Skip to main content

Exosomes repairment for sciatic nerve injury: a cell-free therapy

Abstract

Sciatic nerve injury (SNI) is a common type of peripheral nerve injury typically resulting from trauma, such as contusion, sharp force injuries, drug injections, pelvic fractures, or hip dislocations. It leads to both sensory and motor dysfunctions, characterized by pain, numbness, loss of sensation, muscle atrophy, reduced muscle tone, and limb paralysis. These symptoms can significantly diminish a patient’s quality of life. Following SNI, Wallerian degeneration occurs, which activates various signaling pathways, inflammatory factors, and epigenetic regulators. Despite the availability of several surgical and nonsurgical treatments, their effectiveness remains suboptimal. Exosomes are extracellular vesicles with diameters ranging from 30 to 150 nm, originating from the endoplasmic reticulum. They play a crucial role in facilitating intercellular communication and have emerged as highly promising vehicles for drug delivery. Increasing evidence supports the significant potential of exosomes in repairing SNI. This review delves into the pathological progression of SNI, techniques for generating exosomes, the molecular mechanisms behind SNI recovery with exosomes, the effectiveness of combining exosomes with other approaches for SNI repair, and the changes and future outlook for utilizing exosomes in SNI recovery.

Introduction

SNI, a common peripheral nerve injury typically caused by trauma such as contusions, sharp force injuries, drug injections, pelvic fractures, or hip dislocations, leads to sensory and motor dysfunctions including pain, numbness, loss of sensation, muscle atrophy, reduced muscle tone, and limb paralysis, significantly affecting the patient’s quality of life [1,2,3,4]. Wallerian degeneration follows SNI, triggering a variety of signaling pathways, inflammatory factors, and epigenetic regulators [5,6,7]. To date, numerous approaches have been explored to treat SNI in clinical practice, including nerve tissue and stem cell transplantation, the administration of neurotrophic drugs and nerve growth factors, as well as physical and laser therapy [8, 9]. Direct nerve anastomosis is only suitable for cases involving short gaps [10]. Autologous nerve transplantation is a viable option for addressing large nerve defects, but the effectiveness of this approach, particularly when implemented using microsurgical techniques, is hindered by several challenges, including insufficient availability of nerves sources, potential donor site dysfunction and discrepancies of donor nerve and graft size [11, 12]. Although various surgical and non-surgical therapies have been approved for clinical use, their therapeutic effects remain unsatisfactory. Therefore, there exists a pressing necessity to explore novel therapeutic interventions for SNI [13].

Exosomes are endoplasm-derived extracellular vesicles, ranging from 30 to 150 nm in diameter, that encapsulate a diverse array of bioactive molecules, predominantly nucleic acids and proteins, which play an indispensable role in facilitating intercellular communication [14,15,16,17]. Recent studies have revealed that exosomes possess favorable characteristics such as good stability, biocompatibility, the ability to permeate biological barriers, low immunogenicity, therapeutic targeting, and prolonged blood circulation [18,19,20]. Genetically engineered cells can endow exosomes with new functions by equipping them with functional proteins, enhancing their capability for specific therapeutic and diagnostic applications [21, 22]. Moreover, exosomes can serve as transporters carrying small molecules or nucleic acid medications for the targeted delivery of drugs to specific cells or tissues [23,24,25]. Fayazi et al. suggests that exosomes derived from stem cells may exhibit neuroprotective capabilities in SNI by facilitating myelin regeneration and functional recovery [26]. Researchers have found that exosomes from various stem cell sources can promote the repair of damaged neural tissue [27, 28]. Exosomes are known to play a crucial role in the repair of SNI, yet there is a scarcity of research elucidating the specific mechanisms by which exosomes facilitate this repair process. Herein, this review delves into the pathological progression of SNI, techniques for generating exosomes, the molecular mechanisms behind SNI recovery with exosomes, the effectiveness of combining exosomes with other approaches for SNI repair, and the changes and future outlook for utilizing exosomes in SNI recovery.

Pathological mechanism of SNI

The pathological process following SNI primarily involves changes in the phenotype of Schwann cells (SCs) and the activation of macrophages [29]. Eliminating residual myelin is a crucial step in restoring the sciatic nerve after demyelination because residual myelin can have an inhibitory effect [30]. Various genes or proteins regulate and facilitate the phagocytosis of myelin fragments by SCs and macrophages [31,32,33]. Once myelin fragments are cleared, SCs migrate and cluster together to form myelin, which protect the axon and facilitate its repair and regeneration [34].

Phenotypic changes in SCs

SCs are a type of glial cells responsible for forming and maintaining the myelin around axons, as well as promoting axon regeneration. The peripheral nervous system (PNS) possesses an inherent regenerative ability primarily attributed to SCs [35, 36]. Researchers have demonstrated that activated SCs exhibit increased cellular uptake capacity compared to normal SCs, potentially due to changes in specific proteins such as the upregulation of RAB7A, ARF6, VPS45, and ARF1, and the downregulation of RAB11A, NEDD4, and DNM3 [37]. These findings provide new insights into the mechanisms of myelin fragment clearance and myelin restoration during the subacute phase of nerve damage. Moreover, SCs can be divided into non-myelinated and myelinated phenotypes. Myelinated SCs are the dominant glial cell type in the PNS and play a crucial role in various aspects, including the development, maintenance, regeneration, and overall functioning of peripheral nerves [38]. Non-myelinated SCs, known as Remak cells, provide support to fine caliber axons during both normal conditions and after injury, while the myelinated SCs in the distal nerve stump become dedifferentiated and activated following SNI [39]. In the process recognized as Waller degeneration, the SCs dedifferentiated into non-myelinated type, and then begin to degrade cellular debris and remaining damaged myelin [40]. Additionally, the exchange of interaction and information between dedifferentiated SCs and other cells is crucial for regeneration after a SNI. This communication involves various bioactive components, including lipids, proteins, mRNA, lncRNA and circRNA [41,42,43] (Fig. 1).

Fig. 1
figure 1

Pathological mechanisms of SNI. SCs, Schwann cells; CCL2, C C motif ligand 2; IL-1α, Interleukin-1α; IL-1β, Interleukin-1β; TNF-α, Tumor Necrosis Factor-α

Activation of macrophages

Macrophages exhibit significant variability in their behavior and can differentiate into distinct phenotypes based on cues from their surrounding environment. They can polarize into either M1 macrophage, a pro-inflammatory phenotype, which is identified by the presence of CD86 markers on their surface, or M2 macrophage, an anti-inflammatory phenotype, marked by CD206. This polarization allows macrophages to adapt and respond appropriately to different physiological conditions, playing versatile roles in the immune system’s response to various stimuli or injuries [44, 45]. The neuroinflammatory response triggers peripheral immune cells, such as circulating macrophages, to gather or accumulate at the site of injury [46].

After an injury, macrophages are key immune cells that migrate towards the site of the damage. Their role is multifaceted. Initially, they help clear the debris, such as phagocytizing myelin remnants, which is essential because these remnants can inhibit regeneration if not removed [47]. By clearing these inhibitory materials, macrophages prevent them from suppressing the signals that encourage regrowth and repair [48]. Additionally, macrophages switch roles from being primarily phagocytic to promoting regeneration. They release factors that stimulate the regrowth of axons, the long nerve fibers that are often damaged during injuries [49]. This switch is crucial for the transition from an inflammatory state, which is necessary for cleaning up the site of injury, to a regenerative state, which is essential for healing and the restoration of function [47,48,49]. Thus, macrophages are indispensable not only for their role in debris clearance but also in facilitating and promoting the healing and regrowth of nerve tissues post-injury.

Interaction between SCs and macrophages

SCs play a pivotal role in the recruitment of macrophages to the site of nerve injury through the secretion of various ligands, including cytokines and chemokines. These signaling molecules are crucial for initiating and guiding the immune response following nerve damage. Key cytokines and chemokines released by SCs include: firstly, C C motif ligand 2 (CCL2): This chemokine is particularly important for attracting monocytes, which can differentiate into macrophages, to the injury site. Secondly, IL-1α and IL-1β: these interleukins are pro-inflammatory cytokines that help in amplifying the inflammatory response, which is essential for clearing the damaged tissue and preparing for regeneration. Thirdly, TNF-α: tumor necrosis factor-alpha is another pro-inflammatory cytokine that promotes inflammation and aids in the recruitment and activation of macrophages. By secreting these molecules, SCs actively contribute to the inflammatory environment needed for healing and regeneration, signaling macrophages to come to the injury site where they can perform their crucial roles in debris clearance and tissue repair [50,51,52,53]. Fourth, SCs-derived gel protein [54] and periosteal protein [55] are also important in promoting macrophage motility and infiltration into the injury site in vivo. Fifth, in Schwan cell-derived exosomes (SCs-Exo), a lncRNA called axon regeneration-associated transcription is upregulated after SNI to enhance axon regeneration and promote recovery of motor function through recruitment and activation of pro-regenerative macrophages [56]. Meanwhile, macrophages also play a crucial role in regulating the maturation process of SCs after nerve injury. Macrophages express a protein called Growth arrest-specific 6 (Gas6) that is involved in controlling the function of SCs. The absence of Gas6 within macrophages leads to an altered response from SCs and impairs their ability to effectively remyelinate [57]. Moreover, microvesicles released by CD206 + macrophages aid in promoting migration and proliferation of SCs [58]. Therefore, this interaction between macrophages and SCs is essential for promoting proper nerve regeneration.

