Next Article in Journal
Gut Microbiota Dysbiosis in Japanese Female Patients with Nontuberculous Mycobacteria-Associated Lung Disease: An Observational Study
Previous Article in Journal
Interleukin-37 Suppresses the Function of Type 2 Follicular Helper T in Allergic Rhinitis
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Extracellular Vesicles as Emerging Therapeutic Strategies in Spinal Cord Injury: Ready to Go

1
Aoyang Institute of Cancer, Affiliated Aoyang Hospital of Jiangsu University, 279 Jingang Road, Suzhou 215600, China
2
Zhenjiang Key Laboratory of High Technology Research on sEVs Foundation and Transformation Application, School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang 212013, China
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Biomedicines 2025, 13(5), 1262; https://doi.org/10.3390/biomedicines13051262 (registering DOI)
Submission received: 8 April 2025 / Revised: 17 May 2025 / Accepted: 20 May 2025 / Published: 21 May 2025
(This article belongs to the Section Molecular and Translational Medicine)

Abstract

:
Spinal cord injury (SCI) is a prevalent central nervous system disorder that causes significant disability and mortality. Unfortunately, due to the complex pathophysiological mechanisms involved, there remains a critical paucity of effective therapeutic interventions capable of achieving neural tissue regeneration and functional recovery enhancement in SCI patients. The advancements in extracellular vesicles (EVs) as a cell-free therapy for SCI have displayed notable benefits. These include their small size, low immunogenicity, capacity to target specific areas, and ability to cross the blood‒brain barrier (BBB). EVs offer the potential to not only repair tissue damage and stimulate regeneration but also effectively deliver and release them at the site of SCI when combined with diverse biomaterials. This review explores the biological role and importance of EVs in treating SCI, highlighting the combined use of modified EVs with different biomaterials and their potential for future applications. It presents new and hopeful treatment approaches for individuals afflicted with SCI.

Graphical Abstract

1. Introduction

Spinal cord injury (SCI) is a severe injury that results in permanent damage to motor dysfunction and sensory perception and causes various complications, such as neuropathic pain and cardiac arrhythmias [1]. Globally, SCI affects more than 20 million people, with different countries reporting an annual incidence of approximately 23.77 cases per million people [2,3]. This condition significantly reduces the quality of life for patients, leading to significant financial and psychological burdens. In clinical practice, the primary therapeutic measures following SCI include prompt surgical spinal decompression, medication, and rehabilitation [4,5]. While recent clinical research has improved the therapeutic outcomes for SCI patients, there are still limitations in fully restoring spinal cord function. Therefore, it is crucial to develop novel therapeutic approaches that promote spinal cord recovery.
Extracellular vesicles (EVs) are a promising and highly recognized new method for repairing tissue damage following cell therapy. EVs are lipid bilayer vesicles secreted by almost all cell types, carrying various biomolecules such as nucleic acids, proteins, and lipids [6]. EVs play a role as “intercellular messengers” in various biological processes such as immune responses, tissue repair, tumorigenesis, and metastasis by transporting these biomolecules [7,8]. The International Society for Extracellular Vesicles (ISEV) recommends dividing EVs into large EVs (>200 nm) and small EVs (<200 nm) based on their size [9]. Large EVs are derived from cell membrane budding, including microvesicles and apoptotic bodies [10]. Small EVs are membrane-bound vesicles released into the extracellular environment after the fusion of multivesicular bodies (MVBs), carrying functional molecules of the parent cells [11,12]. Of note, there is still a lack of recognized separation methods. Large EVs and small EVs cannot be strictly separated and often have cross contamination [9]. EVs released by different types of cells have a vesicle-like shape and encapsulate multiple biomolecules. However, the characteristics of EVs, such as their size, content composition, and surface markers, vary depending on the cell types they originate from [13,14]. This diversity reflects the influence of different cell sources. Therefore, cells from different sources influence the bioactive components and function of EVs [15,16,17].
In the past, the transplantation of stem cells such as mesenchymal stem cells and neural stem cells has been widely used in the treatment of SCI [18,19]. Stem cells can promote the repair of nerve function by secreting neurotrophic factors and replace damaged cells through their multidirectional differentiation ability. However, stem cell transplantation still has some defects such as low survival rate in vivo, immune rejection and tumorigenic risk, which hinder its clinical transformation [20,21]. In recent years, EVs have emerged as crucial players in the initiation, progression, and recovery of central nervous system injury. Studies have shown that EVs are not only easy to store and transport, but also overcome the immunogenicity and tumorigenicity of cell therapy [7]. Specifically, EVs possess the ability to enhance the regenerative capacity of neurons and improve the external microenvironment in the injured area following SCI [22,23,24]. Additionally, in order to achieve sufficient targeting and prolong the half-time of EVs, engineered EVs strategy combined with targeted peptide modification and biomaterials has become a hot topic in SCI [25,26,27]. In view of the complex pathophysiological mechanism of SCI, the multi-targeted combination therapy strategy can avoid the limitations of single therapy and significantly enhance the homing ability, enrichment efficiency, and sustained release of EVs at the injured site. Here, we provide a brief overview of the mechanisms underlying SCI, and summarize the possible therapeutic benefits of EVs derived from various cell types and engineered EVs in treating SCI. Additionally, we will explore the emerging strategies involving the combination of EVs with various biomaterials as a potential treatment approach for SCI.

2. Pathophysiology of SCI

Based on the underlying pathophysiology, SCI can be categorized into primary injury and secondary injury (Figure 1). Primary injury, which is the disruption of the spinal cord’s structural integrity caused by physical factors such as compression, contusion, or stretching, causes irreversible mechanical damage, leading to hemorrhage, disruption of the blood–spinal cord barrier (BSCB) and nerve fiber bundles, and spinal shock [28,29,30]. This initial injury is followed by a series of secondary events, including ischemia, hypoxia, inflammatory response, oxidative stress, neuronal death, demyelination, and scar formation, further exacerbating the extent of injury [30,31]. During the acute phase and subacute phase, disruption of the microvasculature and BSCB results in changes to the local microenvironment at the site of the injury, including infiltration of immune cells (e.g., neutrophils, macrophages, and monocytes) and release of inflammatory factors, vasoactive peptides, and damage-associated molecular patterns (DAMP) [32,33]. These factors further intensify the inflammatory cascade response, for example, chemokine CCL2 promotes the migration of peripheral immune cells, activation of microglia, and neuronal apoptosis [34]. Moreover, ischemia induces prolonged edema and vascular thrombosis, leading to the accumulation of excessive reactive oxygen species (ROS), reactive nitrogen species, and other free radicals at the site of the injury, which creates a hypoxic microenvironment and ultimately triggers neuronal apoptosis [35,36]. Subsequently, when the chronic stage of injury is entered, reactive astrocytes aggregate around the cystic cavitation created by necrosis of the spinal cord’s gray matter to form a glial scar. This barrier-like structure may restrict regenerating axons from reaching the injury site, impair neural axon regeneration and tissue repair, and ultimately result in permanent functional deficits [30,37]. Improving the microenvironment of the injury site, regulating oxidative stress and neuroinflammation, and activating endogenous neuronal and axonal regeneration processes are crucial for treating SCI.
Traditionally, surgical procedures, medication administration (such as neurotrophic drugs, dehydrating agents, and hormones), and the incorporation of traditional Chinese medicine approaches are the main clinical treatments for SCI. Unfortunately, there is currently no successful method for repairing damaged nervous tissue and enhancing functional restoration. Recently, researchers have shown a significant interest in EVs as a novel approach for treating SCI. The autocrine, paracrine, and endocrine mechanisms of EVs play a significant role in cellular communication, enabling EVs to effectively cross the BBB and enter the bloodstream through endocytosis. Several studies have shown that EVs, as important mediators of intercellular communication, contribute to promoting axon regeneration, restoring nerve function, and facilitating tissue repair post injury [38,39,40].

