/polym12040971Search in Google Scholar PubMed PubMed Central

[35] Liu M, Wang L, Tong X, Dai J, Li G, Zhang P, et al. Antibacterial polymer nanofiber-coated and high elastin protein-expressing BMSCs incorporated polypropylene mesh for accelerating healing of female pelvic floor dysfunction. Nanotechnol Rev. 2020;9(1):670–82.10.1515/ntrev-2020-0052Search in Google Scholar

[36] Abelseth E, Abelseth L, De la Vega L, Beyer ST, Wadsworth SJ, Willerth SM. 3D printing of neural tissues derived from human induced pluripotent stem cells using a fibrin-based bioink. ACS Biomater Sci Eng. 2019;5(1):234–43.10.1021/acsbiomaterials.8b01235Search in Google Scholar PubMed

[37] Neal RA, Tholpady SS, Foley PL, Swami N, Ogle RC, Botchwey EA. Alignment and composition of laminin-polycaprolactone nanofiber blends enhance peripheral nerve regeneration. J Biomed Mater Res A. 2012;100(2):406–23.10.1002/jbm.a.33204Search in Google Scholar PubMed PubMed Central

[38] Kundu B, Rajkhowa R, Kundu SC, Wang X. Silk fibroin biomaterials for tissue regenerations. Adv Drug Deliv Rev. 2013;65(4):457–70.10.1016/j.addr.2012.09.043Search in Google Scholar PubMed

[39] Neubauer VJ, Trossmann VT, Jacobi S, Döbl A, Scheibel T. Aqueous-organic solvent derived recombinant spider silk gels as depots for drugs. Angew Chem Int Ed Engl. 2021;60(21):11847–51.10.1002/anie.202103147Search in Google Scholar PubMed PubMed Central

[40] Khalid A, Bai D, Abraham AN, Jadhav A, Linklater D, Matusica A, et al. Electrospun nanodiamond-silk fibroin membranes: a multifunctional platform for biosensing and wound-healing applications. ACS Appl Mater Interfaces. 2020;12(43):48408–19.10.1021/acsami.0c15612Search in Google Scholar PubMed

[41] Alessandrino A, Chiarini A, Biagiotti M, Dal Prà I, Bassani GA, Vincoli V, et al. Three-layered silk fibroin tubular scaffold for the repair and regeneration of small caliber blood vessels: from design to in vivo pilot tests. Front Bioeng Biotechnol. 2019;7:356.10.3389/fbioe.2019.00356Search in Google Scholar PubMed PubMed Central

[42] Chung YG, Algarrahi K, Franck D, Tu DD, Adam RM, Kaplan DL, et al. The use of bi-layer silk fibroin scaffolds and small intestinal submucosa matrices to support bladder tissue regeneration in a rat model of spinal cord injury. Biomaterials. 2014;35(26):7452–9.10.1016/j.biomaterials.2014.05.044Search in Google Scholar PubMed PubMed Central

[43] Alessandrino A, Fregnan F, Biagiotti M, Muratori L, Bassani GA, Ronchi G, et al. SilkBridge™: a novel biomimetic and biocompatible silk-based nerve conduit. Biomater Sci. 2019;7(10):4112–30.10.1039/C9BM00783KSearch in Google Scholar

[44] Grabska-Zielińska S, Sionkowska A, Reczyńska K, Pamuła E. Physico-chemical characterization and biological tests of collagen/silk fibroin/chitosan scaffolds cross-linked by dialdehyde starch. Polym (Basel). 2020;12(2):372.10.3390/polym12020372Search in Google Scholar PubMed PubMed Central

[45] Buckley CT, Vinardell T, Thorpe SD, Haugh MG, Jones E, McGonagle D, et al. Functional properties of cartilaginous tissues engineered from infrapatellar fat pad-derived mesenchymal stem cells. J Biomech. 2010;43(5):920–6.10.1016/j.jbiomech.2009.11.005Search in Google Scholar PubMed

