Peripheral Nerve Regeneration: Opportunities and Challenges

Article Information

Rajiv Supra1, Devendra K. Agrawal2*

1 College of Osteopathic Medicine, Touro University, Henderson, Nevada,

2 Department of Translational Research, College of Osteopathic Medicine of the Pacific,

Pomona, California

*Corresponding Author: Devendra K. Agrawal, MSc, Ph.D. (Biochem), Ph.D. (Med Sci), MBA, MS (ITM), FAAAAI, FAHA, FAPS, FIACS, Professor and Director, Department of Translational Research, Western University of Health Sciences, 309 E. Second Street, Pomona, California 91766-1854, USA.

Received: 02 February 2023; Accepted: 06 February 2023; Published: 08 February 2023

Citation:

Rajiv Supra and Devendra K Agrawal. Peripheral Nerve Regeneration: Opportunities and Challenges. Journal of Surgery and Research 5 (2023): 10-18

View / Download Pdf Share at Facebook

Abstract

Peripheral nerve injury has detrimental effects on the quality of life for patients and is a worldwide issue with high rates of morbidity. Research on the molecular mechanisms of nerve injury, microsurgical techniques, and advances in stem cell research have led to substantial progress in the field of translational neurophysiology. Current research on peripheral nerve regeneration aims to accelerate peripheral nerve development through pluripotent stem cells and potential use of smart exosomes, pharmacological agents, and bioengineering of nerve conduits. In this article critically reviewed and summarized various methods used for peripheral nerve regeneration and highlight the opportunities and challenges that come along with these strategies

Keywords

Allografts; Autografts; Erythropoietin; Exosomes; Nerve conduits; Neuronal injury; Peripheral nerve injury; Peripheral nerve regeneration; Pharmacotherapy; Stem cells; Tacrolimus

articles about Allografts Research articles about Allografts review articles about AllograftsAllografts PubMed articles Allografts PubMed Central articles Allografts 2024 articles Allografts 2025 articles Allografts Scopus articles Allografts impact factor journals Allografts Scopus journals Allografts PubMed journals Allografts medical journals Allografts free journals Allografts best journals Allografts top journals Allografts free medical journals Allografts famous journals Allografts Google Scholar indexed journals articles about Autografts Research articles about Autografts review articles about AutograftsAutografts PubMed articles Autografts PubMed Central articles Autografts 2025 articles Autografts 2026 articles Autografts Scopus articles Autografts impact factor journals Autografts Scopus journals Autografts PubMed journals Autografts medical journals Autografts free journals Autografts best journals Autografts top journals Autografts free medical journals Autografts famous journals Autografts Google Scholar indexed journals articles about Erythropoietin Research articles about Erythropoietin review articles about ErythropoietinErythropoietin PubMed articles Erythropoietin PubMed Central articles Erythropoietin 2026 articles Erythropoietin 2027 articles Erythropoietin Scopus articles Erythropoietin impact factor journals Erythropoietin Scopus journals Erythropoietin PubMed journals Erythropoietin medical journals Erythropoietin free journals Erythropoietin best journals Erythropoietin top journals Erythropoietin free medical journals Erythropoietin famous journals Erythropoietin Google Scholar indexed journals articles about Exosomes Research articles about Exosomes review articles about ExosomesExosomes PubMed articles Exosomes PubMed Central articles Exosomes 2027 articles Exosomes 2028 articles Exosomes Scopus articles Exosomes impact factor journals Exosomes Scopus journals Exosomes PubMed journals Exosomes medical journals Exosomes free journals Exosomes best journals Exosomes top journals Exosomes free medical journals Exosomes famous journals Exosomes Google Scholar indexed journals articles about Nerve conduits Research articles about Nerve conduits review articles about Nerve conduitsNerve conduits PubMed articles Nerve conduits PubMed Central articles Nerve conduits 2028 articles Nerve conduits 2029 articles Nerve conduits Scopus articles Nerve conduits impact factor journals Nerve conduits Scopus journals Nerve conduits PubMed journals Nerve conduits medical journals Nerve conduits free journals Nerve conduits best journals Nerve conduits top journals Nerve conduits free medical journals Nerve conduits famous journals Nerve conduits Google Scholar indexed journals articles about Neuronal injury Research articles about Neuronal injury review articles about Neuronal injuryNeuronal injury PubMed articles Neuronal injury PubMed Central articles Neuronal injury 2029 articles Neuronal injury 2030 articles Neuronal injury Scopus articles Neuronal injury impact factor journals Neuronal injury Scopus journals Neuronal injury PubMed journals Neuronal injury medical journals Neuronal injury free journals Neuronal injury best journals Neuronal injury top journals Neuronal injury free medical journals Neuronal injury famous journals Neuronal injury Google Scholar indexed journals articles about Peripheral nerve injury Research articles about Peripheral nerve injury review articles about Peripheral nerve injuryPeripheral nerve injury PubMed articles Peripheral nerve injury PubMed Central articles Peripheral nerve injury 2030 articles Peripheral nerve injury 2031 articles Peripheral nerve injury Scopus articles Peripheral nerve injury impact factor journals Peripheral nerve injury Scopus journals Peripheral nerve injury PubMed journals Peripheral nerve injury medical journals Peripheral nerve injury free journals Peripheral nerve injury best journals Peripheral nerve injury top journals Peripheral nerve injury free medical journals Peripheral nerve injury famous journals Peripheral nerve injury Google Scholar indexed journals articles about Peripheral nerve regeneration Research articles about Peripheral nerve regeneration review articles about Peripheral nerve regenerationPeripheral nerve regeneration PubMed articles Peripheral nerve regeneration PubMed Central articles Peripheral nerve regeneration 2031 articles Peripheral nerve regeneration 2032 articles Peripheral nerve regeneration Scopus articles Peripheral nerve regeneration impact factor journals Peripheral nerve regeneration Scopus journals Peripheral nerve regeneration PubMed journals Peripheral nerve regeneration medical journals Peripheral nerve regeneration free journals Peripheral nerve regeneration best journals Peripheral nerve regeneration top journals Peripheral nerve regeneration free medical journals Peripheral nerve regeneration famous journals Peripheral nerve regeneration Google Scholar indexed journals articles about Stem cells Research articles about Stem cells review articles about Stem cellsStem cells PubMed articles Stem cells PubMed Central articles Stem cells 2032 articles Stem cells 2033 articles Stem cells Scopus articles Stem cells impact factor journals Stem cells Scopus journals Stem cells PubMed journals Stem cells medical journals Stem cells free journals Stem cells best journals Stem cells top journals Stem cells free medical journals Stem cells famous journals Stem cells Google Scholar indexed journals