3. Industrial Production of EVs

EVs, as a cell-free therapy, have received widespread attention in the fields of tissue damage repair and regenerative medicine [41,42,43]. With the advancement of technology and in-depth research, problems such as low yield and inherent heterogeneity of EVs have emerged. The industrial production of EVs is still limited by cultivation methods and separation technologies. The traditional cultivation of EVs involves the production of cells in cell culture flasks, followed by the collection of culture supernatants and the extraction of EVs. This method has limited cultivation space and requires frequent passage with a large amount of labor, which hinders its clinical application [44]. Although 2D cell culture has a simple operation process, the yield of EVs is low and the labor and economic costs are high [45]. Recently, three-dimensional (3D) cell culture systems such as hollow fiber bioreactors, stirred tank bioreactors, suspended bioreactors, and 3D microbeads have achieved mechanical stimulation on cells to promote the biogenesis of EVs [44,45,46]. Under the same conditions, this method can achieve large-scale production of EVs and reduce the manufacturing costs. Three-dimensional cell culture simulates the in vivo microenvironment under physiological conditions and promotes cells–matrix interactions, thereby increasing the yield and functional activity of EVs [44,47]. Studies have shown that compared with 2D culture, the 3D culture of hucMSCs using vertical wheel bioreactors resulted in a 9.4-fold increase in the number of EVs collected per batch and a greater amount of EVs released per cell [48]. In addition, 3D cell culture enriched EVs with more bioactive proteins and miRNAs related to metabolism, immune regulation, and neural growth [49,50]. However, the high shear rates, materials, and structures of bioreactors can affect the yield and functional activity of EVs. Moreover, the production, function, and targeting of EVs could be significantly improved through gene editing technology or drug stimulation [51,52]. However, it also faces challenges such as technical complexity and safety. The large-scale cultivation of gene-edited cells requires a high cost. The drug-stimulated batch processing mode cannot achieve continuous production, limiting clinical applications.
EVs derived from different cell sources have different characteristics of size, content, and surface markers. The traditional methods for EV separation include ultracentrifugation, size exclusion chromatography, ultrafiltration, and immunoprecipitation [53,54]. Of note, ultracentrifugation is regarded as the “gold standard” for EV separation and is widely used for EV separation and purification in various samples [55,56]. However, this technology has limitations such as complex separation procedures, and poor purity and uniformity of EV separation [53,57]. It is crucial to overcome the limitations of EV isolation technology in order to further explore the molecular basis of targeted delivery of EVs in disease treatment. Recently, microfluidic technology based on multiple microchannel single-cell analysis precisely separated EV subpopulations. Compared with traditional methods, microfluidic technology can reduce sample and reagent losses, and improve separation efficiency and accuracy [58,59]. Asymmetric flow field-flow fractionation (AF4) separates EV subpopulations based on differences in fluid flow properties and fluid dynamics, featuring high reproducibility, specificity, rapidity, convenience, and no labeling [60,61]. Liangsupree et al. isolated sEV subpopulations with diameters of less than 50 nm, 50–80 nm, and 80–120 nm using AF4 technology [62]. AF4 separation technology is beneficial for in-depth research on the biogenesis, molecular mechanisms, and metabolic regulation characteristics of EV subpopulations [7,61]. Therefore, effective large-scale production and separation technologies contribute to the production of EVs at clinical-grade quality. Future research needs to formulate standardized production strategies for EVs, which will help achieve breakthrough progress in the treatment of diseases such as SCI.

4. The Emerging Roles of EVs from Various Cell Sources in SCI

Cell therapy has been shown to be a promising therapeutic target for treating refractory diseases. Controversially, its clinical application is limited by immune reactions and high costs associated with storing and transporting reagents [21,63]. To address these challenges, researchers have turned to cell-free therapies, specifically exploring EVs from various cell sources as a safer alternative. EVs from various cell sources, such as neural stem cells (NSCs) and mesenchymal stem cells (MSCs), such as human umbilical cord MSCs (hucMSCs) and adipose-derived stem cells (ADSCs), have demonstrated immunomodulation, anti-inflammation, tissue repair, and regeneration properties [16,64,65]. In the treatment of SCI, EVs offer distinct advantages over cell therapy [17,66,67]. In this discussion, we mainly examine the importance of EVs derived from different cell sources, such as NSCs and MSCs, as well as Schwann cells (SCs), astrocytes, and macrophages (Figure 2).

4.1. NSC-Derived EVs

NSC-derived extracellular vesicles (NSC-EVs) have multifaceted biological capacities, including regulating microglial polarization, inducing neural proliferation, and maintaining cellular homeostasis [16,68,69]. They offer a promising therapeutic avenue for nerve repair and regeneration. For instance, NSC-sEVs are enriched with Netrin1, which promotes neural differentiation functionality by upregulating the expression of transcription factors Hand2 and Phox2b through the Hand2/Phox2b axis [70]. miR-374-5p highly expressed in NSC-sEVs can promote autophagy and inhibit neuronal apoptosis through the regulation of the target gene SKT-4, alleviating spinal cord injury [71]. miR-124-3p abundance in NSC-sEVs can mediate the PI3K/AKT/NF-κB signaling pathway, which leads to the suppression of pro-inflammatory microglia and astrocyte activation, thus facilitating the recovery of motor function after SCI [72]. Altogether, NSC-sEVs exhibit anti-inflammatory properties, inhibit neuronal apoptosis, and establish a conducive microenvironment for neuronal differentiation and tissue regeneration.

4.2. MSC-Derived EVs

MSCs have aided significant advancement in tissue engineering and cell therapy due to their characteristics of multidirectional differentiation and paracrine signaling. At the onset of acute injury, factors such as IL-4, IL-13, and CCL2 secreted by MSCs regulate macrophage polarization, thereby reducing the extent of secondary injury and promoting functional recovery after SCI [73,74]. Compared with stem cells from other sources, hucMSCs have great potential in immunomodulation and regeneration [75,76,77]. HucMSC-derived sEVs (hucMSC-sEVs) have unique advantages, such as easy access, high efficiency of amplification and culture in vitro, and low immunogenicity [41,78]. miR-199a-3p/145-5p abundance in hucMSC-sEVs affects the ubiquitination of the TrkA by NGF/TrkA pathway, thereby inhibiting the detrimental effects of LPS on neuronal differentiation and promoting axon regeneration [64]. Interestingly, under the condition of microelectric fields, hucMSC-sEVs enriched with lncRNA-MALAT1 promoted neural tissue repair and motor function recovery through the miRNA-22-3p/SIRT1/AMPK axis [79].
sEVs derived from ADSC (ADSC-sEVs) contain neurotrophic, immunomodulatory, and angiogenic factors. These vesicles promote cell proliferation, alleviate scar tissue formation, prevent neuronal cell death, and decrease inflammation through paracrine signaling [80,81]. Moreover, under hypoxic conditions ADSCs secrete more sEVs (ADSC-Hyp-sEVs). LncGm37494 enriched in ADSC-Hyp-sEVs can target and inhibit miR-130b-3p and upregulate PPARγ expression, thereby promoting microglial polarization and alleviating the inflammatory microenvironment [65]. Collectively, the strengths of ADSC-sEVs in promoting cell proliferation and inhibiting neuronal apoptosis and inflammatory responses provide valuable insights for the treatment of SCI [82,83].

4.3. EVs Derived from Other Cells

SCI involves complex pathophysiological mechanisms, such as immune regulation involving macrophages and neurotoxicity mediated by A1-type astrocytes. Mounting studies suggest that EVs derived from different cell types, including SCs, astrocytes, and macrophages, are valuable in promoting SCI functional recovery.
SCs, as glial cells, facilitate axon regeneration and myelin formation and maintain the structural integrity of axons [84,85]. Schwann cell-derived sEVs (SC-sEVs) increase mitochondrial autophagy and reduce necrotic apoptosis through the AMPK signaling pathway [86]. SC-sEVs also promote TLR2 expression in astrocytes and downregulate the deposition of chondroitin sulfate proteoglycans (CSPGs) by activating the NF-κB/PI3K axis, promoting nerve functional recovery after SCI [85]. MFG-E8 is highly enriched in SC-sEVs, which can promote the polarization of M2 macrophages and microglia, and attenuate inflammatory responses by activating SOCS3 and inhibiting STAT3 phosphorylation [66].
In the later stage of SCI, pericytes and fibroblasts that accumulate at the site of injury are the “main force” of a glial scar. The presence of glial scarring restricts axonal growth and extension, which is ultimately detrimental to nerve functional recovery. Astrocytes exhibit significant advantages in neuroprotection [87]. For example, astrocyte-derived extracellular vesicles (AS-EVs) can increase the expression of MOB1 and the phosphorylation of YAP by activating the Hippo pathway, thus promoting neuronal elongation and recovery [88]. Additionally, AS-sEVs can reduce fibrosis in injured tissues and promote tissue repair and motor functional recovery [17,89].
Due to the destruction of the spinal cord vascular network, ischemia and cell death after SCI are challenging and lead to spinal cord dysfunction. Many studies have reported the role of growth factors in neurological diseases. M2 macrophages are conducive to new blood vessel formation and facilitating nerve development following SCI [90,91]. It is reported that M2 macrophage-derived EVs enriched with glial cell line-derived neurotrophic factor (GDNF) collected by transfection can significantly promote the outgrowth of axons and dendrites [92]. Additionally, M2-sEVs can promote the migration and proliferation of cerebral endothelial cells by upregulating proangiogenic factors HIF-1α and VEGF, thus facilitating vascular repair and nerve regeneration after SCI [67]. The ubiquitin thioesterase (OTULIN), which is highly enriched in M2-sEVs, mediates the activation of the Wnt/β-catenin axis and induces angiogenesis and tissue regeneration at the site of injury [90]. Interestingly, research has shown that neurotrophic factor basic fibroblast growth factor (bFGF) increases the release of neuronal EVs and the enrichment of various proteins by regulating mechanisms related to vesicle fusion, such as calcium and VAMP3 [93].
In addition to the EVs from different cell sources mentioned above, EVs secreted by Treg cells, M2 microglia, endothelial cells, etc., also play beneficial roles in inhibiting neuroinflammation, enhancing axonal regeneration, and promoting neurological recovery [94,95,96]. However, the clinical application of EVs still faces multiple limitations. For example, systemic administration such as the tail vein injection of EVs accelerates the capture and clearance of EVs by the mononuclear phagocytic cell system. A small amount of EVs can cross the blood–brain barrier and reach the site of spinal cord injury [97]. In addition, although EVs have lower immunogenicity than cell therapy, their surfaces may still carry major histocompatibility complex (MHC) molecules from the donor cells, causing host immune rejection. The safety of long-term multiple administration still needs to be verified [98]. Existing SCI research is mostly based on rodent models, whose damage mechanisms and repair abilities differ from those of humans, which may overestimate the therapeutic efficacy of EVs. The resolutions of these obstacles are crucial for establishing EVs as a dependable option for clinical SCI treatment.