Preparation and functions of exosomes

Preparation

In the realm of exosomes research and their practical application, the identification and extraction techniques of exosomes form the basis of the study. Although it presents a challenge to achieve complete isolation of exosomes from other types of extracellular vesicles, a number of standardized methods for the identification and extraction of exosomes have been established and are used by researchers. Various techniques can be employed to isolate exosomes from cell cultures or body fluids, depending on their size and source. To date, researchers have developed five types of exosome separation techniques: Ultra-high-speed centrifugation, size-based separation technology, in situ polymer precipitation, immunoaffinity capture technique and, more recently, microfluidic derived technology [59]. The most common separation methods used in exosome studies are ultra-high speed centrifugation and exosome extraction kits based on polyethylene glycol precipitation [60]. Various techniques have been utilized in the pursuit of comprehending the constitution of exosomes. These methods encompass transport electron microscopy, scanning electron microscopy, cryo-electron microscope, atomic force microscopy, nanoparticle tracking analysis, dynamic light scattering, trypsin digestion, mass spectrometry, enzyme linked immunosorbent assay analysis, labeled protein expression detection such as Western blot analysis and flow cytometry and these approaches rely on the scrutiny of exosome morphology, particle size, and surface markers for the purpose of exosome identification [61,62,63].

Functions

Researchers are constantly collecting and analyzing exosomes from various biopsies to discover the dynamic properties and molecular features of exosomes and to exploit the powerful functions of exosomes. In general, exosomes have diagnostic, therapeutic and prognostic effects. The inherent architecture and distinctive cellular functionalities of exosomes render them exceptionally promising as conveyors of natural medication or genetic material, possessing substantial diagnostic capabilities within the realms of regenerative medicine, immunotherapy, and vaccination experiments. However, to maximize their potential, it is imperative to establish dependable and effective techniques for isolating these vesicles [64]. Exosomes exhibit significant potential as diagnostic biomarkers for a range of medical conditions, including cardiovascular diseases, Parkinson’s disease, other neurodegenerative diseases such as Alzheimer’s disease and cancer [65]. Treatment strategies for exosomes can be classified into three categories, namely direct interventions, indirect interventions, and alternative therapies. Direct therapy takes advantage of exosomes’ ability to transfer proteins and nucleic acids between cells, using exosomes as carriers of drugs to treat diseases or injuries [65]. Exosomes serve as biomarkers when employing indirect techniques. Alternative strategies seek to impede the advancement of diseases by interrupting the circulation of exosomes that also include harmful disease-associated cargo such as viral miRNA and proteins [66], immunosuppressive factors promoting tumor metastasis [67] or tumor signaling molecules impeding therapeutic medications [68]. An increasing body of research has demonstrated that exosomes play a critical role in the process of tumorigenesis, growth, metastasis and drug resistance [69]. The loading of exosomes originating from tumors aligns with the genetic composition of parental tumor cells [70]. Furthermore, exosomes exhibit remarkable stability in circulation, effectively safeguarding their contents against degradation [71]. Therefore, exosomes and their transport products are beginning to be considered as novel biomarkers for cancer prognosis assessment.

Exosomes for the treatment of SNI

Stem cell-derived exosomes repair SNI

ADSCs-Exo

Advancing research into the mechanisms of sciatic nerve regeneration, identifying novel therapeutic targets, and exploring innovative treatment methods are crucial steps toward enhancing the therapy for SNI. Recent research has shown that adipose-derived stem cells (ADSCs), when cultured with SCs, can enhance myelin-related markers, playing a significant role in nerve injury repair and regeneration [72]. In their in vitro studies, Wang and colleagues discovered that a hypocapnic environment stimulates adipose-derived stem cells (ADSCs) to release smaller exosomes, which more efficiently concentrate and transport intracellular miR-218 to recipient cells. Subsequent in vivo studies demonstrated that this process further accelerates SNI healing by enhancing myelination, promoting tissue regeneration, and restoring motor function [73]. Chen and colleagues evaluated the efficacy of ADSCs-Exo in a rat model of sciatic nerve transection with a 10 mm gap. Compared to the control group, the ADSCs-Exo treatment group exhibited significant improvements in axon regeneration, myelination, and recovery from denervated muscle atrophy [74]. FK506, an immunosuppressant known to promote nerve regeneration, was studied for its effects when combined with ADSCs-Exo after treatment. In a murine model of sciatic nerve compression injury, the local application of these ADSCs-Exo significantly reduced macrophage autophagy following spinal segmental nerve compression injury [75]. Proteomic analysis of ADSCs-Exo revealed that HSPA8 and EEF1A1 show promising potential in reducing autophagy through exosome mediation [75]. Additionally, HDAC, ITGB1, and APP have been identified as potential candidates involved in the exosome-mediated enhancement of nerve regeneration [76]. Furthermore, Yin and colleagues administered ADSCs-Exo to rats with SNI via tail vein injection. Their results indicate that ADSCs-Exo positively influence neuronal axon regeneration by reducing SCs apoptosis, decreasing autophagy, and encouraging Bungner ligament formation, thereby promoting fiber regeneration following SNI [77]. They also found that ADSCs-Exo moderately reduced autophagy in damaged SCs by downregulating Kpna2 through miRNA-26b, facilitating myelin regeneration [78]. ADSCs-Exo can promote SNI regeneration through up-regulating the expression of Bcl-2 mRNA and down-regulating the expression of Bax mRNA, thereby reducing cell apoptosis and promoting the proliferation of SCs [42].

Mesenchymal stem cell-derived exosomes (MSCs-Exo)

Mesenchymal stem cells (MSCs) are pluripotent stem cells that can be derived from kinds of sources such as bone marrow, umbilical cord, adipose tissue and dental pulp [79]. Akyurekli et al. reported that the therapeutic efficacy of MSCs appears to their paracrine factors [80]. Exosomes are now recognized as crucial paracrine mediators of MSCs, facilitating intercellular communication and preserving a dynamic, homeostatic microenvironment that supports tissue regeneration [81, 82]. Compared to MSCs, MSCs-Exo are easier to collect and store, and they face fewer ethical constraints [83]. MSCs-Exo, as a cell derivative, may play a crucial role in recovery after nerve injury [84].

Several researches have reported on the use of MSCs-Exo in the repairment of SNI in recent years [85,86,87,88,89]. Local injections of LPS pre-MSCs-Exo at nerve injury sites were found to deactivate the NF-κB/NLRP3 signaling axis by transferring TSG-6, promoting macrophage polarization to the M2 phenotype, thereby encouraging axon and myelin regeneration and enhancing nerve function recovery [85]. Zhang et al. found that MSCs-Exo carrying miR-181c-5p can reduce neuropathic pain induced by chronic compression of SCI by inhibiting neurogenic inflammation [86]. In a 12 mm sciatic nerve defect rat model, Zhang et al. found that human umbilical cord mesenchymal stem cells-derived exosomes (HUCMSCs-Exo) enhance the survival and migratory capacity of olfactory ensheathing cells under hypoxic conditions via activation of the BDNF signaling pathway, and the combined use of olfactory sensory cells and HUCMSCs-Exo can aid in restoring motor and sensory function after SNI [87]. Additionally, exosomes derived from bone marrow mesenchymal stem cells (BMSCs-Exo) can facilitate sciatic nerve regeneration, potentially through miRNA regulation of regenerative genes like VEGFA and S100b [88]. Moreover, studies on exosomes secreted by adipose-derived mesenchymal stem cells (ADMSCs-Exo) after acute sciatic neurotomy have shown that, compared to the control group, the exosome transplantation group exhibited increased axonal regeneration, improved function, and a higher quantity of regenerated nerve fibers due to the presence of signaling proteins like glial cell-derived neurotrophic factor, fibroblast growth factor-1, brain-derived neurotrophic factor, insulin-like growth factor-1, and nerve growth factor, which promote nerve survival and axon growth [89].

MSCs-Exo could present both challenges and opportunities in advancing regenerative strategies. Thus, comprehensive research is essential to unravel the mechanisms of MSCs-Exo in treating SNI while also evaluating their efficacy and safety.