Article Details

Introduction

Peripheral nerve injury is a clinical problem with detrimental consequences that results in roughly 150 billion dollars in annual costs for injuries to the median and ulnar nerve; about 87% of these costs being from lost production [1]. Etiologies of peripheral nerve injuries include vibration injuries, ischemia, penetrating trauma, and electrocution [2]. In a civilian setting, motor vehicle accidents, lacerations by sharp instruments, or bone fractures are the most common mechanisms of injury in peripheral nerve damage [3]. Explosives and injuries from gunshots are the main reasons for peripheral nerve injury in a warfare setting [4]. Although patients are treated for peripheral nerve damage, many do not go on to full recovery resulting in loss of sensory and motor function [5].

Peripheral nerve surgery has been a major modality of treatment in recent years. Refinements in microsurgical techniques has improved clinical management in patients with peripheral nerve injury. A deeper understanding of the molecular basis of neuron growth has allowed for improved functional outcomes [6,7]. Direct end to end microsurgical epineural nerve repair has been the gold standard for treating peripheral nerve damage in settings with good blood supply and soft tissue [8]. Microsurgical repair requires clean-cut nerve transection with minimal neuron loss and mobilization of distal and proximal stumps [6]. When primary surgical intervention does not allow for repair of peripheral nerves, autogenous nerve grafting, artificial nerve conduits, pharmacologic agents, and stem cell therapies have shown potential in regenerating peripheral nerves [9]. This article aims to provide a critical overview of current research on peripheral nerve regeneration and future perspectives.

Nerve Repair with Grafts

Significant gaps between nerves of greater than 2 cm makes primary end to end nerve repair obsolete. Such gaps present after considerable damage for example from gunshot wounds [4]. In cases of severe damage and enlarged gaps, nerve grafting has become the treatment of choice. This process involved harvesting a portion of a nerve and suturing it between proximal and distal stumps at the site of injury. Nerve stumps are first trimmed of excess scar tissue in which motor and sensory fascicles are then properly aligned [7]. The nerve graft is then placed between the gap with some redundancy in the repair in case the nerve is in proximity with flexion of a joint. Autografts and allografts are the methods used for nerve grafting [6]. Schwann cells (SCs) are provided by autografts and are essential for axon regeneration. Factors such as size of nerve gap, location of nerve repair, and donor-site morbidity are considered and can affect the choice of autogenous nerve grafting [10,11]. Grafts are sutured to the epineurium of nerves depending on the nerve type and location. Nerve autografts are harvested from sensory nerve sites that are expendable. The sural nerve, for example, is a major graft used for peripheral nerve repair. It enables roughly 40 cm of graft to be taken from each leg. Alternative sites include lateral and medial cutaneous nerves of the forearm, superficial and deep peroneal nerves, posterior and lateral cutaneous nerves from the thigh, and dorsal cutaneous branch of the ulnar nerve [12]. For upper extremity reconstruction, Ray et al. suggest using the anterior branch of the medial antebrachial cutaneous nerve [13]. Using nerve autografts involves sacrificing a functioning nerve that is otherwise expendable [14]. The nerve that is harvested for autografting undergoes Wallerian degeneration and provides mechanical structure for axons to grow [15]. Additionally, within the repair site, there are unavoidable issues such as fascicle mismatch, fibrosis of sutures, and damage to the nerve from tissue handling. All these factors in combination lead to poorer outcomes for patients [16].

Nerve allografts are also used as an alternative but require immunosuppression. Techniques used to reduce allograft antigenicity include lyophilization, irradiation, and cold preservation [17]. At approximately 24 months after nerve repair, migration of SCs occurs into the nerve allograft at which point systemic use of immunosuppressive agents can be withdrawn [6]. Nerve allografts go through a process of decellularization, a technique that degrades enzymes, resulting in a nerve scaffold that is acellular. The advantage of this technique compared with nerve conduits in the internal structure maintains an environment ideal for axonal regeneration and repair [9]. Recent research currently supports the use of decellularized nerve allografts for gaps up to 30 mm in length [18].

Pharmacotherapy

Currently, no pharmacological agents have been used in a clinical setting, however many recent studies suggest pharmacotherapy as a potential strategy to accelerate nerve regeneration [19]. Potential pharmacologic agents for peripheral nerve regeneration are summarized in Table 1.

Erythropoietin (EPO), a cytokine that controls red blood cell production is often used in the settings of renal failure [20]. EPO, in the nervous system, has been investigated to prevent neuron apoptosis [21]. The mechanism of this, however, is yet to be understood. EPO is thought to be produced by perineuronal SCs in the peripheral nervous system and from increased expression of calcitonin gene-related peptide (CGRP), PI3K/Akt, JAK/STAT-3, and NF-kB signaling pathways get activated which promotes neuron survival [22]. Since EPO levels have been shown to decline after injury, EPO may be insufficient to provide continual neuroprotection. Studies reveal, however, that even after a week of post sciatic nerve injury, EPO promoted motor function improvement in murine models of peripheral nerve injury [23]. This reinforces the theory that prolonged administration of EPO may be required for peripheral nerve healing [24]. Additionally, EPO has been shown to have anti-apoptotic and anti-inflammatory effects in murine models of several conditions in the central nervous system. Its utility is being tested in phase 2 and phase 3 clinical trials for traumatic brain injury and spinal cord injuries [25].