5. Application of Engineered EVs in SCI Treatment

For a long time, we have been perplexed by the clinical challenges of SCI and the need to improve patient prognosis and quality of life. In view of their advantages of low cytotoxicity, low immunogenicity, and high biocompatibility, EVs have received extensive attention from researchers as important carriers for intercellular communication [99,100]. Recently, engineered EVs modified with peptides or loaded with contents have shown advantages in targeted delivery and in vivo recycling, providing an economical, convenient, and effective treatment for SCI.
Engineered EVs with surface-immobilized specific peptide segments can target and bind to specific cell receptors at the injury site, thereby enhancing EVs’ homing ability [27,101]. Exo-pep-11 was constructed by chemically coupling the EphA4 receptor targeting peptide with ADSC-sEVs, which could specifically bind to NSCs and stimulate its proliferation and differentiation [102]. For instance, it has been discovered that the intranasal administration of RGD (Arg-Gly-Asp) peptide-modified EVs could target endothelial cells overexpressing integrin αvβ3 at ischemic regions of SCI, significantly increasing the specific accumulation of EVs at the lesion site and contributing to the stability of the BSCB [27].
Exogenous drugs, biomolecules, and cytokines can be embedded into EVs through techniques such as electroporation or ultrasound [103]. The CAQK peptide can specifically bind to reactive astrocytes with a high expression of chondroitin sulfate proteoglycans (CSPG) in the lesion area. Based on the CAQK peptide chemical modification of induced neural stem cell (iNSC)-derived EVs, C-EVs-siRNA was constructed by electroporation loaded with CCL2-siRNA. C-EVs-siRNA promoted the synergistic effect of CCL2-siRNA and EVs to target and inhibit inflammation and neuronal apoptosis after SCI [104]. Moreover, EV membranes can protect drugs from enzymatic degradation and escape immune system clearance, thereby improving drug delivery efficiency. FTY720-NSC-sEVs, engineered NSC-sEVs loaded with FTY720, can effectively avoid the serious adverse reactions caused by the systemic administration of FTY720, reduce the expression of AQP-4 at the injury site, and upregulate claudin-5 to synergistically alleviate spinal cord edema [27]. Furthermore, FTY720-NSC-sEVs can mediate the downregulation of PTEN to activate the PI3K/AKT signaling pathway, thereby mitigating the effects of neuronal apoptosis and improving functional recovery after SCI. An engineered EV with neuroprotective and anti-inflammatory abilities, Cur@EVs-cl-NGF, was combined with a nerve growth factor (NGF) and curcumin (Cur) based on M2-sEVs. NGF can effectively enhance the ability of EVs to promote axon extension and inhibit neuronal apoptosis [105]. When Cur@EVs-cl-NGF is transported to the site of the injury, the localized high expression of MMP9 in SCI can efficiently dissociate NGF. Cur@EVs-cl-NGF can effectively solve the problems of low expression of endogenous NGF, short half-life, and low uptake of Cur. It has shown great advantages in relieving inflammation, neuroprotection, and biosafety.
Engineered EVs can effectively utilize drugs with optimal doses and avoid systemic toxicity. However, engineered EVs still have off-target risk. RGD peptides can bind to normal vascular endothelial cells, causing cross reactivity between targeted peptide modified EVs and non-specific tissues [27]. Moreover, EVs loaded with drugs can promote EVs inactivation in addition to reducing cycle stability. Engineered EVs need to improve targeting accuracy, preparation standardization, and preservation stability to facilitate clinical translation.

6. EVs Combined with Biomaterials for SCI

Emerging evidence indicates that a local injection of EVs for SCI can be rapidly cleared and metabolized by the human circulatory system. This prevents EVs from remaining at the injured site and releasing continuously, thereby significantly restricting their potential in facilitating the functional recovery of SCI [106,107]. Of note, the implantation of pre-formed biomaterials in situ offers a promising solution for SCI [108]. Injectable biomaterials as targeted delivery carriers can enhance the therapeutic effects of EVs by effectively retaining them at the injury site and facilitating sustained release [109]. Interestingly, significant headway has been made in exploring the application of biomaterials in conjunction with EVs for the treatment of SCI (Figure 3 and Table 1).
Given the potent value of this combination strategy in improving the therapeutic outcomes of EV-based treatments for SCI, various clinical trials evaluating the efficacy of biomaterials in the interventional therapy of SCI are emerging. Table 2 summarizes the current data on biomaterials used in interventional treatments for SCI from domestic and international clinical trials. Currently, most clinical trials involving the use of MSCs combined with biomaterials are still in their early stages. Moreover, clinical studies on EVs combined with biomaterials have not yet been initiated. However, we anticipate that as more research data emerge, the combination of EVs with different biomaterials will become widely adopted for SCI treatment and eventually transition to clinical practice.

6.1. EVs Combined with Fibrin Gels

Fibrin, as an FDA-approved biomaterial, has received wide attention. It has high biosafety, pro-wound healing, and degradability properties, which may make it a good scaffold for the repairment of SCI [111,118,119]. Interestingly, the degradation products of fibrin gels have also been shown to be biologically active and can promote cell proliferation, adhesion, and collagen synthesis [120].
Fibrin gels have been reported to enhance the differentiation and proliferation of NSCs and MSCs [108,121,122]. Fibrin gels can provide a matrix for the targeted delivery of sEVs and promote nerve growth, thereby significantly enhancing the role of sEVs in alleviating the microenvironment at the lesion site [110]. Additionally, nerve growth factor inducible (VGF) enriched in MSC-sEVs plays a key role in promoting remyelination and oligodendrogenesis. MSC-sEVs encapsulated in fibrin gel (Gel-sEVs) significantly promoted the expression levels of neural nucleus (NeuN), neural class III β-Tubulin (Tuj1), and doublecortin (Dcx), thereby effectively promoting neurogenesis and the recovery of motor function in SCI [111]. However, fibrin hydrogels, as a biocarrier for sEV loading, have not been explored for their effective retention of sEVs and slow release to the injured site. The degradation rate and mechanical properties of fibrin gel still need to be further improved [110,123].

6.2. EVs Combined with Collagen Scaffolds

The main element of the extracellular matrix is collagen, which is biodegradable. Collagen scaffolds possess three-dimensional porous characteristics and are superior to other biomaterials in promoting cell adhesion and growth properties [112,124,125]. The structure of collagen scaffolds benefits material exchange and provides a conducive environment for cell growth and differentiation.
A novel bio-specificity peptide (BSP) can be specifically bound to MSC-sEVs encapsulated with paclitaxel (PTX) and a linearly ordered collagen scaffold, thereby constructing a multifunctional collagen scaffold (LBMP). Compared with traditional binding methods, such as physical adsorption and chemical coupling, BSP, as a versatile linker molecule, is able to effectively preserve the integrity of the sEV membrane and its surface collagen [113]. After 7 days of LBMP implantation, the slow releasing rate of the drug reached 70%. After 60 days, LBMP gradually degraded with tissue regeneration at the lesion site. In the LBMP, MSC-sEVs promoted the migration of endogenous NSCs, while collagen scaffolds could effectively retain NSCs. LBMP has significant advantages in synergistically inducing neuronal differentiation, axonal growth, and inhibiting fiber scar deposition. Remarkably, collagen scaffolds still have problems such as poor mechanical properties [126]. Exogenous peptides in collagen scaffolds are difficult to remove and may cause biosafety concerns [112]. The current studies on the immunogenicity of collagen are also insufficiently in depth and need to be further explored to facilitate the advancement of collagen scaffolds in the clinical treatment of SCI [127].

6.3. EVs Combined with Bioactive Hydrogels

Hydrogels, as mimics of the natural extracellular matrix (ECM), possess a nano arrangement with mucoadhesive, elastic, and neuroinductive properties [107,128,129]. By injecting hydrogels with a sustained release of EVs, they can effectively fill the gap at the site of SCI and deliver EVs specifically to the injury site for therapeutic action [115,130].