MDSCs-Exo

Muscle-derived stem cell exosomes (MDSCs-Exo) overexpressing miR-214 significantly promote SCs proliferation and migration, increase neurotrophic factor levels in dorsal root ganglion neurons, stimulate axonal growth, and support the regeneration of neural structures after peripheral nerve injury by inhibiting PTEN protein expression and activating the JAK2/STAT3 signaling pathway [90]. However, research on using MDSCs-Exo for SNI repair is limited, and further studies are required to evaluate the safety and efficacy of this treatment approach.

Schwann cell-derived exosomes (SCs-Exo)

SCs are a type of glial cell found in the PNS, and they play a vital role in maintaining neural homeostasis and promoting regeneration following peripheral nerve injury [91]. SCs supply nutrients to aid in axon regeneration and are the primary cell type responsible for myelin formation along the axon [92]. SCs-Exo, a second-generation derivative containing crucial informational substances from SCs, have been shown to play a significant role in neurodegeneration, neurodevelopment, and neuroprotection [93, 94].

SCs-Exo have been found to significantly enhance axon regeneration after SNI in vitro and in vivo. They induce alterations in growth cone morphology by reducing the activity of the GTPase RhoA, which is associated with growth cone collapse and axon retraction [95]. In a rat model of sciatic nerve compression, Sun and colleagues found that the ischemic and hypoxic microenvironment following SNI significantly reduced miR-146a-5p levels in SCs-Exo, prompting the conversion of M2 macrophages to M1 macrophages and thereby impeding axon regeneration and functional recovery [96]. Chen and colleagues created an animal model for sciatic nerve transection and demonstrated that Schwann cell-like cells (SCLCs) derived from human amniotic mesenchymal stem cells facilitate sciatic nerve recovery via the exosome-induced SOX2/FN1 signaling pathway [97]. Experiments showed that nerve grafts containing SCLCs-derived exosomes (SCLCs-Exo) significantly enhance sciatic nerve function recovery, reduce gastrocnemius atrophy, and promote axonal growth and myelination. Furthermore, SCLCs-Exo can stimulate SCs to migrate, proliferate, form myelin, and secrete neurotrophic factors [98].

Therefore, these findings deepen our understanding of how SCs-Exo contribute to repairing SNI, offering promising application prospects and valuable insights for their clinical use. Nevertheless, further research is required to fully realize the clinical potential of SCs-Exo in treating SNI patients.

Blood-derived exosomes repair SNI

In recent years, a few studies have documented the use of blood-derived exosomes to treat SNI, though their precise mechanisms remain unclear and are likely to be the focus of future research. The findings indicate that the elevated expression of miR-21 in blood-derived exosomes following sciatic nerve ligation may partially contribute to reducing neuropathic pain [99]. Besides, Platelet-rich plasma-derived exosomes (PRP-Exo), harvested from platelet-rich plasma, exhibit tissue healing abilities and have been noted for their efficacy in addressing injuries in both the central and peripheral nervous systems [100, 101]. In a rat model of SNI, using ultrasound to target and destroy microvesicles enhances the delivery of PRP-Exo to the injury site, significantly concentrating PRP-Exo in the damaged nerve and improving therapeutic outcomes [102].

Mechanisms of exosomes repairing SNI

Exosomes are small extracellular vesicles that play a crucial role in intercellular communication and have been widely studied for their potential in regenerative medicine, including nerve regeneration. Here’s a summary of how various stem cell-derived exosomes contribute to sciatic nerve recovery through different mechanisms (Table 1).

Table 1 Mechanisms of exosomes to repair SNI

Adipose Stem Cell-derived Exosomes (ADSCs-Exo) facilitate sciatic nerve recovery by modulating key signaling pathways, including down-regulating Kpna2, up-regulating Bcl-2 mRNA (which promotes cell survival), and down-regulating Bax mRNA (which promotes apoptosis) [42, 78].

Muscle-derived stem cells exosomes (MDSCs-Exo) can target and inhibit PTEN protein expression, a known negative regulator of the PI3K/Akt pathway, and activate the JAK2/STAT3 signaling pathway, both of which are critical for cell survival and proliferation [90].

MSCs-Exo affects axonal and myelin regeneration through multiple signaling pathways. LPS pre-MSCs-Exo focus on the inactivation of the NF-κB/NLRP3 signaling axis by transferring TNF-stimulating gene 6 (TSG-6) [85]. This action helps reduce inflammation, which is a significant factor in nerve damage and recovery. Besides, HUCMSCs-Exo could activate the brain-derived neurotrophic factor (BDNF) signaling, and play a crucial role for nerve growth and survival [87]. Moreover, BMSCs-Exo promote both vascular and neuronal growth through regulating the expression of VEGFA and S100b [88].

SC-Exo stimulate nerve regeneration by SOX2/FN1 axis, up-regulating SOX10, OCT-6, EGR2, MBP and MPI [97].

Overall, these exosomes harness various biological mechanisms to support nerve repair and regeneration, offering promising therapeutic avenues for conditions like SNI (Fig. 2).

Fig. 2
figure 2

Mechanisms associated with exosome repair of SNI. ADSCs-Exo, Adipose stem cell-derived exosomes; MDSCs-Exo, Muscle-derived stem cell-derived exosomes; MSCs-Exo, Mesenchymal stem cell-derived exosomes; LPS pre-MSCs-Exo, Lipopolysaccharide-treated mesenchymal stem cell-derived exosomes; HUCMSCs-Exo, Human umbilical cord mesenchymal stem cell-derived exosomes; BMSCs-Exo, Bone marrow mesenchymal stem cell-derived exosomes; SCs-Exo, Schwann cells-derived exosomes

Exosomes combined with other methods to repair SNI

Exosome combined with chitin catheter

Chitin catheters are ideal biomaterials for the construction of nerve catheters due to their good biodegradability, high antibacterial activity, high biocompatibility and non-toxicity [103]. Small-gap tubulization has considerable advantages over epineural sutures in terms of shortening the operative duration, enhancing the precision of axon docking and reducing the formation of neuromas [104]. However, hollow chitin catheters lack the biomechanical support and growth factors needed for nerve regeneration. On the contrary, exosomes can supply these critical factors, and when used in combination with chitin catheters, they enhance nerve regeneration by providing essential growth factors [105]. To increase the speed of nerve regeneration, it is critical to use a suitable transport medium. The chitin catheter effectively retains the bioactivity and bioavailability of exosomes while also demonstrating a strong affinity towards exosome binding.

Li et al. enhanced chitin catheters by incorporating polydopamine loaded with MSCs-Exo to bridge sciatic nerve defects measuring 2 mm in rats, and observed that the modified chitin catheter facilitated the sustained release of exosomes, which contributed to accelerated nerve healing and improved nerve function [105]. Rao et al. used a chitin catheter to transport exosomes and repair sciatic nerve defects in rats, discovering that the combined application of chitin catheters and gingival mesenchymal stem cell-derived exosomes (GMSCs-Exo) significantly enhanced nerve fiber quantity and caliber, promoted myelin formation, and improved nerve conduction and limb motor function [106]. Consequently, they advocate that the novel and effective integration of GMSCs-Exo with biodegradable chitin catheters represents a promising strategy for peripheral nerve repair [106]. Namini et al. facilitated axon regeneration and enhanced functional recovery of a sciatic nerve defect in rats by incorporating human endometrial stem cell-derived exosomes into the nuclear sheath of a fibroin/polylactic acid nerve catheter [84]. Yang et al. reported the therapeutic effect of a nerve guidance catheter (NGC) as an exosome carrier for the repair of SNI in rats. They initially encapsulated neurotrophin-3 into exosomes derived from ADSCs (ADSCs-ExoNT-3), which were then loaded into the NGC to create the ADSCs-ExoNT-3-NGC for bridging sciatic nerve defects in rats. After implantation of the NGC in vivo, ADSCs-ExoNT-3-NGC was found to significantly promote sural nerve regeneration and improve gastrocnemius function [107]. Generally, the chitin catheter, known for its good biocompatibility and degradability, offers an optimal biological microenvironment for the gradual release of exosomes, bridging nerve defects, and facilitating nerve regeneration.

Exosomes carrying drugs

At present, there are no documented instances of using exosomes for drug delivery to the sciatic nerve. However, theoretically, it remains a viable method. Moreover, researchers have been modified exosomes to use them as therapeutic drug carriers due to their relatively small molecular structure, natural molecular transport properties, good biocompatibility, and ability to penetrate blood-spinal cord barrier [108, 109]. Zeng et al. [110], Gao et al. [111], and Yue et al. [112] demonstrated that exosome-based drugs, including azide-modified Ile-Lys-Val-Ala-Val peptide, berberine, and resveratrol, can effectively repair spinal cord injuries by reducing inflammation, promoting neuronal differentiation, and enhancing drug delivery across the blood-brain barrier. Exosomes, as drug carriers, offer numerous benefits. They enhance drug stability within the body, improve solubility, enable targeted delivery, and facilitate the transport of drugs across both the blood-brain and blood-spinal cord barriers. Consequently, exosomes represent a promising drug delivery system for PNI repair.