[46] Wang C, Jia Y, Yang W, Zhang C, Zhang K, Chai Y. Silk fibroin enhances peripheral nerve regeneration by improving vascularization within nerve conduits. J Biomed Mater Res A. 2018;106(7):2070–7.10.1002/jbm.a.36390Search in Google Scholar PubMed

[47] Yang Y, Chen X, Ding F, Zhang P, Liu J, Gu X. Biocompatibility evaluation of silk fibroin with peripheral nerve tissues and cells in vitro. Biomaterials. 2007;28(9):1643–52.10.1016/j.biomaterials.2006.12.004Search in Google Scholar PubMed

[48] Tang X, Ding F, Yang Y, Hu N, Wu H, Gu X. Evaluation on in vitro biocompatibility of silk fibroin-based biomaterials with primarily cultured hippocampal neurons. J Biomed Mater Res A. 2009;91(1):166–74.10.1002/jbm.a.32212Search in Google Scholar PubMed

[49] 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):5526–35.10.1016/j.biomaterials.2007.09.001Search in Google Scholar PubMed

[50] Du J, Liu J, Yao S, Mao H, Peng J, Sun X, et al. Prompt peripheral nerve regeneration induced by a hierarchically aligned fibrin nanofiber hydrogel. Acta Biomater. 2017;55:296–309.10.1016/j.actbio.2017.04.010Search in Google Scholar PubMed

[51] Tang X, Xue C, Wang Y, Ding F, Yang Y, Gu X. Bridging peripheral nerve defects with a tissue engineered nerve graft composed of an in vitro cultured nerve equivalent and a silk fibroin-based scaffold. Biomaterials. 2012;33(15):3860–7.10.1016/j.biomaterials.2012.02.008Search in Google Scholar

[52] Ricard-Blum S. The collagen family. Cold Spring Harb Perspect Biol. 2011;3(1):a004978.10.1101/cshperspect.a004978Search in Google Scholar

[53] Goldberga I, Li R, Duer MJ. Collagen structure-function relationships from solid-state NMR spectroscopy. Acc Chem Res. 2018;51(7):1621–9.10.1021/acs.accounts.8b00092Search in Google Scholar

[54] Wessing B, Lettner S, Zechner W. Guided bone regeneration with collagen membranes and particulate graft materials: a systematic review and meta-analysis. Int J Oral Maxillofac Implant. 2018;33(1):87–100.10.11607/jomi.5461Search in Google Scholar

[55] Schönborn K, Willenborg S, Schulz JN, Imhof T, Eming SA, Quondamatteo F, et al. Role of collagen XII in skin homeostasis and repair. Matrix Biol. 2020;94:57–76.10.1016/j.matbio.2020.08.002Search in Google Scholar

[56] Urciuolo A, Quarta M, Morbidoni V, Gattazzo F, Molon S, Grumati P, et al. Collagen VI regulates satellite cell self-renewal and muscle regeneration. Nat Commun. 2013;4:1964.10.1038/ncomms2964Search in Google Scholar

[57] Otake K, Toriumi T, Ito T, Okuwa Y, Moriguchi K, Tanaka S, et al. Recovery of sensory function after the implantation of oriented-collagen tube into the resected rat sciatic nerve. Regen Ther. 2020;14:48–58.10.1016/j.reth.2019.12.004Search in Google Scholar

[58] Keilhoff G, Stang F, Wolf G, Fansa H. Bio-compatibility of type I/III collagen matrix for peripheral nerve reconstruction. Biomaterials. 2003;24(16):2779–87.10.1016/S0142-9612(03)00084-XSearch in Google Scholar

[59] Alluin O, Wittmann C, Marqueste T, Chabas JF, Garcia S, Lavaut MN, et al. Functional recovery after peripheral nerve injury and implantation of a collagen guide. Biomaterials. 2009;30(3):363–73.10.1016/j.biomaterials.2008.09.043Search in Google Scholar PubMed

[60] Borić M, Vicente FA, Jurković DL, Novak U, Likozar B. Chitin isolation from crustacean waste using a hybrid demineralization/DBD plasma process. Carbohydr Polym. 2020;246:116648.10.1016/j.carbpol.2020.116648Search in Google Scholar PubMed