6.3.1. EVs Combined with Hyaluronic Acid Hydrogels

Hyaluronic acid (HA) regulates a range of physiological processes including inflammation, angiogenesis, and wound healing. HA is present in almost all human tissues and body fluids and has high biocompatibility. However, HA is non-adhesive by itself and needs to be combined with biomaterials such as hydrogels and nanoparticles to expand its application [131,132]. In recent years, researchers have designed novel adhesive peptide-modified HA hydrogels, which optimize the SCI therapeutics involved in existing studies.
Based on the high adhesion of integrins on sEV membranes in combination with laminin, sEVs were implanted into HA hydrogel scaffolds (pGel-sEVs) modified with the laminin-derived peptide PPFLMLLKGSTR [117]. The adhesion rate of sEVs was much higher than that of the hydrogel in the peptide-modified pGel group. The sustained release of sEVs in pGel could be maintained for 11 days, with a cumulative release surpassing 90%. In contrast to equivalent intravenous sEVs via the tail vein, pGel-embedded sEVs demonstrated a diffusion pattern radiating from the lesion center to the periphery, displaying a strong correlation with the lesion site. Recently, a composite patch has been developed for the sequential release of SCs-sEVs and methylprednisolone (MP) at different pathological stages of SCI [35]. There is no significant difference in the properties of sEVs released by the composite patch compared with native sEVs. This composite patch can induce M2 macrophage polarization through the TLR4/NF-κB and MAPK signaling axis and modulate the Akt/mTOR signaling axis to inhibit neuronal apoptosis, leading to an improved microenvironment following SCI. Compared with traditional invasive treatment strategies such as local injection, scaffold implantation, and microneedle array patch, this transdermal drug delivery patch can reduce secondary losses and achieve a painless treatment of SCI [102,113,133]. Collectively, the hydrogel serves as a natural carrier for the sustained release and retention of sEVs, effectively filling irregular cavities at the site of the SCI and preventing secondary damage [116]. Notably, the binding and release of integrins and adhesion proteins in sEVs can be unstable. The release of sEVs following pGel-sEV implantation is influenced by the local microenvironment at the injury site, highlighting the significance of stable and continuous sEV release for effective SCI treatment [117].

6.3.2. EVs Combined with FE Hydrogels

In view of the disadvantages of hydrogels that affect neuronal development, such as poor mechanical properties and low resistance [134,135], it is imperative to enhance the performance of hydrogel. Wang et al. [115] prepared FE hydrogels consisting of F127 and PCE polymers, which are injectable, biocompatible, adhesive, and self-repairing. The MSCs-derived EVs (FE@EVs) loaded into the FE hydrogels by electrostatic interactions showed good, stable release compared with free MSCs-EVs, and approximately 90% of the EVs could be released from the FE hydrogels to the injury site for 56 days. In addition, FE@EVs significantly alleviate the injured microenvironment via activating MAP-2 and neurofilaments, inhibiting fiber scar formation and promoting axonal regeneration.

6.3.3. EVs Combined with Gelatin Methacrylate Hydrogels

Gelatin methacryloyl hydrogels (GelMA) consist of methacrylic anhydride and gelatin, which have good biocompatibility, thermal stability, and adaptability to various chemical modifications [136,137]. GelMA hydrogel was mixed with 3D-cultured MSCs-derived sEVs (3D-sEVs) to make microarray patches with nanostructures (GelMA-MN@3D-sEVs) [114]. According to quantitative proteomics analysis, 3D-sEVs are enriched with more proteins and miRNAs in the modulation of a local microenvironment and neural repair compared with 2D-sEVs. Additionally, 3D-sEVs showed an enrichment of the phospholipase D signaling pathway associated with axon growth and synapse development, as well as the Fc-epsilon-RI and mTOR signaling pathways associated with the regulation of neuroinflammation. Although specific mechanisms have not been elucidated, the combination of GelMA-MN and 3D-sEVs holds significant promise for the guidance of SCI clinical therapy.
The combination of EVs with different biomaterials as a new strategy in regenerative medicine has been widely used in studies on tissue regeneration and functional recovery. In addition, synthetic polymer scaffolds serve as another type of biomaterial that can repair or fill SCI-diseased tissues [138,139]. The synthetic polymers reported in existing research mainly include polyethylene glycol (PEG), polylactic acid (PLA), polyvinyl alcohol (PVA), and so on. Compared with natural polymers, synthetic polymer scaffolds have stable molecular structures, porosities, and mechanical properties [140,141,142]. Furthermore, synthetic polymers can be designed as biomaterials with corresponding biological properties according to different needs. Nevertheless, many biosafety issues with synthetic polymers limit their clinical applications. For example, the degradation products of synthetic polymers can cause local inflammatory reactions and aggravate tissue damage conditions [141,143].
Although the combined application of EVs and biomaterials has demonstrated significant advantages, its clinical application still faces many challenges. Loading EVs through ultrasound or chemical modification may disrupt the membrane integrity of EVs and induce immunogenicity [144,145]. Although the combined application of EVs with biomaterials such as hydrogels can prolong the release time, it is difficult to change accordingly in line with the different requirements of SCI acute and chronic phases [146,147]. In addition, the porosity and mechanical strength of the biomaterials need to match the physical characteristics of spinal cord tissue [146]. Otherwise, they may compress the lesion site or fail to effectively support axonal regeneration. In future studies, methods for overcoming the limitations of biomaterial applications are vital to the advancement of therapeutic strategies for SCI.

7. Conclusions and Outlook

SCI is a complicated process that prevents tissue function recovery after injury, greatly impacting the quality of life of patients. EVs, as good alternative materials capable of overcoming the inherent limitations of cell therapy, have become a focal point in SCI treatment research and have entered the clinical trial stage [78]. A series of studies have demonstrated the vast potential of EVs from various cell sources for tissue injury repair and neural regeneration [148,149]. As a potential cell-free therapy, EVs offer a safe and effective treatment strategy for SCI patients. Currently, the core factor of the slow clinical conversion process is the lack of international unified production and quality control standards for EVs, including separation and purification technology, large-scale production, and storage [150]. Notably, EVs-mediated cell-free therapies still have many obstacles to overcome. Current interdisciplinary technology integration is not yet mature. The non-specific accumulation of engineered EVs in the liver, spleen, and other organs may cause inflammatory reactions and damage normal tissues [151,152]. The limitations of EVs combined with biomaterials in the treatment of SCI need to be gradually overcome through in-depth research. A successful clinical translation will depend on the simultaneous optimization of precise delivery systems, in-depth mechanism analysis, and standardized production [113,120]. In the future, we must develop standardized separation and characterization techniques for EVs and establish production processes that comply with standardized operating procedures. We must also construct systematic toxicology evaluation standards and develop intelligent biomaterials through technological innovation [153]. We believe that with further research, these problems can be resolved.
In conclusion, previous studies have demonstrated the efficacy of EVs and their combined applications in promoting tissue regeneration and functional recovery following SCI. Nevertheless, future studies should aim to provide adequate evidence regarding the safety and reliability of these strategies in clinical settings, ultimately offering a promising treatment strategy for SCI with potential clinical applications.

Author Contributions

J.J. (Jiajia Jiang) and W.X. conceived and revised the paper. J.J. (Jiali Jiang) and S.C. drafted the manuscript and prepared the figures and tables. Z.W. and Q.B. undertook manuscript reviewing and editing. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by the National Natural Science Foundation of China (grant no. 82272179, 81971757) and the Jiangsu Province’s Major Project in Research and Development (BE2020680).

Data Availability Statement

Not applicable to this article, as no new data were created or analyzed in this study.

Acknowledgments

Figures were generated using BioRender.com.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

SCIspinal cord injury
EVsextracellular vesicles
MVBsmultivesicular bodies
BSCBblood–spinal cord barrier
DAMPdamage-associated molecular patterns
ROSreactive oxygen species
BBBblood‒brain barrier
NSCsneural stem cells
MSCsmesenchymal stem cells
hucMSCshuman umbilical cord MSCs
ADSCsadipose-derived stem cells
SCsSchwann cells
CNScentral nervous system
lncRNAlong non-coding RNA
Hyp-sEVssEVs under hypoxic conditions
AS-sEVsastrocyte-derived small extracellular vesicles
PKCprotein kinase C
M2-sEVsM2 macrophage-derived small extracellular vesicles
NGFnerve growth factor
MMP9metalloproteinase 9
FEF127-polycitrate-polyethyleneimine hydrogel
PTXpaclitaxel
ECMextracellular matrix
HAhyaluronic acid
3Dthree-dimensional
MPmethylprednisolone
PEGpolyethylene glycol
PLApolylactic acid
PVApolyvinyl alcohol