Exosomes combined with hydrogels

Hydrogel, known for its high-water content and diverse physical properties, is a type of polymer material. Recently, Liu et al. constructed a rat model of SNI and found that compared with hard hydrogels, soft hydrogels can better inhibit the inflammatory response caused by PNI through the rapid release of exosomes and promote nerve regeneration [113]. Furthermore, the study revealed that the release of exosomes, which is regulated by the hardness of the hydrogels, plays a crucial role in tissue repair, offering valuable insights for the clinical use of hydrogels [113]. In vitro studies by Yang et al. have shown that electricity conductive hydrogels (ECH) loaded with BMSCs-Exo can promote Schwann cell attachment and migration. Moreover, the exosomes in the system can regulate M2 macrophage polarization through the NF-κB pathway, thus mitigating inflammatory pain in diabetic PNI. Further, both in vitro and in vivo experiments showed that ECH loaded with BMSCs-Exo promotes myelin axon regeneration through the MEK/ERK signaling pathway, improving muscle denervation atrophy and aiding functional recovery [114]. Therefore, the combination of exosomes with hydrogel presents a promising strategy for the repairment of sciatic nerve injuries (Fig. 3).

Fig. 3
figure 3

Exosomes combine with other strategies, mainly including chitin catheter, drugs and hydrogel, to repair SNI.SCs, Schwann cells

Challenges faced by exosome-mediated repair of SNI

Low extraction yield

Due to the shortcomings of various methods for isolating exosomes, standardizing these techniques is crucial, yet there is currently no consensus. Significant differences in RNA and protein composition are observed across different isolation methods, highlighting the need for standardization of exosome composition for clinical applications [115]. Although ultracentrifugation is the predominant technique used in scientific research, exosomes isolated by this method tend to be less pure and may include other extracellular vesicles of similar diameter [116]. In addition, isolating exosomes from biological fluids or cell culture supernatants often results in a low yield [117,118,119].

Low targeting

Optical imaging showed rapid accumulation of exosomes in the liver and spleen after intravenous injection on account of the presence of blood spinal cord barrier and blood brain barrier, indicating their unexpected biodistribution and short half-life, with exosomes having a much shorter half-life than nanocarriers (60 min versus several hours) [120]. Exosomes are also quickly eliminated by glomerular filtration, bile excretion and phagocytosis of the reticuloendothelial system [121], so the targeting of exosomes is very low. The incapacity to accurately characterize and quantify the relevant cargo carried by exosomes, as well as the incapacity to target them against specific receptors, leads to an off-target problem [40]. Low targeting and off-target lead to more rapid degradation of the drug and less effective treatment [122].

Other challenges

Exosomes face further challenges in the treatment of sciatic nerve damage. Firstly, the storage, transportation, and preservation of exosomes are critical. Secondly, various issues related to exosome sources and optimal culture conditions need to be resolved, including isolation, purification, replication, and the ideal dose, frequency, and method of administration of exosomes [83]. Third, the contents of exosomes require further study to identify which components have therapeutic potential for treating SNI and which may be harmful. Fourth, it is crucial to conduct more studies to explore the relationship between the frequency of injections, dosage, and the therapeutic efficacy of exosomes, and to determine whether single or multiple administrations could have adverse effects, all in order to sustain long-term efficacy [108] (Fig. 4).

Fig. 4
figure 4

The dilemma facing exosome repair of SNI, such as low targeting, low mounts and hard storage

Conclusions

Currently, there is no definitive and effective treatment method that can fully restore nerve function after SNI. Exosomes sourced from various origins could aid in SNI repair by modulating multiple signaling pathways, including Kpna2, Bcl-2 mRNA/Bax mRNA, PTEN, JAK2/STAT3, TSG-6/NF-κΒ/NLRP3, BDNF, and VEGFA/S100, among others. Additionally, innovative approaches such as engineered exosomes, exosomes combined with chitin catheters, and exosomes paired with hydrogels show great promise in repairing SNI and are key focuses of ongoing research. However, there are substantial challenges in using exosomes for SNI repair, including low extraction yields, poor targeting accuracy, and challenges in achieving long-term drug delivery. The current research provides a robust theoretical basis for the preclinical exploration and practical application of exosomes in SNI treatment, offering significant potential for advancements. In summary, utilizing exosomes is proving to be an effective method for repairing SNI. In the future, comparing the effects of exosome treatment with other methods for SNI will be a new research direction.

Data availability

Not applicable.

Abbreviations

SNI:

Sciatic nerve injury

PNS:

Peripheral nervous system

SCs:

Schwann cells

ADSCs:

Adipose-derived stem cells

MDSCs:

Muscle-derived stem cells

MSCs:

Mesenchymal stem cells

SCs-Exo:

Schwan cell-derived exosomes

ADSCs-Exo:

Adipose stem cell-derived exosomes

MDSCs-Exo:

Muscle-derived stem cell-derived exosomes

LPS pre-MSCs-Exo:

Lipopolysaccharide-treated mesenchymal stem cell-derived exosomes

HUCMSCs-Exo:

Human umbilical cord mesenchymal stem cell-derived exosomes

BMSCs-Exo:

Bone marrow mesenchymal stem cell-derived exosomes

SCLCs-Exo:

Schwann cell like cells-derived exosomes

PRP-Exo:

Platelet-rich plasma-derived exosomes

GMSCs-Exo:

Gingival mesenchymal stem cell-derived exosomes

ADMSCs-Exo:

Adipose-derived mesenchymal stem cells

ADSCs-ExoNT−3 :

Neurotrophin-3 into exosomes secreted from ADSCs

Gas6:

Growth arrest-specific 6

NGC:

Nerve guidance catheter

ECH:

Electricity conductive hydrogels

TSG-6:

TNF-stimulating gene 6

References

  1. Gordon T, Borschel GH. The use of the rat as a model for studying peripheral nerve regeneration and sprouting after complete and partial nerve injuries. Exp Neurol Jan. 2017;287(Pt 3):331–47. https://doi.org/10.1016/j.expneurol.2016.01.014.

    Article  CAS  Google Scholar 

  2. Diogo CC, Camassa JA, Pereira JE, et al. The use of sheep as a model for studying peripheral nerve regeneration following nerve injury: review of the literature. Neurol Res Oct. 2017;39(10):926–39. https://doi.org/10.1080/01616412.2017.1331873.

    Article  Google Scholar 

  3. Welch JA. Peripheral nerve injury. Semin Vet Med Surg Small Anim Nov. 1996;11(4):273–84. https://doi.org/10.1016/s1096-2867(96)80020-x.

    Article  CAS  Google Scholar 

  4. Guo D, Lu X, Xu X, et al. Therapeutic effect of Vinorine on sciatic nerve injured rat. Neurochem Res Feb. 2018;43(2):375–86. https://doi.org/10.1007/s11064-017-2432-4.

    Article  CAS  Google Scholar 

  5. Yi S, Tang X, Yu J, Liu J, Ding F, Gu X. Microarray and qPCR analyses of Wallerian Degeneration in Rat sciatic nerves. Front Cell Neurosci. 2017;11:22. https://doi.org/10.3389/fncel.2017.00022.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Yu J, Gu X, Yi S. Ingenuity Pathway Analysis of Gene expression profiles in distal nerve stump following nerve Injury: insights into Wallerian Degeneration. Front Cell Neurosci. 2016;10:274. https://doi.org/10.3389/fncel.2016.00274.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Fawcett JW, Keynes RJ. Peripheral nerve regeneration. Annu Rev Neurosci. 1990;13:43–60. https://doi.org/10.1146/annurev.ne.13.030190.000355.

    Article  CAS  PubMed  Google Scholar 

  8. Sayad-Fathi S, Nasiri E, Zaminy A. Advances in stem cell treatment for sciatic nerve injury. Expert Opin Biol Ther Apr. 2019;19(4):301–11. https://doi.org/10.1080/14712598.2019.1576630.

    Article  Google Scholar 

  9. Sasso LL, de Souza LG, Girasol CE, Marcolino AM, de Jesus Guirro RR, Barbosa RI. Photobiomodulation in sciatic nerve crush injuries in rodents: a systematic review of the literature and perspectives for clinical treatment. J Lasers Med Sci Summer. 2020;11(3):332–44. https://doi.org/10.34172/jlms.2020.54.

    Article  Google Scholar 

  10. Li R, Liu Z, Pan Y, Chen L, Zhang Z, Lu L. Peripheral nerve injuries treatment: a systematic review. Cell Biochem Biophys Apr. 2014;68(3):449–54. https://doi.org/10.1007/s12013-013-9742-1.