Tacrolimus (FK506) is another promising pharmacological agent that can be used for peripheral nerve injuries. Tacrolimus is an immunosuppressant that acts through the inhibition of calcineurin [26]. It is currently being used for organ rejection treatments and has effects on nerve regeneration through the binding of FK506 to heat shock protein 90 (Hsp90) and protein 52 (FKBP-52) and activating the ERK pathway [27]. In murine models with nerve transection, short term administration of FK506 resulted in accelerated rates of nerve regeneration and improved functionality [28]. In rats with delayed nerve repair, FK506 reversed the effects of chronic axotomy on nerve regeneration [29]. Although FK506 has been shown to increase Wallerian degeneration through increasing the proliferation of SCs, the long-term effects are not beneficial because of extensive atrophy in SCs [30]. Due to the systemic side effects and immunosuppression due to FK506, it is difficult to justify its use in peripheral nerve repair. However, when FK506 was administered in a vein graft across a sciatic nerve gap, motor functionality increased as well as axon outgrowth [31]. In future studies, dose response studies need to be considered to minimize immunosuppression and maximize axon regeneration.

N-acetyl cysteine (NAC) is known for its antidote effects against acetaminophen toxicity as well as mucolytic effects [32]. Its mechanism of action includes donating cysteine in the synthesis of glutathione, and it can prevent apoptotic signaling in neuronal cells through the RAS-ERK pathway [33]. Bcl-2 is upregulated by NAC and the mRNA of pro-apoptotic proteins, such as caspase-3 and Bax, are downregulated [34]. In a murine model of sciatic and sural nerve transections, NAC administration resulted in increased survival of neurons [35]. Motor neurons have also been shown to be protected by NAC following rhizotomy and ventral root avulsion [36]. NAC also provided optimal nerve protection against cadmium-induced neurotoxicity [37]. Motor neurons following NAC administration, however, do not seem to respond similarly as peripheral nerves. Following sciatic nerve transection in murine models, no growth promoting effects was noticed on motor neurons in their growth into the distal nerve stump [38]. Since NAC is already used clinically, its clinical use for peripheral nerve regeneration is feasible. The neuroprotective effects NAC on humans is worth noting and the inability to promote axon regeneration in motor neurons is a subject that is yet to be determined through further research in experimental and clinically relevant animal models.

Geldanamycin, an antibiotic that was used as a chemotherapeutic agent, has also shown promising data as a neuroprotective agent. Geldanamycin binds strongly to Hsp90 similarly to tacrolimus [39]. In a murine saphenous nerve crush model, Geldanamycin was shown to accelerate the rate of axon regeneration and promoted earlier functional recovery in motor neurons after tibial nerve crush injury [40]. However, the poor water solubility and hepatotoxic side effects of Geldanamycin have made it difficult to use in human subjects [41]. Newer analogues of the drug are yet to be investigated.

Pharmacologic Agent

Mechanism of Action

Effect

Reference

 

Erythropoietin

-Activates NFkB, JAK/STAT, and PI3K/Akt pathways

-Increases rate of axon regeneration

20,21,22

 

-Increases calcitonin gene-related peptide

-Increases axon sprouting Neuroprotection

 
 

-Increases axon density and improves motor axon growth

 

Tacrolimus

-Binds Hsp-90

-Increases myelinated axons

26,27,28

 

-Calcineurin Inhibition

-Neuroprotection

 

-Induces proliferation of Schwann cells

-Increases axon regeneration

 

N-acetylcysteine

-Decreases apoptotic signaling

-Reduces rates of neuronal death

32,33,35

 

-Upregulates Bcl-2 mRNA

-Increases sensory nerve regeneration

 

-Downregulates Bax and caspase 3

   

Geldanamycin

-Binds Hsp-90

-Increases rate of axon regeneration

39,40,41

 
 

Table 1: Potential pharmacologic agents for peripheral nerve regeneration

Stem Cell Therapy

The effect of stem cell therapy with or without in vitro incorporation into a biomaterial-based scaffold has been well investigated by many researchers. The tissue and nerve graft are sutured to the site of injury to bridge the nerve defect with local administration of stem cells. Following nerve graft transplant, stem cells differentiate into SCs and promote axon growth. Many different sources for stem cells have been applied and studied in nerve tissue engineering. For example, embryonic stem cells can differentiate into all embryonic germ layers and create all types of tissues in the body except for fetal cells. Neurospheres from human embryonic stem cells can differentiate into cells with similar features as SCs [42]. Embryonic stem cells that differentiate into SCs express SC markers, S100 and p75, and induce myelination of neurons in the dorsal root ganglion [43]. Microinjection of murine embryonic stem cell-derived neural progenitor cells into epineurium of transected sciatic nerves in rates led to substantial functional and morphological recovery. Injected stem cells can differentiate into SC-like cells months after they are transplanted [44]. Although embryonic stem cells have shown promising results in murine models, they have been shown to induce formation of teratomas [45,46]. Moreover, the use of embryonic stem cells has been known to trigger ethical controversy.

Mesenchymal stem cells are multipotent and can be found in many tissues of the body. Specifically, bone marrow mesenchymal stem cells (BMMSCs) can be retrieved from bone marrow aspiration and can be cultured. Cultured BMMSCs lack immune recognition and be transplanted to hosts without rejection, thus making it one of the most widely used sources for nerve repair [47,48]. BMMSCs can differentiate into SC-like cells, increasing neurite outgrowth [49]. Studies show when seeding BMMSCs into silk fibroin-based nerve conduits, the levels of S100, a SC marker, increased and subsequently elevated many growth factors which ultimately accelerated the recovery in sciatic nerve injury murine models [50]. Compared to the plain nerve graft, the acellular graft with BMMSCs exhibited faster rates of axon growth, and walking track when a 10 mm sciatic nerve defect was bridged in murine models [50]. Bone marrow stem cells have also been shown to increase angiogenesis through elevating levels of vascular endothelial growth factor [51]. Moreover, BMMSCs secrete neurotrophic factors like extracellular matrix proteins, collagen, fibronectin, and glial cell line-derived neurotrophic factor. When a silicone tube containing BMMSCs was transplanted in murine sciatic nerve gap models, rat walking behavior improved, muscle atrophy reduced, and axon regeneration was stimulated [52]. BMMSCs have also been shown to treat peripheral nerve injuries in primate. SCs induced by BMMSCs was filled into a nerve conduit with a collagen sponge to bridge the 20 mm median nerve injury in cynomolgus primates. When cells were transplanted, the growth of axons were observed without massive cell proliferation [53]. Additionally, another study revealed a nerve graft engineered with chitosan and BMMSCs resulted in better repair to a 50 mm median nerve injury than plain nerve conduits in primate models [54].