References

  1. Chen, M.; Lin, Y.; Guo, W.; Chen, L. BMSC-derived exosomes carrying miR-26a-5p ameliorate spinal cord injury via negatively regulating EZH2 and activating the BDNF-TrkB-CREB signaling. Mol. Neurobiol. 2024, 61, 8156–8174. [Google Scholar] [CrossRef] [PubMed]
  2. Lu, Y.; Shang, Z.; Zhang, W.; Pang, M.; Hu, X.; Dai, Y.; Shen, R.; Wu, Y.; Liu, C.; Luo, T.; et al. Global incidence and characteristics of spinal cord injury since 2000-2021: A systematic review and meta-analysis. BMC Med. 2024, 22, 285. [Google Scholar] [CrossRef] [PubMed]
  3. Fernandes, S.; Oatman, E.; Weinberger, J.; Dixon, A.; Osei-Owusu, P.; Hou, S. The susceptibility of cardiac arrhythmias after spinal cord crush injury in rats. Exp. Neurol. 2022, 357, 114200. [Google Scholar] [CrossRef]
  4. Manzari-Tavakoli, A.; Babajani, A.; Vousooghi, N.; Moghimi, A.; Tarasi, R.; Safaeinejad, F.; Norouzi, S.; Bahrami, S.; Niknejad, H. Therapeutic potential of placenta-derived stem cells cultivated on noggin-loaded nanochitosan/polypyrrole-alginate conductive scaffold to restore spinal cord injury. Stem Cell Res. Ther. 2024, 15, 497. [Google Scholar] [CrossRef]
  5. Khiar-Fernández, N.; Zian, D.; Vázquez-Villa, H.; Martínez, R.F.; Escobar-Peña, A.; Foronda-Sainz, R.; Ray, M.; Puigdomenech-Poch, M.; Cincilla, G.; Sánchez-Martínez, M.; et al. Novel antagonist of the type 2 lysophosphatidic acid receptor (LPA2), UCM-14216, ameliorates spinal cord injury in mice. J. Med. Chem. 2022, 65, 10956–10974. [Google Scholar] [CrossRef]
  6. Arasu, U.T.; Kärnä, R.; Härkönen, K.; Oikari, S.; Koistinen, A.; Kröger, H.; Qu, C.; Lammi, M.J.; Rilla, K. Human mesenchymal stem cells secrete hyaluronan-coated extracellular vesicles. Matrix Biol. 2017, 64, 54–68. [Google Scholar] [CrossRef]
  7. Liu, W.; Wang, X.; Chen, Y.; Yuan, J.; Zhang, H.; Jin, X.; Jiang, Y.; Cao, J.; Wang, Z.; Yang, S.; et al. Distinct molecular properties and functions of small EV subpopulations isolated from human umbilical cord MSCs using tangential flow filtration combined with size exclusion chromatography. J. Extracell. Vesicles 2025, 14, e70029. [Google Scholar] [CrossRef] [PubMed]
  8. Dong, X.; Jiang, D.; Wang, L.; Zhao, J.; Yu, L.; Huang, Y.; Wu, X.; Zhu, Y.; Zhao, Y.; Zhao, Q.; et al. VPS28 regulates brain vasculature by controlling neuronal VEGF trafficking through extracellular vesicle secretion. iScience 2022, 25, 104042. [Google Scholar] [CrossRef]
  9. Welsh, J.A.; Goberdhan, D.C.I.; O’Driscoll, L.; Buzas, E.I.; Blenkiron, C.; Bussolati, B.; Cai, H.; Di Vizio, D.; Driedonks, T.A.P.; Erdbrügger, U.; et al. Minimal information for studies of extracellular vesicles (MISEV2023): From basic to advanced approaches. J. Extracell. Vesicles 2024, 13, e12404. [Google Scholar] [CrossRef]
  10. Sheneman, K.R.; Cummins, T.D.; Merchant, M.L.; Hood, J.L.; Uriarte, S.M.; Lawrenz, M.B. Yersinia pestis actively inhibits the production of extracellular vesicles by human neutrophils. J. Extracell. Vesicles 2025, 14, e70074. [Google Scholar] [CrossRef]
  11. Yanagawa, K.; Yoshimori, T. Rubicon regulates exosome secretion via the non-autophagic pathway. Autophagy 2025, 21, 1160–1162. [Google Scholar] [CrossRef] [PubMed]
  12. Kim, J.; Lee, M.C.; Jeon, J.; Rodríguez-delaRosa, A.; Endo, Y.; Kim, D.S.; Madrigal-Salazar, A.D.; Seo, J.W.; Lee, H.; Kim, K.T.; et al. Combinational regenerative inductive effect of bio-adhesive hybrid hydrogels conjugated with hiPSC-derived myofibers and its derived EVs for volumetric muscle regeneration. Bioact. Mater. 2025, 43, 579–602. [Google Scholar] [CrossRef] [PubMed]
  13. Kalluri, R.; LeBleu, V.S. The biology, function, and biomedical applications of exosomes. Science 2020, 367, eaau6977. [Google Scholar] [CrossRef] [PubMed]
  14. Heo, J.S.; Choi, Y.; Kim, H.S.; Kim, H.O. Comparison of molecular profiles of human mesenchymal stem cells derived from bone marrow, umbilical cord blood, placenta and adipose tissue. Int. J. Mol. Med. 2016, 37, 115–125. [Google Scholar] [CrossRef]
  15. Karoichan, A.; Boucenna, S.; Tabrizian, M. Therapeutics of the future: Navigating the pitfalls of extracellular vesicles research from an osteoarthritis perspective. J. Extracell. Vesicles 2024, 13, e12435. [Google Scholar] [CrossRef]
  16. Rong, Y.; Liu, W.; Wang, J.; Fan, J.; Luo, Y.; Li, L.; Kong, F.; Chen, J.; Tang, P.; Cai, W. Neural stem cell-derived small extracellular vesicles attenuate apoptosis and neuroinflammation after traumatic spinal cord injury by activating autophagy. Cell Death. Dis. 2019, 10, 340. [Google Scholar] [CrossRef]
  17. Lu, Y.; Chen, C.; Wang, H.; Du, R.; Ji, J.; Xu, T.; Yang, C.; Chen, X. Astrocyte-derived sEVs alleviate fibrosis and promote functional recovery after spinal cord injury in rats. Int. Immunopharmacol. 2022, 113, 109322. [Google Scholar] [CrossRef]
  18. Zhong, D.; Cao, Y.; Li, C.; Li, M.; Rong, Z.; Jiang, L.; Guo, Z.; Lu, H.; Hu, J. Neural stem cell-derived exosomes facilitate spinal cord functional recovery after injury by promoting angiogenesis. Exp. Biol. Med 2020, 245, 54–65. [Google Scholar] [CrossRef]
  19. Huang, J.; Shi, L.; Yang, Y.; Zhao, F.; Chen, R.; Liao, W.; Zhu, J.; Yang, D.; Wu, X.; Han, S. Mesenchymal cell-derived exosomes and miR-29a-3p mitigate renal fibrosis and vascular rarefaction after renal ischemia reperfusion injury. Stem Cell Res. Ther. 2025, 16, 135. [Google Scholar] [CrossRef]
  20. Kao, Y.; Song, W.; Zhang, R.; Gu, G.; Qiu, H.; Shen, W.; Zhu, H.; Liu, Y.; Yang, Y.; Liu, H.; et al. Synergistic restoration of spinal cord injury through hyaluronic acid conjugated hydrogel-polydopamine nanoparticles combined with human mesenchymal stem cell transplantation. Bioact. Mater. 2025, 46, 569–581. [Google Scholar] [CrossRef]
  21. Wen, R.; Long, G.; He, X.; Zhang, K.; Ma, W.; Shen, Y.; Xiao, Z.; Zhao, Y.; Liu, D.; Dai, J.; et al. Revisiting the unobtrusive role of exogenous stem cells beyond neural circuits replacement in spinal cord injury repair. Theranostics 2025, 15, 1552–1569. [Google Scholar] [CrossRef] [PubMed]
  22. Hwang, J.; Jang, S.; Kim, C.; Lee, S.; Jeong, H.S. Role of stem cell-derived exosomes and microRNAs in spinal cord injury. Int. J. Mol. Sci. 2023, 24, 13849. [Google Scholar] [CrossRef] [PubMed]
  23. Hu, X.; Liu, Z.; Zhou, X.; Jin, Q.; Xu, W.; Zhai, X.; Fu, Q.; Qian, H. viaSmall extracellular vesicles derived from mesenchymal stem cell facilitate functional recovery in spinal cord injury by activating neural stem cells the ERK1/2 pathway. Front. Cell. Neurosci. 2022, 16, 954597. [Google Scholar] [CrossRef] [PubMed]
  24. Pan, D.; Zhu, S.; Zhang, W.; Wei, Z.; Yang, F.; Guo, Z.; Ning, G.; Feng, S. Autophagy induced by Schwann cell-derived exosomes promotes recovery after spinal cord injury in rats. Biotechnol. Lett. 2022, 44, 129–142. [Google Scholar] [CrossRef]
