Editorial Open Access
Copyright ©The Author(s) 2024. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Stem Cells. May 26, 2024; 16(5): 467-478
Published online May 26, 2024. doi: 10.4252/wjsc.v16.i5.467
Mesenchymal stem cells’ “garbage bags” at work: Treating radial nerve injury with mesenchymal stem cell-derived exosomes
Mazhar Mushtaq, Doaa Hussein Zineldeen, Muhammad Abdul Mateen, Khawaja Husnain Haider, Department of Basic Sciences, Sulaiman AlRajhi University, Albukairiyah 52736, AlQaseem, Saudi Arabia
Doaa Hussein Zineldeen, Medical Biochemistry and Molecular Biology Department, Faculty of Medicine, Tanta University, Tanta 6632110, Egypt
ORCID number: Khawaja Husnain Haider (0000-0002-7907-4808).
Author contributions: Zineldeen DH contributed to writing and generating the visual abstract of this manuscript; Mushtaq M and Mateen MA were involved in writing up and revising this article; Mateen MA contributed to the figures; and Haider KH participated in writing, finalizing, and submitting the manuscript.
Conflict-of-interest statement: All the authors report no relevant conflicts of interest for this article.
Open-Access: This article is an open-access article that was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution NonCommercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: https://creativecommons.org/Licenses/by-nc/4.0/
Corresponding author: Khawaja Husnain Haider, BSc, BPharm, MPharm, PhD, Chairman, Full Professor, Department of Basic Sciences, Sulaiman AlRajhi University, AlMadina Road, Albukairiyah 52736, AlQaseem, Saudi Arabia. kh.haider@sr.edu.sa
Received: February 20, 2024
Revised: April 3, 2024
Accepted: April 25, 2024
Published online: May 26, 2024

Abstract

Unlike central nervous system injuries, peripheral nerve injuries (PNIs) are often characterized by more or less successful axonal regeneration. However, structural and functional recovery is a senile process involving multifaceted cellular and molecular processes. The contemporary treatment options are limited, with surgical intervention as the gold-standard method; however, each treatment option has its associated limitations, especially when the injury is severe with a large gap. Recent advancements in cell-based therapy and cell-free therapy approaches using stem cell-derived soluble and insoluble components of the cell secretome are fast-emerging therapeutic approaches to treating acute and chronic PNI. The recent pilot study is a leap forward in the field, which is expected to pave the way for more enormous, systematic, and well-designed clinical trials to assess the therapeutic efficacy of mesenchymal stem cell-derived exosomes as a bio-drug either alone or as part of a combinatorial approach, in an attempt synergize the best of novel treatment approaches to address the complexity of the neural repair and regeneration.

Key Words: Exosome, Mesenchymal stem cells, Nerve injury, Stem cells, Secretome, Regeneration

Core Tip: The extracellular vesicles constituting the insoluble component of the secretome were once considered cell’s garbage bags. They have become a hot area of research since realizing their significance as an essential means of intracellular communication. They have shown promise for therapeutic applications for repairing and regenerating the damaged tissues via delivering their payload to the resident reparative cells and supporting them in the intrinsic repair process. Although stem cell-derived exosomes have been extensively studied for peripheral nerve injury repair in experimental animal models, their use for radial nerve injury repair in a patient, as the pilot study by Civelek et al, is expected to pave the way for assessment in future clinical trials.



INTRODUCTION

Stem cells and their derived paracrine factors are potential therapeutic modalities to treat spinal cord injury, stroke, and neurodegenerative diseases[1]. Among different stem cell types, mesenchymal stem cells (MSCs) have remained at the forefront of characterization and assessment in the preclinical and clinical settings as choice cells for their regenerative properties in nerve injury repair[2]. They foster the survival and regeneration of neurons via multifaceted mechanisms that primarily include differentiation into morpho-functionally competent neural cells and the release of trophic factors, thereby establishing a conducive microenvironment for neural tissue repair[3]. Some of the latest published studies highlighting their findings in preclinical and clinical settings for peripheral nerve injury (PNI) treatment have been summarized in Tables 1 and 2[4-20].