Studies using adipose stem cells (ASCs) have also shown promising data in nerve regeneration, although use has been limited to its ethical controversy. ASCs can be retrieved through less invasive methods and can exhibit greater clinical potential [55]. ASCs can differentiate into spindle shaped cells that can express SC markers and can stimulate the formation of myelin sheaths [56,57]. Biochemical studies using ASCs showed differentiation into SC phenotypes and improvement of mean amplitudes of muscle action potential and axon diameter after transplantation to bridge a 1 cm murine sciatic nerve gap. Fibrin conduits seeded with ASCs seemed to have better regenerative effects than primary SC-seeded fibrin conduits [58,59]. ASCs were also incorporated into silicone nerve conduits with a type I collagen gel to bridge a 7 mm defect of murine facial nerve defects. Results revealed increased myelinated fibers, myelin thickness, and an over functional improvement in the facial palsy [55]. Of note is the donor age and harvest site of ASCs that can greatly limit the growth properties of ASCs [60,61]. To maximize the regenerative potential of ASCs, the quality of ASC must be considered.

Although the stem cell therapy could have beneficial effects on peripheral nerve regeneration, number of potential drawbacks might limit their use. These drawbacks include low rate of cell survival and graft cell death, immune-mediated rejection, attenuated regenerative capacity of engrafted cells, tumorigenesis, and ethical and regulatory issues [62]. In recent years, the use of stem cell-derived or Schwann cell-derived exosomes, a subclass of extracellular vesicles, have shown some promise to mediate intercellular communication in tissue systems [63-65]. Cell-to-cell contact, and secreted signals (secretome) are critical in axon-glia interaction. Indeed, exosomes carry several constitutive molecules and cargo molecules, including proteins such as growth factors, mediators of gene expression including the transcription factors and cytokines capable of eliciting paracrine biological responses, lipids, and various genetic materials such as mRNA, miRNA, and traces of DNA [63-67]. However, many of the cargo materials in exosome could have off target effects. Therefore, it is critical to define an ideal cell type to derive exosomes and prepare “smart exosomes” by packing them with nerve regenerative mediators to increase myelin-related and neurogenic genes, enhance neurite outgrowth, induce migration and proliferation of Schwann cells, and decrease proinflammatory cytokines [68]. Additionally, the smart exosomes could be delivered using an effective delivery system such as minimally invasive intelligent hydrogels in peripheral nerve regeneration [69]. However, well designed, and controlled studies using these novel strategies are limited and warrant further investigation.

Artificial Nerve Conduits

Although axon regeneration has been reported in gaps less than 5 mm, the functional recovery is poor without proper intervention [70]. In case of gaps less than 1 cm between the nerve injury, direct end to end suturing without tension will allow for proper regeneration. However, for defects greater than 3 cm, bringing the nerve ends together without creating substantial amount of tension is impossible. To overcome this limitation, nerve tissue transplant has been researched to region nerve segments [71]. The disadvantage of nerve grafts includes increased surgical complications, diameter mismatch between nerve and graft, and immunological reactions [72]. A nerve conduit has provided an alternative strategy to bridge the defective nerves and promote regeneration (Table 2).

Nerve conduits have been designed using nondegradable synthetic materials providing a safer option than donor grafts and eliminating the need of any immunosuppressant therapy [73]. Following implant of the surgical conduit, axons can regenerate from the proximal to distal stump [74]. Nerve conduits are designed to enhance peripheral nerve reconstruction and axon guidance as well as preventing formation of fibrous scar tissues [73,75]. Ideal nerve conduits have low immunogenicity, are bio-absorbable, and have substantial mechanical properties to maintain the alignment between proximal and distal nerve sites [76-78]. Different biomaterials have been studied and have been classified as natural polymers and synthetic materials [77,79].

Polyglycolic acid (PGA) is an FDA approved synthetic polymer used for nerve conduits. Using PGA allowed for more flexible and porous material which enhanced the nerve regeneration process [70,80]. The use of PGA nerve conduits allowed for better reconstruction of long nerve gaps compared to other synthetic polymer materials. Additionally, gaps less than 4 mm led to greater repair outcomes compared to end-to-end regeneration [81,82]. Polylactic acid (PLA) enhances neuron regeneration and is made from biodegradable polyester. PLA nerve conduits can support neuron proliferation and has been shown to improve maturation of axons [70]. PLA conduits, however, generate lactic acid as a byproduct during its degradation which can result in neurotoxicity. Further studies are needed to elucidate its efficacy in peripheral nerve regeneration [83,84].

Poly(L-lactide-co-ε-caprolactone) (PLCL) contains lactic acid. A transparent PLCL conduit can theoretically produce less lactic acid during degradation rendering it a more viable option considering its reduced toxicity. The efficacy of this conduit has been assessed with mixed outcomes. Studies show no promising effects of PLCL when promoting nerve repair in digital nerves, however PLCL has had comparable results in end-to-end repair. These conflicting findings potentially can be explained through the location of the nerve injury. The efficacy of PLCL remains in controversy and further studies are needed on this nerve conduit [85-87].

In addition to synthetic nerve conduits, natural polymers have also been used providing cell growth and support. Collagen and chitosan have been extensively used in nerve conduit fabrication and have minimal biodegradability and immunogenicity. Type I collagen has been the most widely used material and constructs fibrils of the endoneurium in the basal lamina. Collagen conduits have the ability to bridge nerve gaps up to 20 mm and improve functional outcomes in nerve reconstruction. Studies have also revealed reduced symptoms of pain [88-90]. Chitosan, another natural polymer, has also been researched for its ability to increase neurite growth and nerve cell adhesion [91-93]. Gu et al. revealed chitosan fibers promoting migration, adhesion, and proliferation of SCs and axon growth. A case study revealed chitosan repairing a 30 mm defect of the median nerve which resulted in restoration of motor function of thumb abduction [94]. Additionally, in murine models of nerve repair, chitosan has been shown to induce nerve repair in 1 cm gaps between nerves [94].