  25. Lin, Y.; Guo, Q.; Liu, Q.; Wang, W.; Lv, A.; Zhang, L.; Li, L.; Gao, J.; Huang, F. An injectable responsive exosome-releasing hydrogel based on sodium alginate restores motor and bladder function by alleviating the injury microenvironment and facilitating distal nerve repair. Int. J. Biol. Macromol. 2025, 304, 140819. [Google Scholar] [CrossRef]
  26. Li, Q.; Fu, X.; Kou, Y.; Han, N. Engineering strategies and optimized delivery of exosomes for theranostic application in nerve tissue. Theranostics 2023, 13, 4266–4286. [Google Scholar] [CrossRef]
  27. Xie, Y.; Sun, Y.; Liu, Y.; Zhao, J.; Liu, Q.; Xu, J.; Qin, Y.; He, R.; Yuan, F.; Wu, T.; et al. Targeted delivery of RGD-CD146+CD271+ human umbilical cord mesenchymal stem cell-derived exosomes promotes blood-spinal cord barrier repair after spinal cord injury. ACS Nano 2023, 17, 18008–18024. [Google Scholar] [CrossRef]
  28. Shen, K.; Li, X.; Huang, G.; Yuan, Z.; Xie, B.; Chen, T.; He, L. High rapamycin-loaded hollow mesoporous Prussian blue nanozyme targets lesion area of spinal cord injury to recover locomotor function. Biomaterials 2023, 303, 122358. [Google Scholar] [CrossRef]
  29. Yang, J.; Wang, M.; Zheng, S.; Huang, R.; Wen, G.; Zhou, P.; Wang, W.; Zhou, S.; Jiang, X.; Liu, S.; et al. Mesoporous polydopamine delivering 8-gingerol for the target and synergistic treatment to the spinal cord injury. J. Nanobiotechnol. 2023, 21, 192. [Google Scholar] [CrossRef]
  30. Liu, J.A.; Tam, K.W.; Chen, Y.L.; Feng, X.; Chan, C.W.L.; Lo, A.L.H.; Wu, K.L.; Hui, M.N.; Wu, M.H.; Chan, K.K.; et al. Transplanting human neural stem cells with ≈50% reduction of SOX9 gene dosage promotes tissue repair and functional recovery from severe spinal cord injury. Adv. Sci. 2023, 10, e2205804. [Google Scholar] [CrossRef]
  31. Guest, J.; Datta, N.; Jimsheleishvili, G.; Gater, D.R. Pathophysiology, classification and comorbidities after traumatic spinal cord injury. J. Pers Med. 2022, 12, 1126. [Google Scholar] [CrossRef] [PubMed]
  32. Jaffer, H.; Andrabi, S.S.; Petro, M.; Kuang, Y.; Steinmetz, M.P.; Labhasetwar, V. Catalytic antioxidant nanoparticles mitigate secondary injury progression and promote functional recovery in spinal cord injury model. J. Control. Release 2023, 364, 109–123. [Google Scholar] [CrossRef] [PubMed]
  33. Gu, C.; Geng, X.; Wu, Y.; Dai, Y.; Zeng, J.; Wang, Z.; Fang, H.; Sun, Y.; Chen, X. Engineered macrophage membrane-coated nanoparticles with enhanced CCR2 expression promote spinal cord injury repair by suppressing neuroinflammation and neuronal death. Small 2023, 20, e2305659. [Google Scholar] [CrossRef]
  34. Rong, Y.; Ji, C.; Wang, Z.; Ge, X.; Wang, J.; Ye, W.; Tang, P.; Jiang, D.; Fan, J.; Yin, G.; et al. Small extracellular vesicles encapsulating CCL2 from activated astrocytes induce microglial activation and neuronal apoptosis after traumatic spinal cord injury. J. Neuroinflamm. 2021, 18, 196. [Google Scholar] [CrossRef] [PubMed]
  35. Zhu, B.; Gu, G.; Ren, J.; Song, X.; Li, J.; Wang, C.; Zhang, W.; Huo, Y.; Wang, H.; Jin, L.; et al. Schwann cell-derived exosomes and methylprednisolone composite patch for spinal cord injury repair. ACS Nano 2023, 17, 22928–22943. [Google Scholar] [CrossRef]
  36. Wang, H.; Lin, F.; Wu, Y.; Guo, W.; Chen, X.; Xiao, C.; Chen, M. Carrier-free nanodrug based on co-assembly of methylprednisolone dimer and rutin for combined treatment of spinal cord injury. ACS Nano 2023, 17, 12176–12187. [Google Scholar] [CrossRef]
  37. Taylor, E.C.; Fitzpatrick, C.E.; Thompson, S.E.; Justice, S.B. Acute traumatic spinal cord injury. Adv. Emerg. Nurs. J. 2022, 44, 272–280. [Google Scholar] [CrossRef]
  38. Makrygianni, E.; Chrousos, G. Extracellular vesicles and the stress system. Neuroendocrinology 2022, 113, 120–167. [Google Scholar] [CrossRef]
  39. Ollen-Bittle, N.; Roseborough, A.; Wang, W.; Wu, J.; Whitehead, S. Mechanisms and biomarker potential of extracellular vesicles in stroke. Biology 2022, 11, 1231. [Google Scholar] [CrossRef]
  40. Fan, B.; Gu, J.; Wu, J.; Sun, Y.; Huang, R.; Shen, H.; Zhang, X.; Li, Z. Circulating abnormal extracellular vesicles: Their mechanism for crossing blood-brain barrier, effects on central nervous system and detection methods. J. Biomed. Nanotechnol. 2022, 18, 640–659. [Google Scholar] [CrossRef]
  41. Mirab, S.M.; Omidi, A.; Soleimani, M.; Soufi-Zomorrod, M.; Fekrirad, Z. Human umbilical cord mesenchymal stem cell-derived exosomes boost functional performance in an animal model of multiple sclerosis through recruiting oligodendrocytes and attenuating gliosis. Stem Cell Rev. Rep. 2025. [Google Scholar] [CrossRef] [PubMed]
  42. Fang, C.; Qian, J.; Tu, B.Z.; Xia, X.; Jia, C.Y.; Shen, C.L. MiR-124 delivered by extracellular vesicles from mesenchymal stem cell exerts neuroprotective effects by stabilizing the p62-Keap1-Nrf2 pathway after spinal cord injury in rats. Mol. Neurobiol. 2025. [Google Scholar] [CrossRef] [PubMed]
  43. Sun, M.K.; Passaro, A.P.; Latchoumane, C.F.; Spellicy, S.E.; Bowler, M.; Goeden, M.; Martin, W.J.; Holmes, P.V.; Stice, S.L.; Karumbaiah, L. Extracellular vesicles mediate neuroprotection and functional recovery after traumatic brain injury. J. Neurotrauma 2020, 37, 1358–1369. [Google Scholar] [CrossRef]
  44. Hu, Y.; Zheng, L.; Zheng, Z.; Fu, M.; Peng, H.; Ma, S. Microbead encapsulation strategy for efficient production of extracellular vesicles derived from human mesenchymal stem cells. J. Extracell. Vesicles 2025, 14, e70053. [Google Scholar] [CrossRef]
  45. Cao, J.; Wang, B.; Tang, T.; Lv, L.; Ding, Z.; Li, Z.; Hu, R.; Wei, Q.; Shen, A.; Fu, Y.; et al. Three-dimensional culture of MSCs produces exosomes with improved yield and enhanced therapeutic efficacy for cisplatin-induced acute kidney injury. Stem Cell Res. Ther. 2020, 11, 206. [Google Scholar] [CrossRef] [PubMed]
  46. Zhang, J.; Peng, Y.; Guo, M.; Li, C. Large-scale expansion of human umbilical cord-derived mesenchymal stem cells in a stirred suspension bioreactor enabled by computational fluid dynamics modeling. Bioengineering 2022, 9, 274. [Google Scholar] [CrossRef]
  47. Khan, N.L.A.; Muhandiram, S.; Dissanayake, K.; Godakumara, K.; Midekessa, G.; Andronowska, A.; Heath, P.R.; Kodithuwakku, S.; Hart, A.R.; Fazeli, A. Effect of 3D and 2D cell culture systems on trophoblast extracellular vesicle physico-chemical characteristics and potency. Front. Cell Dev. Biol. 2024, 12, 1382552. [Google Scholar] [CrossRef]
  48. Otahal, A.; Kramer, K.; Neubauer, M.; Gulová, S.; Lacza, Z.; Nehrer, S.; Luna, A. Culture of hoffa fat pad mesenchymal stem/stromal cells on microcarrier suspension in vertical wheel bioreactor for extracellular vesicle production. Stem Cell Res. Ther. 2024, 15, 61. [Google Scholar] [CrossRef]
  49. Ene, J.; Liu, C.; Syed, F.; Sun, L.; Berry, D.; Durairaj, P.; Liu, Z.L.; Zeng, C.; Jung, S.; Li, Y. Biomanufacturing and lipidomics analysis of extracellular vesicles secreted by human blood vessel organoids in a vertical wheel bioreactor. Stem Cell Res. Ther. 2025, 16, 207. [Google Scholar] [CrossRef] [PubMed]