Table 1 Recently reported peripheral nerve injury repair in the experimental animal models.
Ref.
Experimental model (in vitro or animal)
Therapeutic modalities
Main findings
MSCs-based therapy
Zhang et al[4], 2024SCs from injured sciatic nerves and HUVECsMSCs treated with PRP-derived exosomesTreatment with PRP-exosome improved MSC survival. Exosome-treated MSCs, co-cultured with SCs, reduced their apoptosis and enhanced SC proliferation after PNI. Similarly, exosome-treated MSCs also had pro-migratory and angiogenic effects. Cytokine array analysis and ELISA showed upregulation of 155 proteins and downregulation of six proteins, with many pro-angiogenic and neurotrophic factors. Western blot revealed the activation of the PI3K/Akt signaling pathway in exosomes-treated MSCs
Sivanarayanan et al[5], 2023Sciatic nerve crush injury in rabbitAllogenic BM-MSCs and their CMBM-MSCs and BM-MSCS-CM treatment improved the regenerative capacity in acute and subacute injury groups, with slightly better improvements in the subacute groups. BM-MSCs supported the healing process of PNI, whereas CM increased the healing process
Yalçın et al[6], 2023Sciatic nerve injury in ratADSCsThe study documented the role of syndecan-1 and heat shock protein 70 in the regenerative effects of ADSCs on PNI. Histology and EMG showed that treatment with ADSCs significantly improved nerve regeneration and its functionality via the release of nerve growth factor
Liu et al[7], 2020Sprague-Dawley ratsSC-like ADSCs are placed on an acellular scaffold after treatment with nerve leachateSprague-Dawley rats were divided into four groups: Scaffold only, untreated ADSCs + scaffold, nerve leachate-treated ADSCs + scaffold, and autograft. Four months after treatment, the average area, density, and thickness of regenerated nerve fibers in the nerve leachate-treated ADSCs + scaffold group significantly increased compared to the untreated ADSCs + scaffold group. These data show the superiority of nerve leachate-treated ADSCs for treating PNI
Kizilay et al[8], 2017Wistar rat model of sciatica nerve injury by clip compressionBM-MSCsThe proximal, distal, and mean latency values were higher in MSC treatment groups vs without MSC-treated animals. The nerve conduction velocity, compound action potential, and the number of axons in MSC-treated animals are higher than in non-MSC-treated animals. Also, myelin damage decreased in MSC-treated animals
Cell-free therapy
Growth factor-based approach for PNI
Shi et al[9], 2022Rat sciatic nerve transection modelIn vitro experimental studies show that BDNF/PLGA sustained-release microsphere treatment improved migration and neural differentiation of ADSCs. In vivo studies indicated that BDNF microsphere treatment significantly reduced the nerve conduction velocity compound amplitude compared to the untreated animals. Moreover, the BDNF microsphere group had more closely arranged and uniformly distributed nerve fibers than the control animals
Li et al[10], 2021Rat sciatic nerve transection modelLeision site injection of a lentivirus expressing FGF13FGF13 treatment successfully recovered motor and sensory functions via axon elongation and remyelination. FGF13 pretreatment enhanced SCs survival and increased cellular microtubule-associated proteins in vitro PNI model. The data supported the role of FGF13 in stabilizing cellular microtubules, which is essential for promoting PNI repair following PNI
Su et al[11], 2020Rat sciatic nerve transection modelComposite nerve conduit with slow-release BDNFThe study used fabricated composite nerve conduits with slow-release BDNF to treat PNI and compare the regeneration potentials of autologous nerve grafts. The BDNF composite conduits remained bioactive for at least three months and successfully regenerated a 10-mm sciatic nerve gap
Lu et al[12], 2019Rat model of sciatic crush injuryIntramuscular delivery of FGF21 once daily for seven daysFGF21 treatment led to functional and morphologic recovery with improved motor and sensory function, enhanced axonal remyelination and re-growth, and increased SC proliferation. Local FGF21 treatment reduced oxidative stress via activation of Nrf-2 and ERK. FGF21 also reduced autophagic cell death in SCs
Exosome-based approach for PNI
Zhu et al[13], 2023Mouse model of spared nerve injuryExosomes from UC-MSCs under hypoxiaAfter 48 h of culture under 3% oxygen in a serum-free culture system, UC-MSCs secreted higher EVs than the control cells. SCs could uptake EVs in vitro and increase their growth and migration. The treatment of animals with EVs accelerated the recruitment of SCs at the PNI site and supported PN repair and regeneration
Hu et al[14], 2023Rat model of the injured sciatic nerveSCs-like cells derived from hA-MSCs. Exosomes from hA-MSCs or SC-like cells from hA-MSCsSC-like cells were successfully differentiated from hA-MSCs and used for exosome collection. Treatment with exosomes from SC-like cells significantly enhanced (vs hA-MSCs-derived exosomes) motor function recovery, reduced gastrocnemius muscle atrophy, and supported axonal regrowth, myelin formation, and angiogenesis in the rat model. They were also more efficiently absorbed by SCs and promoted the proliferation and migration of SCs
Yin et al[15], 2021In vitro model and a rat model of sciatic nerve injuryADSCsExosome treatment inhibited autophagy and karyopherin-α2 levels, which were significantly increased in SCs in the injured sciatic nerve, both in vivo and in vitro. Abrogation of karyopherin-α2 reduced SCs autophagy, with the role of miRNA-26b. Treatment with exosomes supported myelin sheath regeneration in rats with a sciatic NI
Liu et al[16], 2020PNI model rats supported by in vitro studiesADSCs and their derivative exosomesTreatment with ADSC-derived exosomes significantly reduced SC apoptosis after PNI via increased Bcl-2 and decreased Bax mRNA expression, in addition to increasing SC proliferation. Histological data in PNI model rats also observed these effects
Chen et al[17], 2019In vitro model and rat sciatic nerve transection model with a 10-mm gapHuman ADSCs-derived exosomes and in vitroIn vitro studies showed that SCs internalized human ASCs-derived exosomes to enhance their proliferation, migration, myelination, and secretion of neurotrophic factors. Treatment with ASC-exosomes supported axon regeneration in a rat sciatic nerve transection model with a 10-mm gap and supported myelination and restoration of denervation muscle atrophy. This data showed the efficacy of exosomes in promoting PN regeneration by restoring SC function
Masgutov et al[18], 2019Wistar rat sciatic nerve injury modelADSCsADSCs-derived MSCs were delivered using fibrin glue to the traumatic injury, helped to fix the cells at the graft site, and gave extracellular matrix support to the provided cells. The transplanted cells were neuroprotective on DRG L5 sensory neurons and stimulated axon growth and myelination. Also, MSCs promoted nerve angiogenesis and motor function recovery
Table 2 Clinical studies for peripheral nerve injury repair using Schwan cells, stem cells, and their derived exosomes.
NCT#
Study title
Conditions
Interventions
Primary outcome
Sponsor
Collaborators
NCT04346680Intraoperative ADSC administration during nerve releaseNeurotmesis of peripheral nerve disorderADSC administrationElectrophysiological improvement, improvement in EMG - the appearance of activities in denervated muscles, one yearMossakowski MRC Polish Academy of SciencesCentre of Postgraduate Medical Education
NCT03964129BMAC nerve allograft studyPNI upper limbAvance nerve graft with autologous BMACComparison of AEs between patients treated with ANG with BMAC and the historical data of nerve repairs with the ANG only. Long-term study - AEs, such as infection, wound dehiscence, neuropathy, carpal tunnel syndrome, bleeding, seroma, and lymphocele, will be recorded and analyzed. AEs will be mapped to a MedDRA-preferred term and system organ classificationBrooke Army Medical CenterWalter Reed National Military Medical Center; Cleveland Clinic Lerner Research Institute
NCT03359330; PKUPH-PNIMid-term effect observation of biodegradable conduit small gap tublization repairing PNIPNIsDegradable conduit small gap tublizationTo observe the mid-term clinical effect of biodegradable conduit small gap tublization on the repair of PNI in multi-center patients and fresh PNIs in the upper extremitiesPeking University People’s Hospital-
NCT05541250Safety and efficacy of autologous human SCs augmentation in severe peripheral nerve injuryPNIsAutologous human SCThe primary purpose of this phase I study is to evaluate the safety of injecting one’s SCs along with nerve auto-graft after a severe nerve injury, such as a sciatic nerve or brachial plexus injuryUniversity of Miami, Florida, United States (Recruiting)-
NCT04654286Clinical outcomes of HAM and allogeneic MSCs composite augmentation for nerve transfer procedure in brachial plexus injury patientsBrachial plexus neuropathiesNerve transfer or nerve transfer with HAM-MSC composite wrappingAROM pre-surgery and 12-month follow-up for shoulder flexion, extension, abduction, adduction, external rotation, and internal rotation using the MRC scale (ranging from 0-5)Dr. Soetomo General Hospital, Jakarta
Huang et al[19], 2016A clinical study on the treatment of peripheral nerve injury growth factor of mecobalamin combined with nerve150 PNI patientsMecobalamin (0.5 mg, I.V, once a day) combined with NGF (30 mg, I.M injection, once a day) for 3-6 wkTreatment with mecobalamin combined with NGF improved the sensorimotor evaluation of the curative effect made by the British Medical Research Institute of Neurotrauma SocietyGuangxi Basic Science and Technology Plan Project PR China (No.: 20111209)
Civelek et al[20], 2024Effects of exosomes from mesenchymal stem cells on functional recovery of a patient with total radial nerve injury: A pilot studyOne patient with total radial nerve injuryWJ-MSCs derived exosomesThe six-month follow-up based on the BMRC and Mackinnon-Dellon scales showed improved motor (M5, excellent), and sensory functions also showed improvement (S3+, good). These results were achieved without physical therapy. Substantial axonal damage was observed at a ten-week follow-up, but nerve re-innervation was observed by EMG, which also improved significantly during the six-month follow-upDepartment of Neurosurgery, University of Health Sciences

Exosomes are small extracellular vesicles (50-100 nm in size bounded by a lipid bilayer membrane) released by cells as an integral part of their paracrine activity. They contain a specific cargo of bioactive molecules. Once considered the “garbage bags” for cell metabolic waste disposal[21], exosomes are being established as critical regulators of diverse physiological cell processes and essential mediators of intercellular communication[22,23].

MSC-derived exosomes exhibit neuroprotective and regenerative effects by modulating inflammation, promoting cell survival, antioxidant properties, anti-apoptotic and pro-proliferative activities, and stimulating neuronal differentiation[24]. They also have higher biocompatibility and low risk of tumorigenicity, microvascular, immune rejection, etc.; safety concerns are generally associated with using cell grafts[25]. As they copycat the regenerative effects of MSCs, they are potential candidates as the non-cellular alternatives for neurodegeneration. In the following sections, we will focus on advancements in PNI treatment using stem cells and their derived exosomes, recapitulated in Figure 1. Moreover, we will also delve into the clinical trials assessing stem cell-based and their derivative exosome-based approaches for treating PNI patients, with a critical appreciation of clinical experience in the pilot study published by Civelek et al[20] using MSCs-derived exosomes.