[61] Zhang XY, Chen YP, Han J, Mo J, Dong PF, Zhuo YH, et al. Biocompatiable silk fibroin/carboxymethyl chitosan/strontium substituted hydroxyapatite/cellulose nanocrystal composite scaffolds for bone tissue engineering. Int J Biol Macromol. 2019;136:1247–57.10.1016/j.ijbiomac.2019.06.172Search in Google Scholar PubMed

[62] Zhao Y, Wang Y, Gong J, Yang L, Niu C, Ni X, et al. Chitosan degradation products facilitate peripheral nerve regeneration by improving macrophage-constructed microenvironments. Biomaterials. 2017;134:64–77.10.1016/j.biomaterials.2017.02.026Search in Google Scholar PubMed

[63] Jiang Z, Song Y, Qiao J, Yang Y, Zhang W, Liu W, et al. Rat sciatic nerve regeneration across a 10 mm defect bridged by a chitin/CM-chitosan artificial nerve graft. Int J Biol Macromol. 2019;129:997–1005.10.1016/j.ijbiomac.2019.02.080Search in Google Scholar PubMed

[64] Kong Y, Tang X, Zhao Y, Chen X, Yao K, Zhang L, et al. Degradable tough chitosan dressing for skin wound recovery. Nanotechnol Rev. 2020;9(1):1576–85.10.1515/ntrev-2020-0105Search in Google Scholar

[65] Wang Y, Zhao Y, Sun C, Hu W, Zhao J, Li G, et al. Chitosan degradation products promote nerve regeneration by stimulating Schwann cell proliferation via miR-27a/FOXO1 axis. Mol Neurobiol. 2016;53(1):28–39.10.1007/s12035-014-8968-2Search in Google Scholar PubMed

[66] Jiang M, Zhuge X, Yang Y, Gu X, Ding F. The promotion of peripheral nerve regeneration by chitooligosaccharides in the rat nerve crush injury model. Neurosci Lett. 2009;454(3):239–43.10.1016/j.neulet.2009.03.042Search in Google Scholar PubMed

[67] Carvalho CR, López-Cebral R, Silva-Correia J, Silva JM, Mano JF, Silva TH, et al. Investigation of cell adhesion in chitosan membranes for peripheral nerve regeneration. Mater Sci Eng C Mater Biol Appl. 2017;71:1122–34.10.1016/j.msec.2016.11.100Search in Google Scholar PubMed

[68] Xiaosong Gu PZ, Wang X, Ding F, Peng L, Cheng H. Dog sciatic nerve gap repaired by artificial tissue nerve graft. Prog Nat Sci. 2003;13(8):578–83.Search in Google Scholar

[69] Yang Y, Gu X, Tan R, Hu W, Wang X, Zhang P, et al. Fabrication and properties of a porous chitin/chitosan conduit for nerve regeneration. Biotechnol Lett. 2004;26(23):1793–7.10.1007/s10529-004-4611-zSearch in Google Scholar PubMed

[70] Shen H, Shen ZL, Zhang PH, Chen NL, Wang YC, Zhang ZF, et al. Ciliary neurotrophic factor-coated polylactic-polyglycolic acid chitosan nerve conduit promotes peripheral nerve regeneration in canine tibial nerve defect repair. J Biomed Mater Res B Appl Biomater. 2010;95(1):161–70.10.1002/jbm.b.31696Search in Google Scholar PubMed

[71] Zhang W, Zhang L, Liu J, Zhang L, Zhang J, Tang P. Repairing sciatic nerve injury with an EPO-loaded nerve conduit and sandwiched-in strategy of transplanting mesenchymal stem cells. Biomaterials. 2017;142:90–100.10.1016/j.biomaterials.2017.06.024Search in Google Scholar PubMed

[72] Kim IS. Augmentation rhinoplasty using silicone implants. Facial Plast Surg Clin North Am. 2018;26(3):285–93.10.1016/j.fsc.2018.03.003Search in Google Scholar PubMed