    Article  CAS  Google Scholar 

  11. Ray WZ, Mackinnon SE. Management of nerve gaps: autografts, allografts, nerve transfers, and end-to-side neurorrhaphy. Exp Neurol May. 2010;223(1):77–85. https://doi.org/10.1016/j.expneurol.2009.03.031.

    Article  Google Scholar 

  12. Singh A, Asikainen S, Teotia AK, et al. Biomimetic Photocurable three-dimensional printed nerve Guidance channels with aligned Cryomatrix Lumen for Peripheral nerve regeneration. ACS Appl Mater Interfaces Dec. 2018;19(50):43327–42. https://doi.org/10.1021/acsami.8b11677.

    Article  CAS  Google Scholar 

  13. Zhang RC, Du WQ, Zhang JY, et al. Mesenchymal stem cell treatment for peripheral nerve injury: a narrative review. Neural Regen Res Nov. 2021;16(11):2170–6. https://doi.org/10.4103/1673-5374.310941.

    Article  CAS  Google Scholar 

  14. Pegtel DM, Gould SJ, Exosomes. Annu Rev Biochem Jun. 2019;20:88:487–514. https://doi.org/10.1146/annurev-biochem-013118-111902.

    Article  CAS  Google Scholar 

  15. Kalluri R, LeBleu VS. The biology, function, and biomedical applications of exosomes. Sci Feb. 2020;7(6478). https://doi.org/10.1126/science.aau6977.

  16. Tkach M, Théry C. Communication by Extracellular vesicles: where we are and where we need to go. Cell Mar 10. 2016;164(6):1226–32. https://doi.org/10.1016/j.cell.2016.01.043.

    Article  CAS  Google Scholar 

  17. Yu H, Huang Y, Yang L. Research progress in the use of mesenchymal stem cells and their derived exosomes in the treatment of osteoarthritis. Ageing Res Rev Sep. 2022;80:101684. https://doi.org/10.1016/j.arr.2022.101684.

    Article  CAS  Google Scholar 

  18. Jiang XC, Zhang T, Gao JQ. The in vivo fate and targeting engineering of crossover vesicle-based gene delivery system. Adv Drug Deliv Rev Aug. 2022;187:114324. https://doi.org/10.1016/j.addr.2022.114324.

    Article  CAS  Google Scholar 

  19. Yang LT, Patel KD, Rathnam C, et al. Harnessing the therapeutic potential of Extracellular vesicles for Biomedical Applications using multifunctional magnetic nanomaterials. Small Apr. 2022;18(13):2104783. https://doi.org/10.1002/smll.202104783.

    Article  CAS  Google Scholar 

  20. Hessvik NP, Llorente A. Current knowledge on exosome biogenesis and release. Cell Mol Life Sci Jan. 2018;75(2):193–208. https://doi.org/10.1007/s00018-017-2595-9.

    Article  CAS  Google Scholar 

  21. Liang G, Kan S, Zhu Y, Feng S, Feng W, Gao S. Engineered exosome-mediated delivery of functionally active miR-26a and its enhanced suppression effect in HepG2 cells. Int J Nanomed. 2018;13:585–99. https://doi.org/10.2147/ijn.S154458.

    Article  CAS  Google Scholar 

  22. Ye Y, Zhang X, Xie F, et al. An engineered exosome for delivering sgRNA:Cas9 ribonucleoprotein complex and genome editing in recipient cells. Biomater Sci May. 2020;19(10):2966–76. https://doi.org/10.1039/d0bm00427h.

    Article  CAS  Google Scholar 

  23. Luan X, Sansanaphongpricha K, Myers I, Chen H, Yuan H, Sun D. Engineering exosomes as refined biological nanoplatforms for drug delivery. Acta Pharmacol Sin Jun. 2017;38(6):754–63. https://doi.org/10.1038/aps.2017.12.

    Article  CAS  Google Scholar 

  24. Antimisiaris SG, Mourtas S, Marazioti A. Exosomes and exosome-inspired vesicles for targeted drug delivery. Pharm Nov. 2018;6(4). https://doi.org/10.3390/pharmaceutics10040218.

  25. Peng H, Ji W, Zhao R, et al. Exosome: a significant nano-scale drug delivery carrier. J Mater Chem B Sep. 2020;14(34):7591–608. https://doi.org/10.1039/d0tb01499k.

    Article  CAS  Google Scholar 

  26. Fayazi N, Sheykhhasan M, Soleimani Asl S, Najafi R. Stem cell-derived exosomes: a new strategy of neurodegenerative Disease treatment. Mol Neurobiol Jul. 2021;58(7):3494–514. https://doi.org/10.1007/s12035-021-02324-x.

    Article  CAS  Google Scholar 

  27. Hervera A, De Virgiliis F, Palmisano I, et al. Reactive oxygen species regulate axonal regeneration through the release of exosomal NADPH oxidase 2 complexes into injured axons. Nat Cell Biol Mar. 2018;20(3):307–19. https://doi.org/10.1038/s41556-018-0039-x.

    Article  CAS  Google Scholar 

  28. Xu B, Zhang Y, Du XF, et al. Neurons secrete mir-132-containing exosomes to regulate brain vascular integrity. Cell Res Jul. 2017;27(7):882–97. https://doi.org/10.1038/cr.2017.62.

    Article  CAS  Google Scholar 

  29. Dong R, Liu Y, Yang Y, Wang H, Xu Y, Zhang Z. MSC-Derived exosomes-based therapy for peripheral nerve Injury: a Novel Therapeutic Strategy. Biomed Res Int. 2019;2019:6458237. https://doi.org/10.1155/2019/6458237.

    Article  CAS  PubMed  Google Scholar 

  30. Weiss T, Taschner-Mandl S, Bileck A, et al. Proteomics and transcriptomics of peripheral nerve tissue and cells unravel new aspects of the human Schwann cell repair phenotype. Glia Dec. 2016;64(12):2133–53. https://doi.org/10.1002/glia.23045.

    Article  Google Scholar 

  31. Forese MG, Pellegatta M, Canevazzi P, et al. Prostaglandin D2 synthase modulates macrophage activity and accumulation in injured peripheral nerves. Glia Jan. 2020;68(1):95–110. https://doi.org/10.1002/glia.23705.

    Article  Google Scholar 

  32. Brosius Lutz A, Chung WS, Sloan SA, et al. Schwann cells use TAM receptor-mediated phagocytosis in addition to autophagy to clear myelin in a mouse model of nerve injury. Proc Natl Acad Sci U S Sep. 2017;19(38):E8072–80. https://doi.org/10.1073/pnas.1710566114.

    Article  CAS  Google Scholar 

  33. Scheib JL, Höke A. An attenuated immune response by Schwann cells and macrophages inhibits nerve regeneration in aged rats. Neurobiol Aging Sep. 2016;45:1–9. https://doi.org/10.1016/j.neurobiolaging.2016.05.004.

    Article  CAS  Google Scholar 

  34. Church JS, Milich LM, Lerch JK, Popovich PG, McTigue DM. E6020, a synthetic TLR4 agonist, accelerates myelin debris clearance, Schwann cell infiltration, and remyelination in the rat spinal cord. Glia Jun. 2017;65(6):883–99. https://doi.org/10.1002/glia.23132.

    Article  Google Scholar 

  35. Scheib J, Höke A. Advances in peripheral nerve regeneration. Nat Rev Neurol Dec. 2013;9(12):668–76. https://doi.org/10.1038/nrneurol.2013.227.

    Article  CAS  Google Scholar 

  36. Jessen KR, Mirsky R. The repair Schwann cell and its function in regenerating nerves. J Physiol Jul 1. 2016;594(13):3521–31. https://doi.org/10.1113/jp270874.

    Article  CAS  Google Scholar 

  37. Shi G, Hao D, Zhang L, et al. Endocytosis-associated patterns in nerve regeneration after peripheral nerve injury. J Orthop Translat Nov. 2021;31:10–9. https://doi.org/10.1016/j.jot.2021.09.004.

    Article  Google Scholar 

  38. Jessen KR, Mirsky R, Lloyd AC. Schwann cells: development and role in nerve repair. Cold Spring Harb Perspect Biol May. 2015;8(7):a020487. https://doi.org/10.1101/cshperspect.a020487.

    Article  Google Scholar 

  39. Oliveira JT, Yanick C, Wein N, Gomez Limia CE. Neuron-Schwann cell interactions in peripheral nervous system homeostasis, disease, and preclinical treatment. Front Cell Neurosci. 2023;17:1248922. https://doi.org/10.3389/fncel.2023.1248922.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Namini MS, Daneshimehr F, Beheshtizadeh N, et al. Cell-free therapy based on extracellular vesicles: a promising therapeutic strategy for peripheral nerve injury. Stem Cell Res Ther Sep. 2023;19(1):254. https://doi.org/10.1186/s13287-023-03467-5.