Figure 1
Figure 1 The novel cell-based and cell-free therapy approaches for peripheral nerve injury repair and regeneration. MSC: Mesenchymal stem cell; iPSC: Induced pluripotent stem cell; NSC: Neural stem cell; VEGF: Vascular endothelial growth factor; miRNA: MicroRNA; lncRNA: Long non-coding RNA; circRNA: Circular RNA; MHC: Major histocompatibility complex.
PNI AND INTRINSIC REPAIR PROCESS

Unlike the brain’s synaptic plasticity, which allows it to have functional reorganization with limited or no repair or renewal in the event of injury[26], axonal regrowth through peripheral nerve sheaths has been observed in PNI as part of the recovery process[27,28] (Figure 2). However, the recovery process in PNI is influenced by the severity of the injury. In non-severe injuries, i.e., neuropraxia and local demyelination, axonotmesis with intact neural stroma, and loss of funiculus and its contents, the nerve recovers from the damage by the inherent ability to repair[29].

Figure 2
Figure 2 Summary of the intrinsic peripheral nerve injury repair mechanisms and the emerging novel treatment modalities to support intrinsic peripheral nerve injury repair. MSC: Mesenchymal stem cell; iPSC: Induced pluripotent stem cell; NSC: Neural stem cell; VEGF: Vascular endothelial growth factor; miRNA: MicroRNA; lncRNA: Long non-coding RNA; circRNA: Circular RNA; MHC: Major histocompatibility complex.

On the contrary, injuries involving the severing of the nerve may require gold-standard surgical intervention. Diverse cellular and molecular events involving Schwann cells (SCs), macrophages, and extracellular matrix contribute to the repair process, which usually spans a prolonged duration[28]. Neuronal repair in the peripheral nervous system involves more than one mechanism, i.e., axonal regrowth, central nerve cell restoration, and neurogenesis, to ensure functional recovery[30,31].

Axonal regrowth may involve damaged nerve cells from the peripheral ganglia or reactivation of signaling from the intact central nerve cells of the severed axons[32]. On the other hand, central nerve cell restoration involves sprouting, a process wherein new axons, dendrites, and synapses grow from the intact central nerve cell body. Neurogenesis, the growth of new neurons, is possible if the neurons retain some of their multipotent neural stem/progenitor cell population, especially near the injury site[33].

CELLULAR AND MOLECULAR BASIS OF PNI

SCs myelinate the peripheral axons, support the regrowth of axons by secreting laminin, fibronectin, and collagens, and produce neurotrophic factors, i.e., nerve growth factor (NGF), brain-derived neurotrophic factor (BDNF), and glial cell-derived neurotrophic factor. Additionally, they facilitate remyelination by aligning as Bands of Büngner, clear cellular and myelin debris by phagocytosis, and guide proper axonal regeneration by increasing cell adhesion molecules NCAM, L1, and N-cadherin and early re-expression of bHLH family transcription factors protein growth-associated protein-43, and that helps actin and microtubule cytoskeletal recovery[34]. SCs and fibroblasts also secrete endoneurial matrix comprising various components, i.e., collagen, glycoproteins (fibronectins, laminins), glycosaminoglycans, and proteoglycans[35]. These matrix components can inhibit or stimulate axonal repair, and their activity is enhanced during axonal repair after injury.

Joining hands with SCs are macrophages, especially M2 macrophages, that remove the debris from the damaged peripheral nerves. Additionally, they provide a conducive microenvironment for nerve repair and growth via modulating inflammation and by releasing pro-inflammatory cytokines, i.e., interleukin (IL)-1 and tumor necrosis factor-α, to promote SC activation[7]. During the resolution phase of PNI repair, the macrophages also stimulate the release of anti-inflammatory cytokines, i.e., IL-10, to help resolve inflammation and promote tissue healing.

TREATMENT ADVANCES FOR PNI

PNI can result in significant functional impairments, necessitating proper therapeutic options for nerve repair. In clinical settings, microsurgical intervention by nerve autografting is considered the gold-standard treatment for PNI. However, it is limited by the availability of the nerve graft, chances of infection, and neuroma development. The other contemporary treatment options for PNI include direct suturing, SCs transplantation, and electrical stimulation, but with their respective deficiencies, especially when treating significant nerve defects wherein they fail to achieve complete repair. The following section elaborates on the current advancements in PNI treatment using cell-based and cell-free approaches, as summarized in Figure 2[36].

MSCs AND PNI REPAIR

The cell-based therapy approach has come a long way with encouraging data for peripheral nerve repair and regeneration; several stem/progenitor cells have been assessed for their neuronal reparability in pre-clinical models. These include pluripotent cells, i.e., embryonic stem cells (ESCs), induced pluripotent stem cells (iPSCs), and adult tissue-derived stem/progenitor cells, i.e., MSCs, neural stem cells (NSCs), etc. Although pluripotent stem cells have high differentiation potential, using ESCs has ethical issues and moral strings attached. In contrast, using iPSCs (considered surrogate ESCs) has tumorigenic potential due to genomic instability induced during reprogramming[37]. MSCs from amongst the adult tissue-derived stem cells have gained considerable attention in nerve regeneration studies in experimental animal models with their self-renewal, robust nature, excellent cell biology, and multipotentiality[38]. Table 1 summarizes the data published by various recently published experimental animal studies using MSCs. From the different tissue sources of MSCs, adipose tissue-derived MSCs and bone marrow (BM)-derived MSCs have shown excellent neural regeneration and repairability. Their reparability is via multifactorial mechanisms encompassing neuroprotection to differentiation to adopt morphofunctional competent neural cells besides the release of secretome containing neurotrophic factors and exosomes rich in the bioactive payload of microRNAs (miRNAs), enzymes, and protein[39], which will be discussed in the following sections. To enhance their stemness and reparability, they have been combined with NSCs, preconditioned to enhance their paracrine activity, or genetically modified to serve as a source of neurotrophic factors as paracrine factors[40,41].

MSCs have also advanced to the clinical assessment for PNI repair in different clinical studies (Table 2), mainly as an adjunct to other therapeutic interventions, such as intraoperative administration (Clinicaltrials.org ID: NCT04346680), being part of the nerve transfer composite (Clinicaltrials.org ID: NCT04654286), or avance nerve transfer (Clinicaltrials.org ID: NCT03964129).

CELL-FREE THERAPY-BASED APPROACHES FOR PNI REPAIR
Growth factor-based strategy

Besides cell-based therapy, there has been immense interest in using a growth factors-based approach to support neuronal regeneration and functional recovery[42]. Several growth factors have been identified as potential candidates for this end (Table 3). These growth factors contribute via interacting with each other to initiate various signaling pathways to guide and stimulate regeneration and functional recovery of the injured neurons. A generally accepted mechanism of growth factor-based treatment of PNI is that the target-derived growth factors are captured at the nerve terminals via receptor-mediated endocytosis and get retrogradely transported to cell bodies to impart their neurotrophic action.