Material

Type

Characteristics

Application

Reference

Polyglycolic Acid

Synthetic

-Stable

-Nerve conduits

71,81,82

-Biodegradable

-Nerve allografts

Polylactic Acid

Synthetic

-Biocompatible

-Nanoparticles

83,84

-Easy to fabricate

-Nerve conduits

PLCL

Synthetic

-Low immunogenicity

-Nerve conduits

85,86,87

Chitosan

Natural

-Weak Degradability

-Nerve conduits

91,92,93

Collagen

Natural

-Degradable

-Nerve conduits

88,89,90

-Biocompatible

-Membrane

Table 2: Characteristics of materials for nerve conduits and their applications

Conclusion

This review summarized the current research on peripheral nerve repair to highlight current and developing perspectives in the growing field of nerve regeneration. As discussed, there are many opportunities in the use of potential pharmacologic agents for peripheral nerve regeneration, stem cell therapy, and artificial nerve conduits. Potential use of “smart exosomes” with minimally invasive intelligent hydrogels is promising in the regeneration of peripheral nerve. However, these strategies do not come without challenges and warrant careful investigation. Together with a greater understanding of the biology underlying nerve regeneration coupled with latest advances in biotechnology can result into promising breakthroughs in the field of nerve regeneration.

Author Contributions:

Concept and design: RS, DKA; Literature Search: RS, DKA; Critical review and interpretation of the findings: RS, DKA; Drafting the article: RS; Revising and editing the manuscript: RS, DKA; Final approval of the article: RS, DKA.

Funding:

The research work of DKA is supported by the research grants R01 HL144125 and R01HL147662 from the National Heart, Lung, and Blood Institute, National Institutes of Health, USA. The contents of this article are solely the responsibility of the authors and do not necessarily represent the official views of the National Institutes of Health.

Institutional Review Board Statement:

Not applicable.

Informed Consent Statement:

Not applicable

Data Availability Statement:

Not applicable since the information is gathered from published articles.

Acknowledgments:

None

Conflicts of Interest:

The authors declare no conflict of interest.