    Article  CAS  Google Scholar 

  41. López-Guerrero JA, de la Nuez C, Praena B, Sánchez-León E, Krummenacher C, Bello-Morales R. Herpes Simplex Virus 1 spread in oligodendrocytic cells is highly dependent on MAL Proteolipid. J Virol Jan. 2020;31(4). https://doi.org/10.1128/jvi.01739-19.

  42. Liu CY, Yin G, Sun YD, et al. Effect of exosomes from adipose-derived stem cells on the apoptosis of Schwann cells in peripheral nerve injury. CNS Neurosci Ther Feb. 2020;26(2):189–96. https://doi.org/10.1111/cns.13187.

    Article  CAS  Google Scholar 

  43. Ching RC, Wiberg M, Kingham PJ. Schwann cell-like differentiated adipose stem cells promote neurite outgrowth via secreted exosomes and RNA transfer. Stem Cell Res Ther Oct. 2018;11(1):266. https://doi.org/10.1186/s13287-018-1017-8.

    Article  CAS  Google Scholar 

  44. Brown BN, Ratner BD, Goodman SB, Amar S, Badylak SF. Macrophage polarization: an opportunity for improved outcomes in biomaterials and regenerative medicine. Biomaterials May. 2012;33(15):3792–802. https://doi.org/10.1016/j.biomaterials.2012.02.034.

    Article  CAS  Google Scholar 

  45. Gordon S, Taylor PR. Monocyte and macrophage heterogeneity. Nat Rev Immunol Dec. 2005;5(12):953–64. https://doi.org/10.1038/nri1733.

    Article  CAS  Google Scholar 

  46. Lu L, Dong J, Liu Y, et al. New insights into natural products that target the gut microbiota: effects on the prevention and treatment of colorectal cancer. Front Pharmacol. 2022;13:964793. https://doi.org/10.3389/fphar.2022.964793.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Hikawa N, Takenaka T. Myelin-stimulated macrophages release neurotrophic factors for adult dorsal root ganglion neurons in culture. Cell Mol Neurobiol Aug. 1996;16(4):517–28. https://doi.org/10.1007/bf02150231.

    Article  CAS  Google Scholar 

  48. Zigmond RE, Echevarria FD. Macrophage biology in the peripheral nervous system after injury. Prog Neurobiol Feb. 2019;173:102–21. https://doi.org/10.1016/j.pneurobio.2018.12.001.

    Article  CAS  Google Scholar 

  49. Tomlinson JE, Žygelytė E, Grenier JK, Edwards MG, Cheetham J. Temporal changes in macrophage phenotype after peripheral nerve injury. J Neuroinflammation Jun. 2018;15(1):185. https://doi.org/10.1186/s12974-018-1219-0.

    Article  CAS  Google Scholar 

  50. Martini R, Fischer S, López-Vales R, David S. Interactions between Schwann cells and macrophages in injury and inherited demyelinating disease. Glia Nov. 2008;1(14):1566–77. https://doi.org/10.1002/glia.20766.

    Article  Google Scholar 

  51. Karanth S, Yang G, Yeh J, Richardson PM. Nature of signals that initiate the immune response during Wallerian degeneration of peripheral nerves. Exp Neurol Nov. 2006;202(1):161–6. https://doi.org/10.1016/j.expneurol.2006.05.024.

    Article  CAS  Google Scholar 

  52. George A, Schmidt C, Weishaupt A, Toyka KV, Sommer C. Serial determination of tumor necrosis factor-alpha content in rat sciatic nerve after chronic constriction injury. Exp Neurol Nov. 1999;160(1):124–32. https://doi.org/10.1006/exnr.1999.7193.

    Article  CAS  Google Scholar 

  53. Shamash S, Reichert F, Rotshenker S. The cytokine network of Wallerian degeneration: tumor necrosis factor-alpha, interleukin-1alpha, and interleukin-1beta. J Neurosci Apr. 2002;15(8):3052–60. https://doi.org/10.1523/jneurosci.22-08-03052.2002.

    Article  Google Scholar 

  54. Gonçalves AF, Dias NG, Moransard M, et al. Gelsolin is required for macrophage recruitment during remyelination of the peripheral nervous system. Glia Apr. 2010;15(6):706–15. https://doi.org/10.1002/glia.20956.

    Article  Google Scholar 

  55. Allard DE, Wang Y, Li JJ, et al. Schwann cell-derived periostin promotes autoimmune peripheral polyneuropathy via macrophage recruitment. J Clin Invest Oct. 2018;1(10):4727–41. https://doi.org/10.1172/jci99308.

    Article  Google Scholar 

  56. Yin G, Lin Y, Wang P, Zhou J, Lin H. Upregulated lncARAT in Schwann cells promotes axonal regeneration by recruiting and activating proregenerative macrophages. Mol Med Jun. 2022;29(1):76. https://doi.org/10.1186/s10020-022-00501-9.

    Article  CAS  Google Scholar 

  57. Stratton JA, Holmes A, Rosin NL, et al. Macrophages regulate Schwann Cell Maturation after nerve Injury. Cell Rep Sep. 2018;4(10):2561–e25726. https://doi.org/10.1016/j.celrep.2018.08.004.

    Article  CAS  Google Scholar 

  58. Zhan C, Ma CB, Yuan HM, Cao BY, Zhu JJ. Macrophage-derived microvesicles promote proliferation and migration of Schwann cell on peripheral nerve repair. Biochem Biophys Res Commun Dec. 2015;4–11(1–2):343–8. https://doi.org/10.1016/j.bbrc.2015.10.097.

    Article  CAS  Google Scholar 

  59. Yang XX, Sun C, Wang L, Guo XL. New insight into isolation, identification techniques and medical applications of exosomes. J Control Release Aug. 2019;28:308:119–29. https://doi.org/10.1016/j.jconrel.2019.07.021.

    Article  CAS  Google Scholar 

  60. Chen C, Zhang Z, Gu X, Sheng X, Xiao L, Wang X, Exosomes. New regulators of reproductive development. Mater Today Bio Apr. 2023;19:100608. https://doi.org/10.1016/j.mtbio.2023.100608.

    Article  CAS  Google Scholar 

  61. Lässer C, Eldh M, Lötvall J. Isolation and characterization of RNA-containing exosomes. J Vis Exp Jan. 2012;9(59):e3037. https://doi.org/10.3791/3037.

    Article  CAS  Google Scholar 

  62. Zhao R, Zhao T, He Z, Cai R, Pang W. Composition, isolation, identification and function of adipose tissue-derived exosomes. Adipocyte Dec. 2021;10(1):587–604. https://doi.org/10.1080/21623945.2021.1983242.

    Article  CAS  Google Scholar 

  63. Shao H, Im H, Castro CM, Breakefield X, Weissleder R, Lee H. New Technologies for Analysis of Extracellular Vesicles. Chem Rev. 2018;118(4):1917–50. https://doi.org/10.1021/acs.chemrev.7b00534. /02/28 2018.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Li X, Corbett AL, Taatizadeh E, et al. Challenges and opportunities in exosome research-perspectives from biology, engineering, and cancer therapy. APL Bioeng Mar. 2019;3(1):011503. https://doi.org/10.1063/1.5087122.

    Article  CAS  Google Scholar 

  65. Lai JJ, Chau ZL, Chen SY, et al. Exosome Processing and characterization approaches for Research and Technology Development. Adv Sci (Weinh). May 2022;9(15):e2103222. https://doi.org/10.1002/advs.202103222.

  66. Hassanpour M, Rezaie J, Nouri M, Panahi Y. The role of extracellular vesicles in COVID-19 virus infection. Infect Genet Evol Nov. 2020;85:104422. https://doi.org/10.1016/j.meegid.2020.104422.

    Article  CAS  Google Scholar 

  67. Yang C, Robbins PD. The roles of tumor-derived exosomes in cancer pathogenesis. Clin Dev Immunol. 2011;2011:842849. https://doi.org/10.1155/2011/842849.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Marleau AM, Chen CS, Joyce JA, Tullis RH. Exosome removal as a therapeutic adjuvant in cancer. J Transl Med Jun. 2012;27:10:134. https://doi.org/10.1186/1479-5876-10-134.

    Article  CAS  Google Scholar 

  69. Fu M, Gu J, Jiang P, Qian H, Xu W, Zhang X. Exosomes in gastric cancer: roles, mechanisms, and applications. Mol Cancer Mar. 2019;15(1):41. https://doi.org/10.1186/s12943-019-1001-7.

    Article  Google Scholar 

  70. Tovar-Camargo OA, Toden S, Goel A. Exosomal microRNA biomarkers: emerging frontiers in colorectal and other human cancers. Expert Rev Mol Diagn. 2016;16(5):553–67. https://doi.org/10.1586/14737159.2016.1156535.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Baassiri A, Nassar F, Mukherji D, Shamseddine A, Nasr R, Temraz S. Exosomal non coding RNA in LIQUID biopsies as a Promising Biomarker for Colorectal Cancer. Int J Mol Sci Feb. 2020;19(4). https://doi.org/10.3390/ijms21041398.