Table 3 List some commonly studied growth factors for peripheral nerve injury treatment.
Ref.
Growth factor
Sandoval-Castellanos et al[43], 2020Brain-derived neurotrophic factor
Xu et al[44], 2023Ciliary neurotrophic factor
Gu et al[45], 2024Chemokine platelet factor-4
Romano and Bucci[46], 2020Epidermal growth factor
Cintron-Colon et al[47], 2022Glial cell line-derived neurotrophic factor
Ye et al[48], 2022Hepatocyte growth factor
Slavin et al[49], 2021Insulin-like growth factor-1
Alastra et al[50], 2021Nerve growth factor
Li et al[51], 2023NGF+ basic fibroblast growth factor
Golzadeh and Mohammadi[52], 2016Platelet-derived growth factor
Ding et al[53], 2024Transforming growth factor
Xu et al[44], 2023Vascular endothelial growth factor

From the list, neurotrophic growth factors families and cytokines, i.e., NGF, GDN, fibroblast growth factor (FGF), ciliary neurotrophic factor, etc., are critical in oligodendrocyte precursors’ migration, proliferation, and differentiation and regulate axonal interactions and myelination. Some reported signaling pathways underlying these cellular level changes include the mitogen-activated protein kinase, phosphoinositide 3-kinase/Akt, nuclear transcription factor-kappa B, BDNF/Trk, Ras/extracellular regulated protein kinases, and transforming growth factor-β[54]. Understanding their interaction with specific receptors and the downstream signaling is essential for progressing growth factor-based therapeutic intervention. Numerous preclinical and clinical studies have investigated the efficacy of growth factor-based therapies in promoting nerve regeneration in animal models with PNI, as summarized in Tables 1 and 2.

MSC-derived exosomes for PNI

As discussed earlier, the MSCs-based cell therapy approach has shown promise in PNI treatment. Still, their use is not without limitations, i.e., tumorigenesis, triggering an immune response, rejection of the cell graft, off-the-shelf non-availability, logistic issues, etc., hampered the pace of their reckoning as a routine treatment option[55,56]. Hence, exosomes derived from different cell types, i.e., SCs, MSCs, etc., offer potential alternatives to overcome these limitations[57]. Their physiological functions primarily involve long-distance intracellular communication, using their surface proteins and lipid rafts to fuse with the recipient cells and deliver the payload of bioactive molecules. Alternatively, they can be taken up by the recipient cells via endocytosis.

An essential phase in exosome biogenesis is the initiation of intraluminal vesicles (ILVs) through the invagination of the endosomal membrane[58,59]. This is followed by payload encapsulation consisting of proteins, lipids, mRNA, miRNA, long non-coding RNA (lncRNA), circular RNAs (circRNAs), DNA, enzymes, signaling proteins, sphingolipids, etc., and the release of ILVs into the extracellular environment as exosomes[60,61]. Each cell type’s payload composition is distinct under a given set of conditions[62,63], contributing to their functional heterogeneity, i.e., cell survival, apoptosis, proliferation, immunomodulation, etc. More recently, exosome modification protocols are being developed to modulate them for good biocompatibility, low immunogenicity, capability to cross biological membrane barriers, and, more importantly, to carry a specific payload composition of interest. Different exosome modification techniques are summarized in Figure 3. In line with their diverse theragnostic applications, MSCs-derived exosomes have been extensively studied to support neuronal functional recovery and regeneration in PNI experimental animal models[14,17,64].

Figure 3
Figure 3 Payload manipulation of mesenchymal stem cells-derived exosomes.

The treatment of SCs with MSCs-derived exosomes reduces their autophagy via miRNA-26b mediated abrogation of karyopherin subunit α2[15], improves SC proliferation dose-dependently, entering SCs through endocytosis to modulate their gene expression profile and supporting their re-myelination[15,17,65,66]. Exosome-based treatment also exerts neuroprotective effects via PI3/Akt signaling activation[67]. Currently, “smart exosomes” are being developed by reprogramming and modulating their surface characteristics for efficient, targeted uptake by the recipient cells and manipulating their payload for delivery to the recipient cells[68]. For example, one of the essential entities in the payload are miRNAs, small, ncRNA molecules with mega functions as cell function regulators[69], that get delivered to the injured neurons during cellular communication[64,70]. They affect neuron differentiation, proliferation, angiogenesis, axonal regrowth, and other cellular functions[71]. For example, MSCs’ exosome-derived miR-21, miR-124, and miR-133 have been attributed to promoting neuronal regeneration[72].

In the neural injury model, MSCs-derived exosomes overexpressing miRNA-133b transferred miRNA-133b to injured neuronal cells, promoting post-stroke neuronal remodeling and functional recovery[73]. Furthermore, MSC exosomes have demonstrated benefits in brain injury, accelerating recovery through neurosynaptic remodeling, neurogenesis, and angiogenesis[74]. Besides miRNAs, circRNAs are resistant to degradation, act as miRNA sponges in neural apoptosis, angiogenesis, and synaptic plasticity modulation, and hold immense promise for neuroregeneration[75]. MSC-derived exosomal lncRNAs have been reported to enhance neuronal survival and promote axonal regeneration after nerve injury. Notably, lncRNA HOTAIR and MALAT1 have shown significant potential in promoting neuroregeneration by modulating several molecular pathways involved in nerve repair.

MSCS AND THEIR EXOSOMES FOR PNI IN CLINICAL SETTINGS

PNI, arising from a diverse range of etiologies such as trauma and underlying medical conditions, poses substantial challenges in both clinical management and subsequent restoration of functional capacity. MSC-derived exosomes, assessed in clinical settings for treating various disease conditions[76,77], have also progressed to clinical application for treating PNI as a part of the cell-free therapy approach. There are at least five registered clinical trials for the safety and efficacy assessment of SCs, MSCs, and their derived exosomes, although their current status remains unknown (Table 2). MSCs focus on diverse tissue sources, i.e., adipose tissue, BM, and human amniotic membrane (HAM), due to their superior biology, paracrine activity, and differentiation characteristics[78].

Mossakowski Medical Research Council Polish Academy of Sciences has registered a clinical trial entitled “Intraoperative ADSCs Administration During Nerve Release” (ClinicalTrials.gov Identifier: NCT04346680). The trial proposes autologous adipose-derived stem cell (ADSC) transplantation in six patients with failure to reconstruct peripheral nerves. ADSCs will be delivered during a last-chance surgery (neurolysis and nerve release) on a previously reconstructed nerve. The patients included in the study will be subjected to clinical and electrophysiological assessment. The patients will receive ten microinjections of ADSC along the injured nerve, and safety, adverse events, and efficacy, i.e., electromyography (EMG) and sensory threshold, will be assessed. On the other hand, the second registered study, “BMAC Nerve Allograft Study” (ClinicalTrials.gov Identifier: NCT04346680), will adopt a combinatorial approach involving an avance nerve graft combined with BM aspirate concentrate delivery. The third study, “Clinical Outcomes of Human Amniotic Membrane and Allogeneic Mesenchymal Stem Cells Composite Augmentation for Nerve Transfer Procedure in Brachial Plexus Injury Patients” (ClinicalTrials.gov Identifier: NCT04654286), will investigate the safety and efficacy of a composite between HAM and allogeneic ADSCs as a wrapping in the nerve transfer procedure of upper traumatic brachial plexus injury patients, with a focus on the augmentation of axonal regeneration. Another phase I study is entitled “Safety and Efficacy of Autologous Human SCs Augmentation in Severe Peripheral Nerve Injury” (NCT05541250) at the University of Miami, United States. The study, with primary safety and efficacy endpoints, is still in the recruitment stage.