[73] Francel PC, Francel TJ, Mackinnon SE, Hertl C. Enhancing nerve regeneration across a silicone tube conduit by using interposed short-segment nerve grafts. J Neurosurg. 1997;87(6):887–92.10.3171/jns.1997.87.6.0887Search in Google Scholar PubMed

[74] Kormpakis I, Papalois A, Kinnas P, Zoubos AB, Sioutis I, Dimitriadi A, et al. Silicone tubes with thyroid hormone (T3) and BDNF as an alternative to autografts for bridging neural defects. Injury. 2020;51(12):2879–86.10.1016/j.injury.2020.03.026Search in Google Scholar PubMed

[75] Verdú E, Labrador RO, Rodríguez FJ, Ceballos D, Forés J, Navarro X. Alignment of collagen and laminin-containing gels improve nerve regeneration within silicone tubes. Restor Neurol Neurosci. 2002;20(5):169–79.Search in Google Scholar

[76] Liu S, Li H, Ou Yang J, Peng H, Wu K, Liu Y, et al. Enhanced rat sciatic nerve regeneration through silicon tubes filled with pyrroloquinoline quinone. Microsurgery. 2005;25(4):329–37.10.1002/micr.20126Search in Google Scholar PubMed

[77] Primc G. Recent advances in surface activation of polytetrafluoroethylene (PTFE) by gaseous plasma treatments. Polym (Basel). 2020;12(10):2295.10.3390/polym12102295Search in Google Scholar PubMed PubMed Central

[78] Zetti G, Gatti S, Premoselli P, Quattrini A, Comola M, Marchettini P, et al. Morphological and functional evaluation of peripheral nerve regeneration in the rat using an expanded polytetrafluoroethylene (PTFE) microprosthesis. J Invest Surg. 1991;4(4):437–43.10.3109/08941939109141174Search in Google Scholar PubMed

[79] Labroo P, Hilgart D, Davis B, Lambert C, Sant H, Gale B, et al. Drug-delivering nerve conduit improves regeneration in a critical-sized gap. Biotechnol Bioeng. 2019;116(1):143–54.10.1002/bit.26837Search in Google Scholar PubMed

[80] Schmidt CE, Leach JB. Neural tissue engineering: strategies for repair and regeneration. Annu Rev Biomed Eng. 2003;5:293–347.10.1146/annurev.bioeng.5.011303.120731Search in Google Scholar PubMed

[81] Swainson SME, Styliari ID, Taresco V, Garnett MC. Poly(glycerol adipate) (PGA), an enzymatically synthesized functionalizable polyester and versatile drug delivery carrier: a literature update. Polym (Basel). 2019;11:10.10.3390/polym11101561Search in Google Scholar PubMed PubMed Central

[82] Tabesh H, Amoabediny G, Nik NS, Heydari M, Yosefifard M, Siadat SO, et al. The role of biodegradable engineered scaffolds seeded with Schwann cells for spinal cord regeneration. Neurochem Int. 2009;54(2):73–83.10.1016/j.neuint.2008.11.002Search in Google Scholar PubMed

[83] Fujimaki H, Matsumine H, Osaki H, Ueta Y, Kamei W, Shimizu M, et al. Dedifferentiated fat cells in polyglycolic acid-collagen nerve conduits promote rat facial nerve regeneration. Regen Ther. 2019;11:240–8.10.1016/j.reth.2019.08.004Search in Google Scholar

[84] Şentürk M, Çakır M, Tekin A, Küçükkartallar T, Yıldırım MA, Alkan S, et al. Comparison of primary repair and repair with polyglycolic acid coated tube in recurrent laryngeal nerve cuts (an experimental study). Am J Surg. 2020;219(4):632–6.10.1016/j.amjsurg.2019.06.015Search in Google Scholar

[85] Hsu SH, Ni HC. Fabrication of the microgrooved/microporous polylactide substrates as peripheral nerve conduits and in vivo evaluation. Tissue Eng Part A. 2009;15(6):1381–90.10.1089/ten.tea.2008.0175Search in Google Scholar