  72. Yue Y, Yang X, Zhang L, et al. Low-intensity pulsed ultrasound upregulates pro-myelination indicators of Schwann cells enhanced by co-culture with adipose-derived stem cells. Cell Prolif Dec. 2016;49(6):720–8. https://doi.org/10.1111/cpr.12298.

    Article  CAS  Google Scholar 

  73. Wang Y, Yu T, Hu F. Hypocapnia Stimuli-Responsive Engineered exosomes delivering miR-218 facilitate sciatic nerve regeneration. Front Bioeng Biotechnol. 2022;10:825146. https://doi.org/10.3389/fbioe.2022.825146.

    Article  PubMed  PubMed Central  Google Scholar 

  74. Chen J, Ren S, Duscher D, et al. Exosomes from human adipose-derived stem cells promote sciatic nerve regeneration via optimizing Schwann cell function. J Cell Physiol Dec. 2019;234(12):23097–110. https://doi.org/10.1002/jcp.28873.

    Article  CAS  Google Scholar 

  75. Kuo PJ, Rau CS, Wu SC, et al. Exosomes secreted by adipose-derived stem cells following FK506 stimulation reduce autophagy of macrophages in spine after nerve crush Injury. Int J Mol Sci Sep. 2021;6(17). https://doi.org/10.3390/ijms22179628.

  76. Rau CS, Kuo PJ, Wu SC, et al. Enhanced nerve regeneration by Exosomes secreted by adipose-derived stem cells with or without FK506 stimulation. Int J Mol Sci Aug. 2021;9(16). https://doi.org/10.3390/ijms22168545.

  77. Yin G, Liu C, Lin Y, Xie Z, Hou C, Lin H. [Effect of exosomes from adipose-derived stem cells on peripheral nerve regeneration]. Zhongguo Xiu Fu Chong Jian Wai Ke Za Zhi Dec. 2018;15(12):1592–6. https://doi.org/10.7507/1002-1892.201707051.

    Article  Google Scholar 

  78. Yin G, Yu B, Liu C, et al. Exosomes produced by adipose-derived stem cells inhibit schwann cells autophagy and promote the regeneration of the myelin sheath. Int J Biochem Cell Biol Mar. 2021;132:105921. https://doi.org/10.1016/j.biocel.2021.105921.

    Article  CAS  Google Scholar 

  79. Qing L, Chen H, Tang J, Jia X. Exosomes and their MicroRNA Cargo: New players in Peripheral nerve regeneration. Neurorehabil Neural Repair Sep. 2018;32(9):765–76. https://doi.org/10.1177/1545968318798955.

    Article  Google Scholar 

  80. Akyurekli C, Le Y, Richardson RB, Fergusson D, Tay J, Allan DS. A systematic review of preclinical studies on the therapeutic potential of mesenchymal stromal cell-derived microvesicles. Stem Cell Rev Rep Feb. 2015;11(1):150–60. https://doi.org/10.1007/s12015-014-9545-9.

    Article  CAS  Google Scholar 

  81. Wen D, Peng Y, Liu D, Weizmann Y, Mahato RI. Mesenchymal stem cell and derived exosome as small RNA carrier and immunomodulator to improve islet transplantation. J Control Release Sep. 2016;28:238:166–75. https://doi.org/10.1016/j.jconrel.2016.07.044.

    Article  CAS  Google Scholar 

  82. Pashoutan Sarvar D, Shamsasenjan K, Akbarzadehlaleh P. Mesenchymal stem cell-derived exosomes: New Opportunity in Cell-Free Therapy. Adv Pharm Bull Sep. 2016;6(3):293–9. https://doi.org/10.15171/apb.2016.041.

    Article  CAS  Google Scholar 

  83. Schepici G, Silvestro S, Mazzon E. Regenerative effects of exosomes-derived MSCs: an overview on spinal cord Injury Experimental studies. Biomedicines Jan. 2023;13(1). https://doi.org/10.3390/biomedicines11010201.

  84. Namini MS, Ebrahimi-Barough S, Ai J, et al. Tissue-Engineered Core-Shell Silk-Fibroin/Poly-l-Lactic acid nerve Guidance Conduit containing encapsulated exosomes of Human endometrial stem cells promotes peripheral nerve regeneration. ACS Biomater Sci Eng Jun. 2023;12(6):3496–511. https://doi.org/10.1021/acsbiomaterials.3c00157.

    Article  CAS  Google Scholar 

  85. Li C, Li X, Shi Z, et al. Exosomes from LPS-preconditioned bone marrow MSCs accelerated peripheral nerve regeneration via M2 macrophage polarization: involvement of TSG-6/NF-κB/NLRP3 signaling pathway. Exp Neurol Oct. 2022;356:114139. https://doi.org/10.1016/j.expneurol.2022.114139.

    Article  CAS  Google Scholar 

  86. Zhang YU, Ye G, Zhao J, et al. Exosomes carried miR-181c-5p alleviates neuropathic pain in CCI rat models. Acad Bras Cienc. 2022;94(3):e20210564. https://doi.org/10.1590/0001-3765202220210564.

    Article  CAS  Google Scholar 

  87. Zhang Y, Wang WT, Gong CR, Li C, Shi M. Combination of olfactory ensheathing cells and human umbilical cord mesenchymal stem cell-derived exosomes promotes sciatic nerve regeneration. Neural Regen Res Oct. 2020;15(10):1903–11. https://doi.org/10.4103/1673-5374.280330.

    Article  CAS  Google Scholar 

  88. Zhao J, Ding Y, He R, et al. Dose-effect relationship and molecular mechanism by which BMSC-derived exosomes promote peripheral nerve regeneration after crush injury. Stem Cell Res Ther Aug. 2020;18(1):360. https://doi.org/10.1186/s13287-020-01872-8.

    Article  CAS  Google Scholar 

  89. Bucan V, Vaslaitis D, Peck CT, Strauß S, Vogt PM, Radtke C. Effect of Exosomes from Rat adipose-derived mesenchymal stem cells on Neurite Outgrowth and sciatic nerve regeneration after crush Injury. Mol Neurobiol Mar. 2019;56(3):1812–24. https://doi.org/10.1007/s12035-018-1172-z.

    Article  CAS  Google Scholar 

  90. Zeng X, Bian W, Liu Z, et al. Muscle-derived stem cell exosomes with overexpressed miR-214 promote the regeneration and repair of rat sciatic nerve after crush injury to activate the JAK2/STAT3 pathway by targeting PTEN. Front Mol Neurosci. 2023;16:1146329. https://doi.org/10.3389/fnmol.2023.1146329.

    Article  CAS  PubMed  Google Scholar 

  91. Jessen KR, Mirsky R. The origin and development of glial cells in peripheral nerves. Nat Rev Neurosci Sep. 2005;6(9):671–82. https://doi.org/10.1038/nrn1746.

    Article  CAS  Google Scholar 

  92. Kidd GJ, Ohno N, Trapp BD. Biology of Schwann cells. Handb Clin Neurol. 2013;115:55–79. https://doi.org/10.1016/b978-0-444-52902-2.00005-9.

    Article  PubMed  Google Scholar 

  93. Kalani A, Tyagi A, Tyagi N. Exosomes: mediators of neurodegeneration, neuroprotection and therapeutics. Mol Neurobiol Feb. 2014;49(1):590–600. https://doi.org/10.1007/s12035-013-8544-1.

    Article  CAS  Google Scholar 

  94. Lai CP, Breakefield XO. Role of exosomes/microvesicles in the nervous system and use in emerging therapies. Front Physiol. 2012;3:228. https://doi.org/10.3389/fphys.2012.00228.

    Article  CAS  PubMed  Google Scholar 

  95. Lopez-Verrilli MA, Picou F, Court FA. Schwann cell-derived exosomes enhance axonal regeneration in the peripheral nervous system. Glia Nov. 2013;61(11):1795–806. https://doi.org/10.1002/glia.22558.

    Article  Google Scholar 

  96. Sun J, Liao Z, Li Z, et al. Down-regulation miR-146a-5p in Schwann cell-derived exosomes induced macrophage M1 polarization by impairing the inhibition on TRAF6/NF-κB pathway after peripheral nerve injury. Exp Neurol Apr. 2023;362:114295. https://doi.org/10.1016/j.expneurol.2022.114295.

    Article  CAS  Google Scholar 

  97. Chen W, Chang S, Yang C, et al. Schwann cell–like cells derived from human amniotic mesenchymal stem cells promote sciatic nerve repair through an exosome–induced SOX2/FN1 pathway in vitro. Int J Mol Med Jun. 2022;49(6). https://doi.org/10.3892/ijmm.2022.5136.