Unlike the aforementioned clinical trials, pilot study data reported by Civelek et al[20] is unique in using MSCs-derived exosomes for therapeutic intervention as part of the cell-free therapy approach. The authors rationalized using exosomes and anticipated that exosomes would deliver their miRNA payload to the injured nerve, leading to the repair and regeneration of the nerve and reducing the inflammatory activity in the injured area via anti-inflammatory cytokines. The authors used 1 mL exosomes divided into four doses, with 1.25 billion vesicles each, delivered epineurally, after using a sural autograft. Functional sensory and motor recovery were observed as early as the 10th-wk post-grafting, and indications of re-innervation were evidenced by neurological examination and control EMG during a six-month follow-up. These data enhance our understanding of the neurobiological consequences of peripheral nerve damage and emphasize the potential of MSC-derived exosomes, offering avenues for future clinical advancements. Despite encouraging data from the study, it has its limitations. Firstly, it is a pilot study that includes only one patient. Hence, the data needs to reflect the safety and efficacy of the treatment approach, which necessitates more extensive studies involving more participants with a control arm for comparison. Besides, the rationale for exosome dose selection has little justification. There has yet to be an attempt to profile the exosome payload composition, i.e., miRNA, cytokines, growth factors, etc. The authors, therefore, assumed that specific miRNAs might have participated merely based on the published data. These limitations certainly make it challenging to comprehend the underlying mechanism. Nevertheless, despite these limitations, the study is a leap forward in neural repair and regeneration.

CONCLUSION

In conclusion, the recent advances in cell-based (especially MSC- and its derived exosomes) have shown promise in both preclinical and clinical settings; however, the field is still evolving and needs further research before it can be adopted as a routine therapeutic modality for PNI repair and neural regeneration. The pilot trial report from Civelek et al[20] is a leap forward in the clinical arena that has already started evolving but warrants a more extensive, systematic, and well-defined study. A few challenges encountered in using exosomes in neural repair and regeneration necessitating particular focus include optimizing an efficient isolation and purification protocol for clinical-grade exosome preparation, a method to achieve a well-defined payload of exosomes, and an optimal exosome delivery method for treating PNI. Given the complexity of an injured nerve’s repair and regeneration process, it would be prudent to adopt a combinatorial approach by combining exosome delivery with other emerging PNI treatment approaches. For example, it can be integrated with the nanoparticle-based approach, which gives encouraging results in promoting peripheral nerve repair and neuroprotection, besides efficient drug delivery methods[79]. On the same note, synergizing the therapeutic potential of exosomes as a bio-drug with other treatment approaches, including surgical end-to-end anastomoses, is worth exploiting for optimal therapeutic benefits.

Footnotes

Provenance and peer review: Invited article; Externally peer reviewed.