[86] Riboldi SA, Tozzi M, Bagardi M, Ravasio G, Cigalino G, Crippa L, et al. A novel hybrid silk fibroin/polyurethane arteriovenous graft for hemodialysis: proof-of-concept animal study in an Ovine model. Adv Healthc Mater. 2020;9(20):e2000794.10.1002/adhm.202000794Search in Google Scholar

[87] Eivazi Zadeh Z, Solouk A, Shafieian M, Haghbin Nazarpak M. Electrospun polyurethane/carbon nanotube composites with different amounts of carbon nanotubes and almost the same fiber diameter for biomedical applications. Mater Sci Eng C Mater Biol Appl. 2021;118:111403.10.1016/j.msec.2020.111403Search in Google Scholar

[88] Wu Y, Wang L, Guo B, Shao Y, Ma PX. Electroactive biodegradable polyurethane significantly enhanced Schwann cells myelin gene expression and neurotrophin secretion for peripheral nerve tissue engineering. Biomaterials. 2016;87:18–31.10.1016/j.biomaterials.2016.02.010Search in Google Scholar

[89] Hsu SH, Chang WC, Yen CT. Novel flexible nerve conduits made of water-based biodegradable polyurethane for peripheral nerve regeneration. J Biomed Mater Res A. 2017;105(5):1383–92.10.1002/jbm.a.36022Search in Google Scholar

[90] Laurent CP, Vaquette C, Liu X, Schmitt JF, Rahouadj R. Suitability of a PLCL fibrous scaffold for soft tissue engineering applications: a combined biological and mechanical characterisation. J Biomater Appl. 2018;32(9):1276–88.10.1177/0885328218757064Search in Google Scholar

[91] Hiljanen-Vainio MP, Orava PA, Seppälä JV. Properties of epsilon-caprolactone/DL-lactide (epsilon-CL/DL-LA) copolymers with a minor epsilon-CL content. J Biomed Mater Res. 1997;34(1):39–46.10.1002/(SICI)1097-4636(199701)34:1<39::AID-JBM6>3.0.CO;2-OSearch in Google Scholar

[92] Prabhakaran MP, Venugopal JR, Ramakrishna S. Mesenchymal stem cell differentiation to neuronal cells on electrospun nanofibrous substrates for nerve tissue engineering. Biomaterials. 2009;30(28):4996–5003.10.1016/j.biomaterials.2009.05.057Search in Google Scholar

[93] Wang J, Zheng W, Chen L, Zhu T, Shen W, Fan C, et al. Enhancement of Schwann cells function using graphene-oxide-modified nanofiber scaffolds for peripheral nerve regeneration. ACS Biomater Sci Eng. 2019;5(5):2444–56.10.1021/acsbiomaterials.8b01564Search in Google Scholar

[94] Dadaci M, Karagülle N, Sönmez E, Dadaci Z, İşci ET, İnce B, et al. Evaluation of the effectiveness of biodegradable electrospun caprolactoneand poly(lactic acid-ε-caprolactone) nerve conduits for peripheral nerveregenerations in a rat sciatic nerve defect model. Turk J Med Sci. 2016;46(2):539–48.10.3906/sag-1412-110Search in Google Scholar

[95] Zhou Y, Shen Q, Lin Y, Xu S, Meng Q. Evaluation of the potential of chimeric spidroins/poly(L-lactic-co-ε-caprolactone) (PLCL) nanofibrous scaffolds for tissue engineering. Mater Sci Eng C Mater Biol Appl. 2020;111:110752.10.1016/j.msec.2020.110752Search in Google Scholar

[96] Donate R, Monzón M, Alemán-Domínguez ME, Ortega Z. Enzymatic degradation study of PLA-based composite scaffolds. Rev Adv Mater Sci. 2020;59(1):170–5.10.1515/rams-2020-0005Search in Google Scholar

[97] Kim YH, Lin MC, Radke CJ. Degradation modeling of poly-l-lactide acid (PLLA) bioresorbable vascular scaffold within a coronary artery. Nanotechnol Rev. 2020;9(1):1217–26.10.1515/ntrev-2020-0093Search in Google Scholar