  98. Hu T, Chang S, Qi F, et al. Neural grafts containing exosomes derived from Schwann cell-like cells promote peripheral nerve regeneration in rats. Burns Trauma. 2023;11:tkad013. https://doi.org/10.1093/burnst/tkad013.

    Article  PubMed  Google Scholar 

  99. Hori N, Narita M, Yamashita A, et al. Changes in the expression of IL-6-Mediated MicroRNAs in the dorsal root ganglion under neuropathic pain in mice. Synapse Aug. 2016;70(8):317–24. https://doi.org/10.1002/syn.21902.

    Article  CAS  Google Scholar 

  100. Zhang Y, Yi D, Hong Q, et al. Platelet-rich plasma-derived exosomes boost mesenchymal stem cells to promote peripheral nerve regeneration. J Control Release Mar. 2024;367:265–82. https://doi.org/10.1016/j.jconrel.2024.01.043.

    Article  CAS  Google Scholar 

  101. Zhang Y, Yi D, Hong Q, et al. Platelet-rich plasma-derived exosomes enhance mesenchymal stem cell paracrine function and nerve regeneration potential. Biochem Biophys Res Commun Mar. 2024;5:699:149496. https://doi.org/10.1016/j.bbrc.2024.149496.

    Article  CAS  Google Scholar 

  102. Yi D, Zhang Y, Li M, et al. UTMD assisted delivery of PRP-exos promoting peripheral nerve regeneration. Tissue Eng Part Aug. 2023;23. https://doi.org/10.1089/ten.TEA.2023.0133.

  103. Lu CF, Wang B, Zhang PX, et al. Combining chitin biological conduits with small autogenous nerves and platelet-rich plasma for the repair of sciatic nerve defects in rats. CNS Neurosci Ther Jul. 2021;27(7):805–19. https://doi.org/10.1111/cns.13640.

    Article  CAS  Google Scholar 

  104. Peixun Z, Na H, Kou Y, Xiaofeng Y, Jiang B. Peripheral nerve intersectional repair by bi-directional induction and systematic remodelling: biodegradable conduit tubulization from basic research to clinical application. Artif Cells Nanomed Biotechnol Dec. 2017;45(8):1464–6. https://doi.org/10.1080/21691401.2017.1373658.

    Article  Google Scholar 

  105. Li C, Liu SY, Zhang M, et al. Sustained release of exosomes loaded into polydopamine-modified chitin conduits promotes peripheral nerve regeneration in rats. Neural Regen Res Sep. 2022;17(9):2050–7. https://doi.org/10.4103/1673-5374.335167.

    Article  CAS  Google Scholar 

  106. Rao F, Zhang D, Fang T, et al. Exosomes from human gingiva-derived mesenchymal stem cells combined with biodegradable chitin conduits promote rat sciatic nerve regeneration. Stem Cells Int. 2019;2019:2546367. https://doi.org/10.1155/2019/2546367.

    Article  CAS  PubMed  Google Scholar 

  107. Yang Z, Yang Y, Xu Y, et al. Biomimetic nerve guidance conduit containing engineered exosomes of adipose-derived stem cells promotes peripheral nerve regeneration. Stem Cell Res Ther Aug. 2021;6(1):442. https://doi.org/10.1186/s13287-021-02528-x.

    Article  CAS  Google Scholar 

  108. Liu WZ, Ma ZJ, Li JR, Kang XW. Mesenchymal stem cell-derived exosomes: therapeutic opportunities and challenges for spinal cord injury. Stem Cell Res Ther Feb. 2021;3(1):102. https://doi.org/10.1186/s13287-021-02153-8.

    Article  Google Scholar 

  109. Feng J, Zhang Y, Zhu Z, Gu C, Waqas A, Chen L. Emerging exosomes and Exosomal MiRNAs in spinal cord Injury. Front Cell Dev Biol. 2021;9:703989. https://doi.org/10.3389/fcell.2021.703989.

    Article  PubMed  Google Scholar 

  110. Zeng J, Gu C, Sun Y, Chen X. Engineering of M2 macrophages-derived exosomes via click Chemistry for spinal cord Injury Repair. Adv Healthc Mater Apr. 2023;12(11):e2203391. https://doi.org/10.1002/adhm.202203391.

    Article  CAS  Google Scholar 

  111. Gao ZS, Zhang CJ, Xia N, et al. Berberine-loaded M2 macrophage-derived exosomes for spinal cord injury therapy. Acta Biomater May. 2021;126:211–23. https://doi.org/10.1016/j.actbio.2021.03.018.

    Article  CAS  Google Scholar 

  112. Fan Y, Li Y, Huang S, Xu H, Li H, Liu B. Resveratrol-primed exosomes strongly promote the recovery of motor function in SCI rats by activating autophagy and inhibiting apoptosis via the PI3K signaling pathway. Neurosci Lett Sep. 2020;25:736:135262. https://doi.org/10.1016/j.neulet.2020.135262.

    Article  CAS  Google Scholar 

  113. Liu Z, Tong H, Li J, et al. Low-stiffness hydrogels promote peripheral nerve regeneration through the Rapid Release of Exosomes. Front Bioeng Biotechnol. 2022;10:922570. https://doi.org/10.3389/fbioe.2022.922570.

    Article  PubMed  PubMed Central  Google Scholar 

  114. Yang Q, Su S, Liu S, et al. Exosomes-loaded electroconductive nerve dressing for nerve regeneration and pain relief against diabetic peripheral nerve injury. Bioact Mater Aug. 2023;26:194–215. https://doi.org/10.1016/j.bioactmat.2023.02.024.

    Article  CAS  Google Scholar 

  115. Zhang J, Li S, Li L, et al. Exosome and exosomal microRNA: trafficking, sorting, and function. Genomics Proteom Bioinf Feb. 2015;13(1):17–24. https://doi.org/10.1016/j.gpb.2015.02.001.

    Article  CAS  Google Scholar 

  116. Li P, Kaslan M, Lee SH, Yao J, Gao Z. Progress in Exosome isolation techniques. Theranostics. 2017;7(3):789–804. https://doi.org/10.7150/thno.18133.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  117. Samanta S, Rajasingh S, Drosos N, Zhou Z, Dawn B, Rajasingh J. Exosomes: new molecular targets of diseases. Acta Pharmacol Sin Apr. 2018;39(4):501–13. https://doi.org/10.1038/aps.2017.162.

    Article  CAS  Google Scholar 

  118. Yamashita T, Takahashi Y, Takakura Y. Possibility of Exosome-based therapeutics and challenges in production of Exosomes Eligible for therapeutic application. Biol Pharm Bull. 2018;41(6):835–42. https://doi.org/10.1248/bpb.b18-00133.

    Article  CAS  PubMed  Google Scholar 

  119. Charoenviriyakul C, Takahashi Y, Morishita M, Matsumoto A, Nishikawa M, Takakura Y. Cell type-specific and common characteristics of exosomes derived from mouse cell lines: yield, physicochemical properties, and pharmacokinetics. Eur J Pharm Sci Jan. 2017;1:96:316–22. https://doi.org/10.1016/j.ejps.2016.10.009.

    Article  CAS  Google Scholar 

  120. Kurian TK, Banik S, Gopal D, Chakrabarti S, Mazumder N. Elucidating methods for isolation and quantification of exosomes: a review. Mol Biotechnol Apr. 2021;63(4):249–66. https://doi.org/10.1007/s12033-021-00300-3.

    Article  CAS  Google Scholar 

  121. Barile L, Vassalli G, Exosomes. Therapy delivery tools and biomarkers of diseases. Pharmacol Ther Jun. 2017;174:63–78. https://doi.org/10.1016/j.pharmthera.2017.02.020.

    Article  CAS  Google Scholar 

  122. Gutierrez-Millan C, Calvo Díaz C, Lanao JM, Colino CI. Advances in Exosomes-based drug Delivery systems. Macromol Biosci Jan. 2021;21(1):e2000269. https://doi.org/10.1002/mabi.202000269.

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank Le Qi, Zhi-Jia Ma and Ran Meng for identifying relevant literature in the manuscript and for editing the manuscript. The authors declare that artificial intelligence is not used in this study.

Funding

Not applicable.

Author information

Authors and Affiliations

Authors

Contributions

GDX and GJZ wrote portions of the initial draft of this paper and modified the initial draft. XYL contributed to the figures editing. XDF and GJZ designed the work, edited the initial draft of this paper and made final changes to the final version before submission. All authors have read and agreed to the published version of the manuscript.

Corresponding author

Correspondence to Guan-Jie Zhao.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Xin, GD., Liu, XY., Fan, XD. et al. Exosomes repairment for sciatic nerve injury: a cell-free therapy. Stem Cell Res Ther 15, 214 (2024). https://doi.org/10.1186/s13287-024-03837-7

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s13287-024-03837-7

Keywords