Peer-review model: Single blind

Specialty type: Cell and tissue engineering

Country of origin: Saudi Arabia

Peer-review report’s classification

Scientific Quality: Grade B

Novelty: Grade A

Creativity or Innovation: Grade B

Scientific Significance: Grade B

P-Reviewer: Zhang Q, China S-Editor: Wang JJ L-Editor: A P-Editor: Che XX

References
1.  Zhong L, Wang J, Wang P, Liu X, Liu P, Cheng X, Cao L, Wu H, Chen J, Zhou L. Neural stem cell-derived exosomes and regeneration: cell-free therapeutic strategies for traumatic brain injury. Stem Cell Res Ther. 2023;14:198.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 2]  [Reference Citation Analysis (0)]
2.  Zhang RC, Du WQ, Zhang JY, Yu SX, Lu FZ, Ding HM, Cheng YB, Ren C, Geng DQ. Mesenchymal stem cell treatment for peripheral nerve injury: a narrative review. Neural Regen Res. 2021;16:2170-2176.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 20]  [Cited by in F6Publishing: 41]  [Article Influence: 13.7]  [Reference Citation Analysis (0)]
3.  Gutiérrez-Fernández M, Fuentes B, Rodríguez-Frutos B, Ramos-Cejudo J, Vallejo-Cremades MT, Díez-Tejedor E. Trophic factors and cell therapy to stimulate brain repair after ischaemic stroke. J Cell Mol Med. 2012;16:2280-2290.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 42]  [Cited by in F6Publishing: 39]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
4.  Zhang Y, Yi D, Hong Q, Cao J, Geng X, Liu J, Xu C, Cao M, Chen C, Xu S, Zhang Z, Li M, Zhu Y, Peng N. Platelet-rich plasma-derived exosomes boost mesenchymal stem cells to promote peripheral nerve regeneration. J Control Release. 2024;367:265-282.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
5.  Sivanarayanan TB, Bhat IA, Sharun K, Palakkara S, Singh R, Remya 5th, Parmar MS, Bhardwaj R, Chandra V, Munuswamy P, Kinjavdekar P, Pawde AM, Amarpal, Sharma GT. Allogenic bone marrow-derived mesenchymal stem cells and its conditioned media for repairing acute and sub-acute peripheral nerve injuries in a rabbit model. Tissue Cell. 2023;82:102053.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 2]  [Reference Citation Analysis (0)]
6.  Yalçın MB, Bora ES, Erdoğan MA, Çakır A, Erbaş O. The Effect of Adipose-Derived Mesenchymal Stem Cells on Peripheral Nerve Damage in a Rodent Model. J Clin Med. 2023;12.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 1]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
7.  Liu Y, Dong R, Zhang C, Yang Y, Xu Y, Wang H, Zhang M, Zhu J, Wang Y, Sun Y, Zhang Z. Therapeutic effects of nerve leachate-treated adipose-derived mesenchymal stem cells on rat sciatic nerve injury. Exp Ther Med. 2020;19:223-231.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 4]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
8.  Kizilay Z, Aktas S, Kahraman Cetin N, Bakay Ilhan D, Ersoy G, Erken HA. Effect of systemic application of bone marrow-derived mesenchymal stem cells on healing of peripheral nerve injury in an experimental sciatic nerve injury model. Turk Neurosurg. 2017;.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 4]  [Article Influence: 0.6]  [Reference Citation Analysis (0)]
9.  Shi ZL, Fan ZY, Zhang H, Li ST, Yuan H, Tong JH. Localized delivery of brain-derived neurotrophic factor from PLGA microspheres promotes peripheral nerve regeneration in rats. J Orthop Surg Res. 2022;17:172.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 8]  [Article Influence: 4.0]  [Reference Citation Analysis (0)]
10.  Li R, Tao X, Huang M, Peng Y, Liang J, Wu Y, Jiang Y. Fibroblast Growth Factor 13 Facilitates Peripheral Nerve Regeneration through Maintaining Microtubule Stability. Oxid Med Cell Longev. 2021;2021:5481228.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 4]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
11.  Su H, Xu F, Sun H, Fu X, Zhao Y. Preparation and Evaluation of BDNF Composite Conduits for Regeneration of Sciatic Nerve Defect in Rats. J Pharm Sci. 2020;109:2189-2195.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 3]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
12.  Lu Y, Li R, Zhu J, Wu Y, Li D, Dong L, Li Y, Wen X, Yu F, Zhang H, Ni X, Du S, Li X, Xiao J, Wang J. Fibroblast growth factor 21 facilitates peripheral nerve regeneration through suppressing oxidative damage and autophagic cell death. J Cell Mol Med. 2019;23:497-511.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 27]  [Cited by in F6Publishing: 40]  [Article Influence: 6.7]  [Reference Citation Analysis (0)]
13.  Zhu Z, Zhang Y, Huang Z, Hao H, Yan M. Hypoxic culture of umbilical cord mesenchymal stem cell-derived sEVs prompts peripheral nerve injury repair. Front Cell Neurosci. 2023;16:897224.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
14.  Hu T, Chang S, Qi F, Zhang Z, Chen J, Jiang L, Wang D, Deng C, Nie K, Xu G, Wei Z. Neural grafts containing exosomes derived from Schwann cell-like cells promote peripheral nerve regeneration in rats. Burns Trauma. 2023;11:tkad013.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 2]  [Reference Citation Analysis (0)]
15.  Yin G, Yu B, Liu C, Lin Y, Xie Z, Hu Y, Lin H. Exosomes produced by adipose-derived stem cells inhibit schwann cells autophagy and promote the regeneration of the myelin sheath. Int J Biochem Cell Biol. 2021;132:105921.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 20]  [Article Influence: 6.7]  [Reference Citation Analysis (0)]
16.  Liu CY, Yin G, Sun YD, Lin YF, Xie Z, English AW, Li QF, Lin HD. Effect of exosomes from adipose-derived stem cells on the apoptosis of Schwann cells in peripheral nerve injury. CNS Neurosci Ther. 2020;26:189-196.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 40]  [Cited by in F6Publishing: 52]  [Article Influence: 10.4]  [Reference Citation Analysis (0)]
17.  Chen J, Ren S, Duscher D, Kang Y, Liu Y, Wang C, Yuan M, Guo G, Xiong H, Zhan P, Wang Y, Machens HG, Chen Z. Exosomes from human adipose-derived stem cells promote sciatic nerve regeneration via optimizing Schwann cell function. J Cell Physiol. 2019;234:23097-23110.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 48]  [Cited by in F6Publishing: 71]  [Article Influence: 14.2]  [Reference Citation Analysis (0)]
18.  Masgutov R, Masgutova G, Mullakhmetova A, Zhuravleva M, Shulman A, Rogozhin A, Syromiatnikova V, Andreeva D, Zeinalova A, Idrisova K, Allegrucci C, Kiyasov A, Rizvanov A. Adipose-Derived Mesenchymal Stem Cells Applied in Fibrin Glue Stimulate Peripheral Nerve Regeneration. Front Med (Lausanne). 2019;6:68.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 39]  [Cited by in F6Publishing: 33]  [Article Influence: 6.6]  [Reference Citation Analysis (0)]
19.  Huang C, Su G, Wei W, Lu W, Mai Y, Hua S, Zhao Y, Lu J. A clinical study on the treatment of peripheral nerve injury growth factor of mecobalamin combined with nerve. WJNS. 2016;6:75-81.  [PubMed]  [DOI]  [Cited in This Article: ]
20.  Civelek E, Kabatas S, Savrunlu EC, Diren F, Kaplan N, Ofluoğlu D, Karaöz E. Effects of exosomes from mesenchymal stem cells on functional recovery of a patient with total radial nerve injury: A pilot study. World J Stem Cells. 2024;16:19-32.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
21.  de Gassart A, Géminard C, Hoekstra D, Vidal M. Exosome secretion: the art of reutilizing nonrecycled proteins? Traffic. 2004;5:896-903.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 105]  [Cited by in F6Publishing: 110]  [Article Influence: 5.5]  [Reference Citation Analysis (0)]
22.  Ma ZJ, Yang JJ, Lu YB, Liu ZY, Wang XX. Mesenchymal stem cell-derived exosomes: Toward cell-free therapeutic strategies in regenerative medicine. World J Stem Cells. 2020;12:814-840.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 40]  [Cited by in F6Publishing: 49]  [Article Influence: 12.3]  [Reference Citation Analysis (0)]
23.  Couch Y, Buzàs EI, Di Vizio D, Gho YS, Harrison P, Hill AF, Lötvall J, Raposo G, Stahl PD, Théry C, Witwer KW, Carter DRF. A brief history of nearly EV-erything - The rise and rise of extracellular vesicles. J Extracell Vesicles. 2021;10:e12144.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 93]  [Cited by in F6Publishing: 147]  [Article Influence: 49.0]  [Reference Citation Analysis (0)]
24.  Kang X, Zuo Z, Hong W, Tang H, Geng W. Progress of Research on Exosomes in the Protection Against Ischemic Brain Injury. Front Neurosci. 2019;13:1149.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 21]  [Cited by in F6Publishing: 21]  [Article Influence: 4.2]  [Reference Citation Analysis (0)]