[98] Peltoniemi H, Ashammakhi N, Kontio R, Waris T, Salo A, Lindqvist C, et al. The use of bioabsorbable osteofixation devices in craniomaxillofacial surgery. Oral Surg Oral Med Oral Pathol Oral Radiol Endod. 2002;94(1):5–14.10.1067/moe.2002.122160Search in Google Scholar

[99] Zhang S, Liang Y, Qian X, Hui D, Sheng K. Pyrolysis kinetics and mechanical properties of poly(lactic acid)/bamboo particle biocomposites: effect of particle size distribution. Nanotechnol Rev. 2020;9(1):524–33.10.1515/ntrev-2020-0037Search in Google Scholar

[100] Evans GR, Brandt K, Niederbichler AD, Chauvin P, Herrman S, Bogle M, et al. Clinical long-term in vivo evaluation of poly(L-lactic acid) porous conduits for peripheral nerve regeneration. J Biomater Sci Polym Ed. 2000;11:869–78.10.1163/156856200744066Search in Google Scholar

[101] 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):841–8.10.1016/S0142-9612(01)00190-9Search in Google Scholar

[102] Gu F, Zhang L, Teply BA, Mann N, Wang A, Radovic-Moreno AF, et al. Precise engineering of targeted nanoparticles by using self-assembled biointegrated block copolymers. Proc Natl Acad Sci U S A. 2008;105(7):2586–91.10.1073/pnas.0711714105Search in Google Scholar PubMed PubMed Central

[103] Park JH, Kang HJ, Kwon DY, Lee BK, Lee B, Jang JW, et al. Biodegradable poly(lactide-co-glycolide-co-ε-caprolactone) block copolymers – evaluation as drug carriers for a localized and sustained delivery system. J Mater Chem B. 2015;3(41):8143–53.10.1039/C5TB01542ASearch in Google Scholar

[104] Swider E, Koshkina O, Tel J, Cruz LJ, de Vries IJM, Srinivas M. Customizing poly(lactic-co-glycolic acid) particles for biomedical applications. Acta Biomater. 2018;73:38–51.10.1016/j.actbio.2018.04.006Search in Google Scholar PubMed

[105] Akolpoğlu Başaran DD, Gündüz U, Tezcaner A, Keskin D. Topical delivery of heparin from PLGA nanoparticles entrapped in nanofibers of sericin/gelatin scaffolds for wound healing. Int J Pharm. 2021;597:120207.10.1016/j.ijpharm.2021.120207Search in Google Scholar PubMed

[106] Xu F, Zhang K, Lv P, Lu R, Zheng L, Zhao J. NECL1 coated PLGA as favorable conduits for repair of injured peripheral nerve. Mater Sci Eng C Mater Biol Appl. 2017;70(Pt 2):1132–40.10.1016/j.msec.2016.03.043Search in Google Scholar PubMed

[107] Zare EN, Jamaledin R, Naserzadeh P, Afjeh-Dana E, Ashtari B, Hosseinzadeh M, et al. Metal-based nanostructures/PLGA nanocomposites: antimicrobial activity, cytotoxicity, and their biomedical applications. ACS Appl Mater Interfaces. 2020;12(3):3279–300.10.1021/acsami.9b19435Search in Google Scholar PubMed

[108] Hou Y, Wang X, Zhang Z, Luo J, Cai Z, Wang Y, et al. Repairing transected peripheral nerve using a biomimetic nerve guidance conduit containing intraluminal sponge fillers. Adv Healthc Mater. 2019;8(21):e1900913.10.1002/adhm.201900913Search in Google Scholar PubMed

[109] Nieto-Suárez M, López-Quintela MA, Lazzari M. Preparation and characterization of crosslinked chitosan/gelatin scaffolds by ice segregation induced self-assembly. Carbohydr Polym. 2016;141:175–83.10.1016/j.carbpol.2015.12.064Search in Google Scholar PubMed

[110] Ouyang Y, Huang C, Zhu Y, Fan C, Ke Q. Fabrication of seamless electrospun collagen/PLGA conduits whose walls comprise highly longitudinal aligned nanofibers for nerve regeneration. J Biomed Nanotechnol. 2013;9(6):931–43.10.1166/jbn.2013.1605Search in Google Scholar PubMed