  50. Son, J.P.; Kim, E.H.; Shin, E.K.; Kim, D.H.; Sung, J.H.; Oh, M.J.; Cha, J.M.; Chopp, M.; Bang, O.Y. Mesenchymal stem cell-extracellular vesicle therapy for stroke: Scalable production and imaging biomarker studies. Stem Cells Transl. Med. 2023, 12, 459–473. [Google Scholar] [CrossRef]
  51. Wang, J.; Bonacquisti, E.E.; Brown, A.D.; Nguyen, J. Boosting the biogenesis and secretion of mesenchymal stem cell-derived exosomes. Cells 2020, 9, 660. [Google Scholar] [CrossRef] [PubMed]
  52. Zhang, R.; Bu, T.; Cao, R.; Li, Z.; Wang, C.; Huang, B.; Wei, M.; Yuan, L.; Yang, G. An optimized exosome production strategy for enhanced yield while without sacrificing cargo loading efficiency. J. Nanobiotechnol. 2022, 20, 463. [Google Scholar] [CrossRef] [PubMed]
  53. Askeland, A.; Borup, A.; Østergaard, O.; Olsen, J.V.; Lund, S.M.; Christiansen, G.; Kristensen, S.R.; Heegaard, N.H.H.; Pedersen, S. Mass-spectrometry based proteome comparison of extracellular vesicle isolation methods: Comparison of ME-kit, size-exclusion chromatography, and high-speed centrifugation. Biomedicines 2020, 8, 246. [Google Scholar] [CrossRef]
  54. Andriolo, G.; Provasi, E.; Brambilla, A.; Panella, S.; Soncin, S.; Cicero, V.L.; Radrizzani, M.; Turchetto, L.; Barile, L. Methodologies for scalable production of high-quality purified small extracellular vesicles from conditioned medium. Methods Mol. Biol. 2023, 2668, 69–98. [Google Scholar] [CrossRef]
  55. Lozano, N.; Prescilla-Ledezma, A.; Calabuig, E.; Trelis, M.; Arce, J.M.S.; López Hontangas, J.L.; de Pablos, L.M.; Gomez-Samblas, M.; Osuna, A. Circulating extracellular vesicles in sera of chronic patients as a method for determining active parasitism in Chagas disease. PLoS Negl. Trop. Dis. 2024, 18, e0012356. [Google Scholar] [CrossRef]
  56. Filipović, L.; Spasojević Savković, M.; Prodanović, R.; Matijašević Joković, S.; Stevanović, S.; Marco, A.; Kosanović, M.; Brajušković, G.; Popović, M. Urinary extracellular vesicles as a readily available biomarker source: A simplified stratification method. Int. J. Mol. Sci. 2024, 25, 8004. [Google Scholar] [CrossRef] [PubMed]
  57. Jackson, K.K.; Powell, R.R.; Marcus, R.K.; Bruce, T.F. Comparison of the capillary-channeled polymer (C-CP) fiber spin-down tip approach to traditional methods for the isolation of extracellular vesicles from human urine. Anal. Bioanal. Chem. 2022, 414, 3813–3825. [Google Scholar] [CrossRef]
  58. Bu, Y.; Wang, J.; Ni, S.; Lu, Z.; Guo, Y.; Yobas, L. High-performance gel-free and label-free size fractionation of extracellular vesicles with two-dimensional electrophoresis in a microfluidic artificial sieve. Anal. Chem. 2024, 96, 3508–3516. [Google Scholar] [CrossRef]
  59. Lu, Y.; Lin, B.; Liu, W.; Zhang, J.; Zhu, L.; Yang, C.; Song, Y. Isolation of PD-L1 extracellular vesicle subpopulations using DNA computation mediated microfluidic tandem separation. Small Methods 2023, 7, e2300516. [Google Scholar] [CrossRef]
  60. Multia, E.; Liangsupree, T.; Jussila, M.; Ruiz-Jimenez, J.; Kemell, M.; Riekkola, M.L. Automated on-line isolation and fractionation system for nanosized biomacromolecules from human plasma. Anal. Chem. 2020, 92, 13058–13065. [Google Scholar] [CrossRef]
  61. Zhang, H.; Freitas, D.; Kim, H.S.; Fabijanic, K.; Li, Z.; Chen, H.; Mark, M.T.; Molina, H.; Martin, A.B.; Bojmar, L.; et al. Identification of distinct nanoparticles and subsets of extracellular vesicles by asymmetric flow field-flow fractionation. Nat. Cell Biol. 2018, 20, 332–343. [Google Scholar] [CrossRef] [PubMed]
  62. Liangsupree, T.; Multia, E.; Forssén, P.; Fornstedt, T.; Riekkola, M.L. Kinetics and interaction studies of anti-tetraspanin antibodies and ICAM-1 with extracellular vesicle subpopulations using continuous flow quartz crystal microbalance biosensor. Biosens. Bioelectron. 2022, 206, 114151. [Google Scholar] [CrossRef] [PubMed]
  63. Gómez-Ferrer, M.; Villanueva-Badenas, E.; Sánchez-Sánchez, R.; Sánchez-López, C.M.; Baquero, M.C.; Sepúlveda, P.; Dorronsoro, A. HIF-1α and pro-inflammatory signaling improves the immunomodulatory activity of MSC-derived extracellular vesicles. Int. J. Mol. Sci. 2021, 22, 3416. [Google Scholar] [CrossRef]
  64. Wang, Y.; Lai, X.; Wu, D.; Liu, B.; Wang, N.; Rong, L. Umbilical mesenchymal stem cell-derived exosomes facilitate spinal cord functional recovery through the miR-199a-3p/145-5p-mediated NGF/TrkA signaling pathway in rats. Stem Cell Res. Ther. 2021, 12, 117. [Google Scholar] [CrossRef]
  65. Shao, M.; Jin, M.; Xu, S.; Zheng, C.; Zhu, W.; Ma, X.; Lv, F. Exosomes from long noncoding RNA-Gm37494-ADSCs repair spinal cord injury via shifting microglial M1/M2 polarization. Inflammation 2020, 43, 1536–1547. [Google Scholar] [CrossRef] [PubMed]
  66. Ren, J.; Zhu, B.; Gu, G.; Zhang, W.; Li, J.; Wang, H.; Wang, M.; Song, X.; Wei, Z.; Feng, S. Schwann cell-derived exosomes containing MFG-E8 modify macrophage/microglial polarization for attenuating inflammation via the SOCS3/STAT3 pathway after spinal cord injury. Cell Death Dis. 2023, 14, 70. [Google Scholar] [CrossRef]
  67. Huang, J.H.; He, H.; Chen, Y.N.; Liu, Z.; Romani, M.D.; Xu, Z.Y.; Xu, Y.; Lin, F.Y. Exosomes derived from M2 macrophages improve angiogenesis and functional recovery after spinal cord injury through HIF-1α/VEGF axis. Brain Sci. 2022, 12, 1322. [Google Scholar] [CrossRef]
  68. Wu, Z.; Han, T.; Dong, Y.; Ying, W.; Fang, H.; Liu, Y.; Song, P.; Shen, C. Acid-sensing ion channel-1 contributes to the failure of myelin sheath regeneration following spinal cord injury by transcellular delivery of PGE2. Cell. Mol. Biol. Lett. 2024, 29, 149. [Google Scholar] [CrossRef]
  69. Rong, Y.; Liu, W.; Lv, C.; Wang, J.; Luo, Y.; Jiang, D.; Li, L.; Zhou, Z.; Zhou, W.; Li, Q.; et al. Neural stem cell small extracellular vesicle-based delivery of 14-3-3t reduces apoptosis and neuroinflammation following traumatic spinal cord injury by enhancing autophagy by targeting Beclin-1. Aging 2019, 11, 7723–7745. [Google Scholar] [CrossRef]
  70. Ma, L.; Wei, X.; Ma, W.; Liu, Y.; Wang, Y.; He, Y.; Jia, S.; Wang, Y.; Luo, W.; Liu, D.; et al. Neural stem cell-derived exosomal Netrin1 contributes to neuron differentiation of mesenchymal stem cells in therapy of spinal bifida aperta. Stem Cells Transl. Med. 2022, 11, 539–551. [Google Scholar] [CrossRef]
  71. Zhang, L.; Han, P. Neural stem cell-derived exosomes suppress neuronal cell apoptosis by activating autophagy via miR-374-5p/STK-4 axis in spinal cord injury. J. Musculoskelet. Neuronal Interact. 2022, 22, 411–421. [Google Scholar] [PubMed]
  72. Jiang, D.; Gong, F.; Ge, X.; Lv, C.; Huang, C.; Feng, S.; Zhou, Z.; Rong, Y.; Wang, J.; Ji, C.; et al. Neuron-derived exosomes-transmitted miR-124-3p protect traumatically injured spinal cord by suppressing the activation of neurotoxic microglia and astrocytes. J. Nanobiotechnol. 2020, 18, 105. [Google Scholar] [CrossRef] [PubMed]