25.  Nikfarjam S, Rezaie J, Zolbanin NM, Jafari R. Mesenchymal stem cell derived-exosomes: a modern approach in translational medicine. J Transl Med. 2020;18:449.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 92]  [Cited by in F6Publishing: 203]  [Article Influence: 50.8]  [Reference Citation Analysis (0)]
26.  Magee JC, Grienberger C. Synaptic Plasticity Forms and Functions. Annu Rev Neurosci. 2020;43:95-117.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 112]  [Cited by in F6Publishing: 244]  [Article Influence: 61.0]  [Reference Citation Analysis (0)]
27.  Castellanos NP, Bajo R, Cuesta P, Villacorta-Atienza JA, Paúl N, Garcia-Prieto J, Del-Pozo F, Maestú F. Alteration and reorganization of functional networks: a new perspective in brain injury study. Front Hum Neurosci. 2011;5:90.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 21]  [Cited by in F6Publishing: 25]  [Article Influence: 1.9]  [Reference Citation Analysis (0)]
28.  Johnson EO, Zoubos AB, Soucacos PN. Regeneration and repair of peripheral nerves. Injury. 2005;Suppl 4:S24-S29.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 141]  [Cited by in F6Publishing: 133]  [Article Influence: 7.0]  [Reference Citation Analysis (0)]
29.  Li R, Li DH, Zhang HY, Wang J, Li XK, Xiao J. Growth factors-based therapeutic strategies and their underlying signaling mechanisms for peripheral nerve regeneration. Acta Pharmacol Sin. 2020;41:1289-1300.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 109]  [Cited by in F6Publishing: 92]  [Article Influence: 23.0]  [Reference Citation Analysis (0)]
30.  Menorca RM, Fussell TS, Elfar JC. Nerve physiology: mechanisms of injury and recovery. Hand Clin. 2013;29:317-330.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 417]  [Cited by in F6Publishing: 364]  [Article Influence: 33.1]  [Reference Citation Analysis (0)]
31.  Nagappan PG, Chen H, Wang DY. Neuroregeneration and plasticity: a review of the physiological mechanisms for achieving functional recovery postinjury. Mil Med Res. 2020;7:30.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 12]  [Cited by in F6Publishing: 23]  [Article Influence: 5.8]  [Reference Citation Analysis (0)]
32.  Mahar M, Cavalli V. Intrinsic mechanisms of neuronal axon regeneration. Nat Rev Neurosci. 2018;19:323-337.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 241]  [Cited by in F6Publishing: 280]  [Article Influence: 56.0]  [Reference Citation Analysis (0)]
33.  Lennington JB, Yang Z, Conover JC. Neural stem cells and the regulation of adult neurogenesis. Reprod Biol Endocrinol. 2003;1:99.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 69]  [Cited by in F6Publishing: 72]  [Article Influence: 3.4]  [Reference Citation Analysis (0)]
34.  Nocera G, Jacob C. Mechanisms of Schwann cell plasticity involved in peripheral nerve repair after injury. Cell Mol Life Sci. 2020;77:3977-3989.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 214]  [Cited by in F6Publishing: 195]  [Article Influence: 48.8]  [Reference Citation Analysis (0)]
35.  Dubový P. Schwann cells and endoneurial extracellular matrix molecules as potential cues for sorting of regenerated axons: a review. Anat Sci Int. 2004;79:198-208.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 36]  [Cited by in F6Publishing: 38]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
36.  Lopes B, Sousa P, Alvites R, Branquinho M, Sousa AC, Mendonça C, Atayde LM, Luís AL, Varejão ASP, Maurício AC. Peripheral Nerve Injury Treatments and Advances: One Health Perspective. Int J Mol Sci. 2022;23.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 17]  [Cited by in F6Publishing: 62]  [Article Influence: 31.0]  [Reference Citation Analysis (0)]
37.  Prieto González EA, Haider KH.   Genomic Instability in Stem Cells: The Basic Issues. In: Haider KH. Stem cells: From Potential to Promise. Singapore: Springer, 2021: 107-150.  [PubMed]  [DOI]  [Cited in This Article: ]
38.  Kamal M, Kassem D, Haider KH.   Sources and therapeutic strategies of mesenchymal stem cells in regenerative medicine. In: Handbook of stem cell therapy. Singapore: Springer, 2022: 1-28.  [PubMed]  [DOI]  [Cited in This Article: ]
39.  González-González A, García-Sánchez D, Dotta M, Rodríguez-Rey JC, Pérez-Campo FM. Mesenchymal stem cells secretome: The cornerstone of cell-free regenerative medicine. World J Stem Cells. 2020;12:1529-1552.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 55]  [Cited by in F6Publishing: 45]  [Article Influence: 11.3]  [Reference Citation Analysis (3)]
40.  Hosseini SM, Sani M, Haider KH, Dorvash M, Ziaee SM, Karimi A, Namavar MR. Concomitant use of mesenchymal stem cells and neural stem cells for treatment of spinal cord injury: A combo cell therapy approach. Neurosci Lett. 2018;668:138-146.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 22]  [Cited by in F6Publishing: 33]  [Article Influence: 5.5]  [Reference Citation Analysis (0)]
41.  Hosseini SM, Ziaee SM, Haider KH, Karimi A, Tabeshmehr P, Abbasi Z. Preconditioned neurons with NaB and nicorandil, a favorable source for stroke cell therapy. J Cell Biochem. 2018;119:10301-10313.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 8]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
42.  Wan T, Zhang FS, Qin MY, Jiang HR, Zhang M, Qu Y, Wang YL, Zhang PX. Growth factors: Bioactive macromolecular drugs for peripheral nerve injury treatment - Molecular mechanisms and delivery platforms. Biomed Pharmacother. 2024;170:116024.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
43.  Sandoval-Castellanos AM, Claeyssens F, Haycock JW. Biomimetic surface delivery of NGF and BDNF to enhance neurite outgrowth. Biotechnol Bioeng. 2020;117:3124-3135.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in F6Publishing: 10]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
44.  Xu H, Gao Z, Wang Z, Wu W, Li H, Liu Y, Jia S, Hao D, Zhu L. Electrospun PCL Nerve Conduit Filled with GelMA Gel for CNTF and IGF-1 Delivery in Promoting Sciatic Nerve Regeneration in Rat. ACS Biomater Sci Eng. 2023;9:6309-6321.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
45.  Gu M, Cheng X, Zhang D, Wu W, Cao Y, He J. Chemokine platelet factor 4 accelerates peripheral nerve regeneration by regulating Schwann cell activation and axon elongation. Neural Regen Res. 2024;19:190-195.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
46.  Romano R, Bucci C. Role of EGFR in the Nervous System. Cells. 2020;9.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 86]  [Cited by in F6Publishing: 76]  [Article Influence: 19.0]  [Reference Citation Analysis (0)]
47.  Cintron-Colon AF, Almeida-Alves G, VanGyseghem JM, Spitsbergen JM. GDNF to the rescue: GDNF delivery effects on motor neurons and nerves, and muscle re-innervation after peripheral nerve injuries. Neural Regen Res. 2022;17:748-753.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in F6Publishing: 20]  [Article Influence: 6.7]  [Reference Citation Analysis (0)]
48.  Ye K, He A, Wu M, Qiu X, Chen Z, Yin J, Song Q, Huang Y, Xu K, Wei P. In vitro study of decellularized rat tissues for nerve regeneration. Front Neurol. 2022;13:986377.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
49.  Slavin BR, Sarhane KA, von Guionneau N, Hanwright PJ, Qiu C, Mao HQ, Höke A, Tuffaha SH. Insulin-Like Growth Factor-1: A Promising Therapeutic Target for Peripheral Nerve Injury. Front Bioeng Biotechnol. 2021;9:695850.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 18]  [Article Influence: 6.0]  [Reference Citation Analysis (0)]
50.  Alastra G, Aloe L, Baldassarro VA, Calzà L, Cescatti M, Duskey JT, Focarete ML, Giacomini D, Giardino L, Giraldi V, Lorenzini L, Moretti M, Parmeggiani I, Sannia M, Tosi G. Nerve Growth Factor Biodelivery: A Limiting Step in Moving Toward Extensive Clinical Application? Front Neurosci. 2021;15:695592.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in F6Publishing: 5]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
51.  Li M, Xu TM, Zhang DY, Zhang XM, Rao F, Zhan SZ, Ma M, Xiong C, Chen XF, Wang YH. Nerve growth factor-basic fibroblast growth factor poly-lactide co-glycolid sustained-release microspheres and the small gap sleeve bridging technique to repair peripheral nerve injury. Neural Regen Res. 2023;18:162-169.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