References

  1. Taylor CA, Braza D, Rice JB, Dillingham T. The Incidence of Peripheral Nerve Injury in Extremity Trauma. Am J Phys Med Rehabil. 2008 May;87(5):381-385.
  2. Robinson LR. Chapter 19 Traumatic injury to peripheral nerves. In 2004. p. 173-86.
  3. Siemionow M, Brzezicki G. Chapter 8 Current Techniques and Concepts in Peripheral Nerve Repair. In 2009. p. 141-172.
  4. Birch R, Misra P, Stewart MPM, Eardley WGP, Ramasamy A, Brown K, et al. Nerve injuries sustained during warfare. J Bone Joint Surg Br. 2012 Apr;94-B(4):523-528.
  5. Dahlin LB, Wiberg M. Nerve injuries of the upper extremity and hand. EFORT Open Rev. 2017 May;2(5):158-170.
  6. Ray WZ, Mackinnon SE. Management of nerve gaps: Autografts, allografts, nerve transfers, and end-to-side neurorrhaphy. Exp Neurol. 2010 May;223(1):77-85.
  7. Kline DG. Nerve Surgery as It Is Now and as It May Be. Neurosurgery. 2000 Jun;46(6):1285.
  8. Griffin JW, Hogan M v., Chhabra AB, Deal DN. Peripheral Nerve Repair and Reconstruction. J Bone Joint Surg. 2013 Dec;95(23):2144-2151.
  9. Huang YC, Huang YY. Biomaterials and Strategies for Nerve Regeneration. Artif Organs. 2006 Jul;30(7):514-522.
  10. Terzis JK, Kostopoulos VK. Vascularized nerve grafts and vascularized fascia for upper extremity nerve reconstruction. Hand (N Y). 2010 Mar;5(1):19-30.
  11. Colen KL, Choi M, Chiu DTW. Nerve Grafts and Conduits. Plast Reconstr Surg. 2009 Dec;124:e386-394.
  12. Norkus T, Norkus M, Ramanauskas T. Donor, recipient and nerve grafts in brachial plexus reconstruction: anatomical and technical features for facilitating the exposure. Surgical and Radiologic Anatomy. 2005 Dec 25;27(6):524-530.
  13. Ross D, Mackinnon S, Chang YL. Intraneural Anatomy of the Median Nerve Provides “Third Web Space” Donor Nerve Graft. J Reconstr Microsurg. 1992 May 8;8(03):225-232.
  14. Mackinnon SE, Doolabh VB, Novak CB, Trulock EP. Clinical Outcome following Nerve Allograft Transplantation. Plast Reconstr Surg. 2001 May;107(6):1419-1429.
  15. MILLESI H. Peripheral Nerve Surgery Today: Turning Point or Continuous Development? Journal of Hand Surgery. 1990 Jun 17;15(3):281-287.
  16. Moore AM, Ray WZ, Chenard KE, Tung T, Mackinnon SE. Nerve Allotransplantation as it Pertains to Composite Tissue Transplantation. HAND. 2009 Sep 21;4(3):239-244.
  17. Evans PJ, Mackinnon SE, Best TJ, Wade JA, Awerbuck DC, Makino AP, et al. Regeneration across preserved peripheral nerve grafts. Muscle Nerve. 1995 Oct;18(10):1128-1138.
  18. Sachanandani NF, Pothula A, Tung TH. Nerve Gaps. Plast Reconstr Surg. 2014 Feb;133(2):313-319.
  19. Chan KM, Gordon T, Zochodne DW, Power HA. Improving peripheral nerve regeneration: From molecular mechanisms to potential therapeutic targets. Exp Neurol. 2014 Nov;261:826-835.
  20. Singh AK, Szczech L, Tang KL, Barnhart H, Sapp S, Wolfson M, et al. Correction of Anemia with Epoetin Alfa in Chronic Kidney Disease. New England Journal of Medicine. 2006 Nov 16;355(20):2085-2098.
  21. HÖKE A, KESWANI SC. Neuroprotection in the PNS: Erythropoietin and Immunophilin Ligands. Ann N Y Acad Sci. 2008 Jun 28;1053(1):491-501.
  22. Hoke A. Schwann Cells Express Motor and Sensory Phenotypes That Regulate Axon Regeneration. Journal of Neuroscience. 2006 Sep 20;26(38):9646-9655.
  23. 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-American Volume. 2008 Aug;90(8):1644-1653.
  24. Modrak M, Sundem L, Elfar J. Erythropoietin enhanced recovery after peripheral nerve injury. Neural Regen Res. 2017;12(8):1268.
  25. Sirén AL, Faßhauer T, Bartels C, Ehrenreich H. Therapeutic potential of erythropoietin and its structural or functional variants in the nervous system. Neurotherapeutics. 2009 Jan;6(1):108-127.
  26. Starzl ThomasE, Fung J, Venkataramman R, Todo S, Demetris AnthonyJ, Jain A. FK 506 FOR LIVER, KIDNEY, AND PANCREAS TRANSPLANTATION. The Lancet. 1989 Oct;334(8670):1000-1004.
  27. Gold BG, Zhong YP. FK506 Requires Stimulation of the Extracellular Signal-Regulated Kinase 1/2 and the Steroid Receptor Chaperone Protein p23 for Neurite Elongation. Neurosignals. 2004;13(3):122-129.
  28. Yan Y, Sun HH, Hunter DA, Mackinnon SE, Johnson PJ. Efficacy of Short-Term FK506 Administration on Accelerating Nerve Regeneration. Neurorehabil Neural Repair. 2012 Jul 30;26(6):570-580.
  29. Sulaiman OAR, Voda J, Gold BG, Gordon T. FK506 Increases Peripheral Nerve Regeneration after Chronic Axotomy but Not after Chronic Schwann Cell Denervation. Exp Neurol. 2002 May;175(1):127-137.
  30. FANSA H, KEILHOFF G, ALTMANN S, PLOGMEIER K, WOLF G, SCHNEIDER W. The Effect of the Immunosuppressant FK 506 on Peripheral Nerve Regeneration Following Nerve Grafting. Journal of Hand Surgery. 1999 Feb 7;24(1):38-42.
  31. Azizi S, Mohammadi R, Amini K, Fallah R. Effects of topically administered FK506 on sciatic nerve regeneration and reinnervation after vein graft repair of short nerve gaps. Neurosurg Focus. 2012 May;32(5):E5.
  32. Marenzi G, Assanelli E, Marana I, Lauri G, Campodonico J, Grazi M, et al. N -Acetylcysteine and Contrast-Induced Nephropathy in Primary Angioplasty. New England Journal of Medicine. 2006 Jun 29;354(26):2773-2782.
  33. Yan CYI, Greene LA. Prevention of PC12 Cell Death by N -Acetylcysteine Requires Activation of the Ras Pathway. The Journal of Neuroscience. 1998 Jun 1;18(11):4042-4049.
  34. Reid AJ, Shawcross SG, Hamilton AE, Wiberg M, Terenghi G. N-Acetylcysteine alters apoptotic gene expression in axotomised primary sensory afferent subpopulations. Neurosci Res. 2009 Oct;65(2):148-155.
  35. Hart AM, Wiberg M, Youle M, Terenghi G. Systemic acetyl-l-carnitine eliminates sensory neuronal loss after peripheral axotomy: a new clinical approach in the management of peripheral nerve trauma. Exp Brain Res. 2002 Jul 4;145(2):182-189.