[111] Wang G, Hu X, Lin W, Dong C, Wu H. Electrospun PLGA-silk fibroin-collagen nanofibrous scaffolds for nerve tissue engineering. In Vitro Cell Dev Biol Anim. 2011;47(3):234–40.10.1007/s11626-010-9381-4Search in Google Scholar PubMed

[112] Lundborg G, Rosén B, Dahlin L, Holmberg J, Rosén I. Tubular repair of the median or ulnar nerve in the human forearm: a 5-year follow-up. J Hand Surg Br. 2004;29(2):100–7.10.1016/j.jhsb.2003.09.018Search in Google Scholar PubMed

[113] Ashley WW Jr, Weatherly T, Park TS. Collagen nerve guides for surgical repair of brachial plexus birth injury. J Neurosurg. 2006;105(6 Suppl):452–6.10.3171/ped.2006.105.6.452Search in Google Scholar

[114] Stanec S, Stanec Z. Reconstruction of upper-extremity peripheral-nerve injuries with ePTFE conduits. J Reconstr Microsurg. 1998;14(4):227–32.10.1142/9789812778208_0029Search in Google Scholar

[115] Pogrel MA, McDonald AR, Kaban LB. Gore-Tex tubing as a conduit for repair of lingual and inferior alveolar nerve continuity defects: a preliminary report. J Oral Maxillofac Surg. 1998;56(3):319–21. discussion 321–2.10.1016/S0278-2391(98)90107-0Search in Google Scholar

[116] Mackinnon SE, Dellon AL. Clinical nerve reconstruction with a bioabsorbable polyglycolic acid tube. Plast Reconstr Surg. 1990;85(3):419–24.10.1097/00006534-199003000-00015Search in Google Scholar PubMed

[117] Seo K, Terumitsu M, Inada Y, Nakamura T, Shigeno K, Tanaka Y. Prognosis after surgical treatment of trigeminal neuropathy with a PGA-c tube: report of 10 cases. Pain Med. 2016;17(12):2360–8.10.1093/pm/pnw088Search in Google Scholar PubMed

[118] Inada Y, Hosoi H, Yamashita A, Morimoto S, Tatsumi H, Notazawa S, et al. Regeneration of peripheral motor nerve gaps with a polyglycolic acid-collagen tube: technical case report. Neurosurgery. 2007;61(5):E1105–7. discussion E1107.10.1227/01.neu.0000303210.45983.97Search in Google Scholar PubMed

[119] Lei M, Chen Z, Lu H, Yu K. Recent progress in shape memory polymer composites: methods, properties, applications and prospects. Nanotechnol Rev. 2019;8(1):327–51.10.1515/ntrev-2019-0031Search in Google Scholar

[120] Zheng L, Cui HF. Use of chitosan conduit combined with bone marrow mesenchymal stem cells for promoting peripheral nerve regeneration. J Mater Sci Mater Med. 2010;21(5):1713–20.10.1007/s10856-010-4003-ySearch in Google Scholar PubMed

[121] Li X, Wang W, Wei G, Wang G, Zhang W, Ma X. Immunophilin FK506 loaded in chitosan guide promotes peripheral nerve regeneration. Biotechnol Lett. 2010;32(9):1333–7.10.1007/s10529-010-0287-8Search in Google Scholar PubMed

[122] Liu H, Wen W, Hu M, Bi W, Chen L, Liu S, et al. Chitosan conduits combined with nerve growth factor microspheres repair facial nerve defects. Neural Regen Res. 2013;8(33):3139–47.Search in Google Scholar

Received: 2021-03-27
Revised: 2021-05-07
Accepted: 2021-05-07
Published Online: 2021-06-21

© 2021 Wufei Dai et al., published by De Gruyter

This work is licensed under the Creative Commons Attribution 4.0 International License.

Downloaded on 25.4.2024 from https://www.degruyter.com/document/doi/10.1515/ntrev-2021-0028/html
Scroll to top button