52.  Golzadeh A, Mohammadi R. Effect of local administration of platelet-derived growth factor B on functional recovery of peripheral nerve regeneration: A sciatic nerve transection model. Dent Res J (Isfahan). 2016;13:225-232.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in F6Publishing: 11]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
53.  Ding Z, Jiang M, Qian J, Gu D, Bai H, Cai M, Yao D. Role of transforming growth factor-β in peripheral nerve regeneration. Neural Regen Res. 2024;19:380-386.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
54.  Akram R, Anwar H, Javed MS, Rasul A, Imran A, Malik SA, Raza C, Khan IU, Sajid F, Iman T, Sun T, Han HS, Hussain G. Axonal Regeneration: Underlying Molecular Mechanisms and Potential Therapeutic Targets. Biomedicines. 2022;10.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 10]  [Reference Citation Analysis (0)]
55.  Niknejad H, Yazdanpanah G, Ahmadiani A. Induction of apoptosis, stimulation of cell-cycle arrest and inhibition of angiogenesis make human amnion-derived cells promising sources for cell therapy of cancer. Cell Tissue Res. 2016;363:599-608.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 48]  [Cited by in F6Publishing: 51]  [Article Influence: 6.4]  [Reference Citation Analysis (0)]
56.  Zhang J, Huang X, Wang H, Liu X, Zhang T, Wang Y, Hu D. The challenges and promises of allogeneic mesenchymal stem cells for use as a cell-based therapy. Stem Cell Res Ther. 2015;6:234.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 179]  [Cited by in F6Publishing: 195]  [Article Influence: 21.7]  [Reference Citation Analysis (0)]
57.  Yang D, Wei H, Sheng Y, Peng T, Zhao Q, Xie L, Yang J. Circ_0006640 transferred by bone marrow-mesenchymal stem cell-exosomes suppresses lipopolysaccharide-induced apoptotic, inflammatory and oxidative injury in spinal cord injury. J Orthop Surg Res. 2024;19:50.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
58.  Sonbhadra S, Mehak, Pandey LM. Biogenesis, Isolation, and Detection of Exosomes and Their Potential in Therapeutics and Diagnostics. Biosensors (Basel). 2023;13.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
59.  Liu B, Qiao G, Cao W, Li CH, Pan SH, Wang L, Liu Y, Ma L, Cui D. Proteomics Analyses Reveal Functional Differences between Exosomes of Mesenchymal Stem Cells Derived from The Umbilical Cord and Those Derived from The Adipose Tissue. Cell J. 2021;23:75-84.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 3]  [Reference Citation Analysis (0)]
60.  Xu HK, Chen LJ, Zhou SN, Li YF, Xiang C. Multifunctional role of microRNAs in mesenchymal stem cell-derived exosomes in treatment of diseases. World J Stem Cells. 2020;12:1276-1294.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 21]  [Cited by in F6Publishing: 27]  [Article Influence: 6.8]  [Reference Citation Analysis (0)]
61.  Hwang J, Jang S, Kim C, Lee S, Jeong HS. Role of Stem Cell-Derived Exosomes and microRNAs in Spinal Cord Injury. Int J Mol Sci. 2023;24.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
62.  Pathan M, Fonseka P, Chitti SV, Kang T, Sanwlani R, Van Deun J, Hendrix A, Mathivanan S. Vesiclepedia 2019: a compendium of RNA, proteins, lipids and metabolites in extracellular vesicles. Nucleic Acids Res. 2019;47:D516-D519.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 313]  [Cited by in F6Publishing: 457]  [Article Influence: 114.3]  [Reference Citation Analysis (0)]
63.  van Balkom BW, Eisele AS, Pegtel DM, Bervoets S, Verhaar MC. Quantitative and qualitative analysis of small RNAs in human endothelial cells and exosomes provides insights into localized RNA processing, degradation and sorting. J Extracell Vesicles. 2015;4:26760.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 223]  [Cited by in F6Publishing: 197]  [Article Influence: 21.9]  [Reference Citation Analysis (0)]
64.  Jiang M, Wang Y, Wang J, Feng S, Wang X. The etiological roles of miRNAs, lncRNAs, and circRNAs in neuropathic pain: A narrative review. J Clin Lab Anal. 2022;36:e24592.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
65.  Rao F, Zhang D, Fang T, Lu C, Wang B, Ding X, Wei S, Zhang Y, Pi W, Xu H, Wang Y, Jiang B, Zhang P. Exosomes from Human Gingiva-Derived Mesenchymal Stem Cells Combined with Biodegradable Chitin Conduits Promote Rat Sciatic Nerve Regeneration. Stem Cells Int. 2019;2019:2546367.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 49]  [Cited by in F6Publishing: 65]  [Article Influence: 13.0]  [Reference Citation Analysis (0)]
66.  Haertinger M, Weiss T, Mann A, Tabi A, Brandel V, Radtke C. Adipose Stem Cell-Derived Extracellular Vesicles Induce Proliferation of Schwann Cells via Internalization. Cells. 2020;9.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 21]  [Cited by in F6Publishing: 27]  [Article Influence: 6.8]  [Reference Citation Analysis (0)]
67.  Wei JJ, Chen YF, Xue CL, Ma BT, Shen YM, Guan J, Bao XJ, Wu H, Han Q, Wang RZ, Zhao CH. Protection of Nerve Injury with Exosome Extracted from Mesenchymal Stem Cell. Zhongguo Yi Xue Ke Xue Yuan Xue Bao. 2016;38:33-36.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 18]  [Reference Citation Analysis (0)]
68.  Supra R, Wilson DR, Agrawal DK. Therapeutic Potential of "Smart" Exosomes in Peripheral Nerve Regeneration. J Biotechnol Biomed. 2023;6:189-196.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
69.  Haider KH, Khan M, Sen CK.   MicroRNAs with Mega Functions in Cardiac Remodeling and Repair: The Micromanagement of Matters of the Heart. In: Sen CK. MicroRNA in Regenerative Medicine. United States: Academic Press, 2015: 569-600.  [PubMed]  [DOI]  [Cited in This Article: ]
70.  Nasirishargh A, Kumar P, Ramasubramanian L, Clark K, Hao D, Lazar SV, Wang A. Exosomal microRNAs from mesenchymal stem/stromal cells: Biology and applications in neuroprotection. World J Stem Cells. 2021;13:776-794.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 16]  [Cited by in F6Publishing: 18]  [Article Influence: 6.0]  [Reference Citation Analysis (2)]
71.  Gotoh S, Kawabori M, Fujimura M. Intranasal administration of stem cell-derived exosomes for central nervous system diseases. Neural Regen Res. 2024;19:1249-1255.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
72.  Ma ZX, Liu Z, Xiong HH, Zhou ZP, Ouyang LS, Xie FK, Tang YM, Wu ZD, Feng Y. MicroRNAs: protective regulators for neuron growth and development. Neural Regen Res. 2023;18:734-745.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
73.  Li D, Zhang P, Yao X, Li H, Shen H, Li X, Wu J, Lu X. Exosomes Derived From miR-133b-Modified Mesenchymal Stem Cells Promote Recovery After Spinal Cord Injury. Front Neurosci. 2018;12:845.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 75]  [Cited by in F6Publishing: 110]  [Article Influence: 18.3]  [Reference Citation Analysis (0)]
74.  Xin H, Li Y, Buller B, Katakowski M, Zhang Y, Wang X, Shang X, Zhang ZG, Chopp M. Exosome-mediated transfer of miR-133b from multipotent mesenchymal stromal cells to neural cells contributes to neurite outgrowth. Stem Cells. 2012;30:1556-1564.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 613]  [Cited by in F6Publishing: 663]  [Article Influence: 55.3]  [Reference Citation Analysis (0)]
75.  Xiao J, Joseph S, Xia M, Teng F, Chen X, Huang R, Zhai L, Deng W. Circular RNAs Acting as miRNAs' Sponges and Their Roles in Stem Cells. J Clin Med. 2022;11.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 1]  [Article Influence: 0.5]  [Reference Citation Analysis (0)]
76.  Hade MD, Suire CN, Suo Z. Mesenchymal Stem Cell-Derived Exosomes: Applications in Regenerative Medicine. Cells. 2021;10.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 31]  [Cited by in F6Publishing: 164]  [Article Influence: 54.7]  [Reference Citation Analysis (0)]
77.  Tan F, Li X, Wang Z, Li J, Shahzad K, Zheng J. Clinical applications of stem cell-derived exosomes. Signal Transduct Target Ther. 2024;9:17.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
78.  Alvarez-Viejo M, Haider KH.   Mesenchymal stem cells. In: Haider KH. Handbook of stem cell therapy. Singapore: Springer Nature, 2022: 127-162.  [PubMed]  [DOI]  [Cited in This Article: ]
79.  Shi S, Ou X, Cheng D. Nanoparticle-Facilitated Therapy: Advancing Tools in Peripheral Nerve Regeneration. Int J Nanomedicine. 2024;19:19-34.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]