  36. Zhang CG, Welin D, Novikov L, Kellerth JO, Wiberg M, Hart AM. Motorneuron protection by N-acetyl-cysteine after ventral root avulsion and ventral rhizotomy. Br J Plast Surg. 2005 Sep;58(6):765-773.
  37. Moschou M, Kosmidis EK, Kaloyianni M, Geronikaki A, Dabarakis N, Theophilidis G. In vitro assessment of the neurotoxic and neuroprotective effects of N-acetyl-l-cysteine (NAC) on the rat sciatic nerve fibers. Toxicology in Vitro. 2008 Feb;22(1):267-274.
  38. Welin D, Novikova LN, Wiberg M, Kellerth JO, Novikov LN. Effects of N-acetyl-cysteine on the survival and regeneration of sural sensory neurons in adult rats. Brain Res. 2009 Sep;1287:58-66.
  39. Bucci M, Roviezzo F, Cicala C, Sessa WC, Cirino G. Geldanamycin, an inhibitor of heat shock protein 90 (Hsp90) mediated signal transduction has anti-inflammatory effects and interacts with glucocorticoid receptor in vivo. Br J Pharmacol. 2000 Sep;131(1):13-16.
  40. Sun HH, Saheb-Al-Zamani M, Yan Y, Hunter DA, Mackinnon SE, Johnson PJ. Geldanamycin accelerated peripheral nerve regeneration in comparison to FK-506 in vivo. Neuroscience. 2012 Oct;223:114-123.
  41. Neckers L, Workman P. Hsp90 Molecular Chaperone Inhibitors: Are We There Yet? Clinical Cancer Research. 2012 Jan 1;18(1):64-76.
  42. Ziegler L, Grigoryan S, Yang IH, Thakor N v., Goldstein RS. Efficient Generation of Schwann Cells from Human Embryonic Stem Cell-Derived Neurospheres. Stem Cell Rev Rep. 2011 Jun 30;7(2):394-403.
  43. Liu Q, Spusta SC, Mi R, Lassiter RNT, Stark MR, Höke A, et al. Human Neural Crest Stem Cells Derived from Human ESCs and Induced Pluripotent Stem Cells: Induction, Maintenance, and Differentiation into Functional Schwann Cells. Stem Cells Transl Med. 2012 Apr 1;1(4):266-278.
  44. Cui L, Jiang J, Wei L, Zhou X, Fraser JL, Snider BJ, et al. Transplantation of Embryonic Stem Cells Improves Nerve Repair and Functional Recovery After Severe Sciatic Nerve Axotomy in Rats. Stem Cells. 2008 May 1;26(5):1356-1365.
  45. Rippon HJ, Bishop AE. Embryonic stem cells. Cell Prolif. 2004 Feb;37(1):23-34.
  46. Lee G, Chambers SM, Tomishima MJ, Studer L. Derivation of neural crest cells from human pluripotent stem cells. Nat Protoc. 2010 Apr 18;5(4):688-701.
  47. Han Y, Li X, Zhang Y, Han Y, Chang F, Ding J. Mesenchymal Stem Cells for Regenerative Medicine. Cells. 2019 Aug 13;8(8):886.
  48. Aggarwal S, Pittenger MF. Human mesenchymal stem cells modulate allogeneic immune cell responses. Blood. 2005 Feb 15;105(4):1815-1822.
  49. Caddick J, Kingham PJ, Gardiner NJ, Wiberg M, Terenghi G. Phenotypic and functional characteristics of mesenchymal stem cells differentiated along a Schwann cell lineage. Glia. 2006 Dec;54(8):840-849.
  50. Yang Y, Yuan X, Ding F, Yao D, Gu Y, Liu J, et al. Repair of Rat Sciatic Nerve Gap by a Silk Fibroin-Based Scaffold Added with Bone Marrow Mesenchymal Stem Cells. Tissue Eng Part A. 2011 Sep;17(17-18):2231-2244.
  51. Fan L, Yu Z, Li J, Dang X, Wang K. Schwann-like cells seeded in acellular nerve grafts improve nerve regeneration. BMC Musculoskelet Disord. 2014 Dec 21;15(1):165.
  52. Chen CJ, Ou YC, Liao SL, Chen WY, Chen SY, Wu CW, et al. Transplantation of bone marrow stromal cells for peripheral nerve repair. Exp Neurol. 2007 Mar;204(1):443-453.
  53. Wakao S, Hayashi T, Kitada M, Kohama M, Matsue D, Teramoto N, et al. Long-term observation of auto-cell transplantation in non-human primate reveals safety and efficiency of bone marrow stromal cell-derived Schwann cells in peripheral nerve regeneration. Exp Neurol. 2010 Jun;223(2):537-547.
  54. Hu N, Wu H, Xue C, Gong Y, Wu J, Xiao Z, et al. Long-term outcome of the repair of 50 mm long median nerve defects in rhesus monkeys with marrow mesenchymal stem cells-containing, chitosan-based tissue engineered nerve grafts. Biomaterials. 2013 Jan;34(1):100-111.
  55. Watanabe Y, Sasaki R, Matsumine H, Yamato M, Okano T. Undifferentiated and differentiated adipose-derived stem cells improve nerve regeneration in a rat model of facial nerve defect. J Tissue Eng Regen Med. 2017 Feb;11(2):362-374.
  56. Xie S, Lu F, Han J, Tao K, Wang H, Simental A, et al. Efficient generation of functional Schwann cells from adipose-derived stem cells in defined conditions. Cell Cycle. 2017 May 3;16(9):841-851.
  57. Tomita K, Madura T, Sakai Y, Yano K, Terenghi G, Hosokawa K. Glial differentiation of human adipose-derived stem cells: Implications for cell-based transplantation therapy. Neuroscience. 2013 Apr;236:55-65.
  58. di Summa PG, Kalbermatten DF, Pralong E, Raffoul W, Kingham PJ, Terenghi G. Long-term in vivo regeneration of peripheral nerves through bioengineered nerve grafts. Neuroscience. 2011 May;181:278-291.
  59. Kingham PJ, Kalbermatten DF, Mahay D, Armstrong SJ, Wiberg M, Terenghi G. Adipose-derived stem cells differentiate into a Schwann cell phenotype and promote neurite outgrowth in vitro. Exp Neurol. 2007 Oct;207(2):267-274.
  60. Tremp M, Schwabedissen MMZ, Kappos EA, Engels PE, Fischmann A, Scherberich A, et al. The Regeneration Potential after Human and Autologous Stem Cell Transplantation in a Rat Sciatic Nerve Injury Model can be Monitored by MRI. Cell Transplant. 2015 Feb 1;24(2):203-211.
  61. Sowa Y, Imura T, Numajiri T, Nishino K, Fushiki S. Adipose-Derived Stem Cells Produce Factors Enhancing Peripheral Nerve Regeneration: Influence of Age and Anatomic Site of Origin. Stem Cells Dev. 2012 Jul 20;21(11):1852-1862.
  62. Rai V, Moellmer R, Agrawal DK: Stem Cells and angiogenesis: implications and limitations in enhancing chronic diabetic foot ulcer healing. Cells. 2022 Jul 25;11(15):2287.
  63. Thankam FG, Agrawal DK: Infarct zone: a novel platform for exosome trade in cardiac tissue regeneration. J Cardiovasc Transl Res. 2020 Oct;13(5):686-701.
  64. Littig JPB, Moellmer R, Agrawal DK, Rai V: Future applications of exosomes delivering resolvins and cytokines in facilitating diabetic foot ulcer healing. World Journal of Diabetes. 2023 Jan 15;14(1):35-47.
  65. Thankam FG, Huynh J, Fang W, Chen Y, Agrawal DK: Exosomal-ribosomal proteins-driven heterogeneity of epicardial adipose tissue derived stem cells under ischemia for cardiac regeneration. J Tissue Eng Regen Med. 2022 Apr; 16(4): 396-408.
  66. Supra R, Agrawal DK: Mechanobiology of microRNAs in intervertebral disk degeneration. J Spine Res Surg. 2023; 5(1): 1-9.
  67. Supra R, Agrawal DK: Innate immune response in orthopedic implant failure. J Orthop Sports Med. 2023; 5(1): 13-29;
  68. Fang W, Agrawal DK, Thankam FG: ‘Smart-Exosomes’: A Smart Approach for Tendon Regeneration. Tissue Eng Part B 2022 Jun; 28(3): 613-625.
  69. Agrawal DK, Rai V: Commentary: Revival of Motor and Sensory Functions: Is this a catholicon or hollow promise for paraplegia? J Thoracic Cardiovasc Surg Open 2021; 7: 43-44.
  70. 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. Biomed Res Int. 2016;2016:1-18.
  71. Wang X, Hu W, Cao Y, Yao J, Wu J, Gu X. Dog sciatic nerve regeneration across a 30-mm defect bridged by a chitosan/PGA artificial nerve graft. Brain. 2005 Aug 1;128(8):1897-910.
  72. Ray WZ, Mackinnon SE. Management of nerve gaps: Autografts, allografts, nerve transfers, and end-to-side neurorrhaphy. Exp Neurol. 2010 May;223(1):77-85.
  73. Konofaos P, ver Halen J. Nerve Repair by Means of Tubulization: Past, Present, Future. J Reconstr Microsurg. 2013 Jan 9;29(03):149-164.
  74. Aghanasir F, Aghaei H, Imani Fooladi AA, Ebrahimi M, Bagherpour G, Nourani MR. Expression of neutrophil gelatinase-associated lipocalin (NGAL) in peripheral nerve repair. Journal of Receptors and Signal Transduction. 2016 Jul 3;36(4):429-434.
  75. Gerth DJ. Clinical outcomes for Conduits and Scaffolds in peripheral nerve repair. World J Clin Cases. 2015;3(2):141.
  76. Muheremu A, Ao Q. Past, Present, and Future of Nerve Conduits in the Treatment of Peripheral Nerve Injury. Biomed Res Int. 2015;2015:1-6.
  77. Nectow AR, Marra KG, Kaplan DL. Biomaterials for the Development of Peripheral Nerve Guidance Conduits. Tissue Eng Part B Rev. 2012 Feb;18(1):40-50.
  78. Kehoe S, Zhang XF, Boyd D. FDA approved guidance conduits and wraps for peripheral nerve injury: A review of materials and efficacy. Injury. 2012 May;43(5):553-572.
  79. Deal ND, Griffin JW, Hogan M v. Nerve Conduits for Nerve Repair or Reconstruction. Journal of the American Academy of Orthopaedic Surgeons. 2012 Feb;20(2):63-68.
  80. Hasirci V, Arslantunali D, Dursun T, Yucel D, Hasirci N. Peripheral nerve conduits: technology update. Medical Devices: Evidence and Research. 2014 Dec;405.
  81. Dellon AL, Mackinnon SE. An Alternative to the Classical Nerve Graft for the Management of the Short Nerve Gap. Plast Reconstr Surg. 1988 Nov;82(5):849-856.
  82. Siemionow M, Bozkurt M, Zor F. Regeneration and repair of peripheral nerves with different biomaterials: Review. Microsurgery. 2010 Oct;30(7):574-588.
  83. Evans GRD, Brandt K, Widmer MS, Lu L, Meszlenyi RK, Gupta PK, et al. In vivo evaluation of poly(l-lactic acid) porous conduits for peripheral nerve regeneration. Biomaterials. 1999 Jun;20(12):1109-1115.
  84. Hsu S hui, Chan SH, Chiang CM, Chi-Chang Chen C, Jiang CF. Peripheral nerve regeneration using a microporous polylactic acid asymmetric conduit in a rabbit long-gap sciatic nerve transection model. Biomaterials. 2011 May;32(15):3764-3775.
  85. Leibig N, Boyle V, Kraus D, Stark GB, Penna V. C3 Toxin and Poly-DL-Lactide-ε-Caprolactone Conduits in the Critically Damaged Peripheral Nervous System. Ann Plast Surg. 2015 Mar;74(3):350-353.
  86. Chiriac S, Facca S, Diaconu M, Gouzou S, Liverneaux P. Experience of using the bioresorbable copolyester poly(DL-lactide-ε-caprolactone) nerve conduit guide NeurolacTM for nerve repair in peripheral nerve defects: Report on a series of 28 lesions. Journal of Hand Surgery (European Volume). 2012 May 10;37(4):342-349.
  87. Costa Serrão de Araújo G, Couto Neto B, Harley Santos Botelho R, Carpi Malta M. Clinical Evaluation After Peripheral Nerve Repair With Caprolactone Neurotube. HAND. 2017 Mar 8;12(2):168-174.
  88. Houshyar S, Bhattacharyya A, Shanks R. Peripheral Nerve Conduit: Materials and Structures. ACS Chem Neurosci. 2019 Aug 21;10(8):3349-3365.
  89. Onode E, Uemura T, Takamatsu K, Shintani K, Yokoi T, Okada M, et al. Nerve capping with a nerve conduit for the treatment of painful neuroma in the rat sciatic nerve. J Neurosurg. 2020 Mar;132(3):856-864.
  90. Liodaki E, Bos I, Lohmeyer J, Senyaman O, Mauss K, Siemers F, et al. Removal of Collagen Nerve Conduits (NeuraGen) after Unsuccessful Implantation: Focus on Histological Findings. J Reconstr Microsurg. 2013 Jul 1;29(08):517-522.
  91. Ishikawa N, Suzuki Y, Ohta M, Cho H, Suzuki S, Dezawa M, et al. Peripheral nerve regeneration through the space formed by a chitosan gel sponge. J Biomed Mater Res A. 2007 Oct;83A(1):33-40.
  92. Patel M, Vandevord PJ, Matthew H, Wu B, Desilva S, Wooley PH. Video-Gait Analysis of Functional Recovery of Nerve Repaired with Chitosan Nerve Guides. Tissue Eng. 2006 Nov;12(11):3189-3199.
  93. Bak M, Gutlowska O, Wagner E, Gosk J. The role of chitin and chitosan in peripheral nerve reconstruction. Polymers in Medicine. 2017 Oct 2;47(1):43-47.
  94. 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. J Tissue Eng Regen Med. 2012 Feb;6(2):163-168.

© 2016-2024, Copyrights Fortune Journals. All Rights Reserved