  73. Papa, S.; Vismara, I.; Mariani, A.; Barilani, M.; Rimondo, S.; De Paola, M.; Panini, N.; Erba, E.; Mauri, E.; Rossi, F.; et al. Mesenchymal stem cells encapsulated into biomimetic hydrogel scaffold gradually release CCL2 chemokine in situ preserving cytoarchitecture and promoting functional recovery in spinal cord injury. J. Control. Release 2018, 278, 49–56. [Google Scholar] [CrossRef] [PubMed]
  74. Nakajima, H.; Uchida, K.; Guerrero, A.R.; Watanabe, S.; Sugita, D.; Takeura, N.; Yoshida, A.; Long, G.; Wright, K.T.; Johnson, W.E.; et al. Transplantation of mesenchymal stem cells promotes an alternative pathway of macrophage activation and functional recovery after spinal cord injury. J. Neurotrauma 2012, 29, 1614–1625. [Google Scholar] [CrossRef]
  75. Huang, L.; Fu, C.; Xiong, F.; He, C.; Wei, Q. Stem cell therapy for spinal cord injury. Cell Transplant. 2021, 30, 963689721989266. [Google Scholar] [CrossRef]
  76. Yang, Y.; Pang, M.; Du, C.; Liu, Z.Y.; Chen, Z.H.; Wang, N.X.; Zhang, L.M.; Chen, Y.Y.; Mo, J.; Dong, J.W.; et al. Repeated subarachnoid administrations of allogeneic human umbilical cord mesenchymal stem cells for spinal cord injury: A phase 1/2 pilot study. Cytotherapy 2021, 23, 57–64. [Google Scholar] [CrossRef]
  77. Zheng, Y.; Wang, Y.; Liu, W.; Li, Y.; Ma, X.; Abulimiti, M.; Maimaitiaili, N.; Qin, H. Intrathecal transplantation of human umbilical cord mesenchymal stem cells enhances spinal cord injury recovery: Role of miR-124-3p as a biomarker. Exp. Ther. Med. 2025, 29, 57. [Google Scholar] [CrossRef]
  78. Akhlaghpasand, M.; Tavanaei, R.; Hosseinpoor, M.; Yazdani, K.O.; Soleimani, A.; Zoshk, M.Y.; Soleimani, M.; Chamanara, M.; Ghorbani, M.; Deylami, M.; et al. Safety and potential effects of intrathecal injection of allogeneic human umbilical cord mesenchymal stem cell-derived exosomes in complete subacute spinal cord injury: A first-in-human, single-arm, open-label, phase I clinical trial. Stem Cell Res. Ther. 2024, 15, 264. [Google Scholar] [CrossRef]
  79. Li, K.; Liu, Z.; Wu, P.; Chen, S.; Wang, M.; Liu, W.; Zhang, L.; Guo, S.; Liu, Y.; Liu, P.; et al. Micro electrical fields induced MSC-sEVs attenuate neuronal cell apoptosis by activating autophagy via lncRNA MALAT1/miR-22-3p/SIRT1/AMPK axis in spinal cord injury. J. Nanobiotechnol. 2023, 21, 451. [Google Scholar] [CrossRef]
  80. Fu, Y.; Zhang, Y.L.; Liu, R.Q.; Xu, M.M.; Xie, J.L.; Zhang, X.L.; Xie, G.M.; Han, Y.T.; Zhang, X.M.; Zhang, W.T.; et al. Exosome lncRNA IFNG-AS1 derived from mesenchymal stem cells of human adipose ameliorates neurogenesis and ASD-like behavior in BTBR mice. J. Nanobiotechnol. 2024, 22, 66. [Google Scholar] [CrossRef]
  81. Rau, C.S.; Wu, S.C.; Kuo, P.J.; Lin, C.W.; Lu, T.H.; Wu, Y.C.; Tsai, C.W.; Hsieh, C.H. Tracking adipose-derived stem cell exosomes applied in a mouse crush injury model: Insights from fluorescent labeling and spatial transcriptomics—An experimental study. Int. J. Surg. 2024, 111, 1860–1873. [Google Scholar] [CrossRef]
  82. Lu, X.; Lv, C.; Zhao, Y.; Wang, Y.; Li, Y.; Ji, C.; Wang, Z.; Ye, W.; Yu, S.; Bai, J.; et al. TSG-6 released from adipose stem cells-derived small extracellular vesicle protects against spinal cord ischemia reperfusion injury by inhibiting endoplasmic reticulum stress. Stem Cell Res. Ther. 2022, 13, 291. [Google Scholar] [CrossRef] [PubMed]
  83. Sung, S.; Seo, M.; Kim, Y.; Kang, K.; Choi, J.; Lee, S.; Sung, M.; Yim, S.; Lim, J.; Seok, H.; et al. Human epidural AD-MSC exosomes improve function recovery after spinal cord injury in rats. Biomedicines 2022, 10, 678. [Google Scholar] [CrossRef] [PubMed]
  84. Wei, Z.; Fan, B.; Ding, H.; Liu, Y.; Tang, H.; Pan, D.; Shi, J.; Zheng, P.; Shi, H.; Wu, H.; et al. Proteomics analysis of Schwann cell-derived exosomes: A novel therapeutic strategy for central nervous system injury. Mol. Cell. Biochem. 2019, 457, 51–59. [Google Scholar] [CrossRef]
  85. Pan, D.; Li, Y.; Yang, F.; Lv, Z.; Zhu, S.; Shao, Y.; Huang, Y.; Ning, G.; Feng, S. Increasing toll-like receptor 2 on astrocytes induced by Schwann cell-derived exosomes promotes recovery by inhibiting CSPGs deposition after spinal cord injury. J. Neuroinflamm. 2021, 18, 172. [Google Scholar] [CrossRef]
  86. Xu, B.; Zhou, Z.; Fang, J.; Wang, J.; Tao, K.; Liu, J.; Liu, S. Exosomes derived from schwann cells alleviate mitochondrial dysfunction and necroptosis after spinal cord injury via AMPK signaling pathway-mediated mitophagy. Free Radic. Biol. Med. 2023, 208, 319–333. [Google Scholar] [CrossRef]
  87. Sun, R.; Liao, W.; Lang, T.; Qin, K.; Jiao, K.; Shao, L.; Deng, C.; She, Y. Astrocyte-derived exosomal miR-378a-5p mitigates cerebral ischemic neuroinflammation by modulating NLRP3-mediated pyroptosis. Front. Immunol. 2024, 15, 1454116. [Google Scholar] [CrossRef]
  88. Sun, H.; Cao, X.; Gong, A.; Huang, Y.; Xu, Y.; Zhang, J.; Sun, J.; Lv, B.; Li, Z.; Guan, S.; et al. Extracellular vesicles derived from astrocytes facilitated neurite elongation by activating the Hippo pathway. Exp. Cell Res. 2022, 411, 112937. [Google Scholar] [CrossRef] [PubMed]
  89. You, C.; Zhou, W.; Ye, P.; Zhang, L.; Sun, W.; Tian, L.; Peng, B.; Hu, M.; Xu, B. LncRNA 4933431K23Rik modulate microglial phenotype via inhibiting miR-10a-5p in spinal cord injury induced neuropathic pain. Sci. Rep. 2025, 15, 11620. [Google Scholar] [CrossRef]
  90. Luo, Z.; Peng, W.; Xu, Y.; Xie, Y.; Liu, Y.; Lu, H.; Cao, Y.; Hu, J. Exosomal OTULIN from M2 macrophages promotes the recovery of spinal cord injuries via stimulating Wnt/β-catenin pathway-mediated vascular regeneration. Acta Biomater. 2021, 136, 519–532. [Google Scholar] [CrossRef]
  91. 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. 2023, 12, e2203391. [Google Scholar] [CrossRef] [PubMed]
  92. Zhao, Y.; Haney, M.J.; Fallon, J.K.; Rodriguez, M.; Swain, C.J.; Arzt, C.J.; Smith, P.C.; Loop, M.S.; Harrison, E.B.; El-Hage, N.; et al. Using extracellular vesicles released by GDNF-transfected macrophages for therapy of parkinson disease. Cells 2022, 11, 1933. [Google Scholar] [CrossRef] [PubMed]
  93. Kumar, R.; Tang, Q.; Müller, S.A.; Gao, P.; Mahlstedt, D.; Zampagni, S.; Tan, Y.; Klingl, A.; Bötzel, K.; Lichtenthaler, S.F.; et al. Fibroblast growth factor 2-mediated regulation of neuronal exosome release depends on VAMP3/cellubrevin in hippocampal neurons. Adv. Sci. 2020, 7, 1902372. [Google Scholar] [CrossRef]
  94. Guan, P.; Fan, L.; Zhu, Z.; Yang, Q.; Kang, X.; Li, J.; Zhang, Z.; Liu, S.; Liu, C.; Wang, X.; et al. M2 microglia-derived exosome-loaded electroconductive hydrogel for enhancing neurological recovery after spinal cord injury. J. Nanobiotechnol. 2024, 22, 8. [Google Scholar] [CrossRef]
  95. Kong, G.; Xiong, W.; Li, C.; Xiao, C.; Wang, S.; Li, W.; Chen, X.; Wang, J.; Chen, S.; Zhang, Y.; et al. Treg cells-derived exosomes promote blood-spinal cord barrier repair and motor function recovery after spinal cord injury by delivering miR-2861. J. Nanobiotechnol. 2023, 21, 364. [Google Scholar] [CrossRef] [PubMed]
  96. Huang, J.; Zhang, G.; Li, S.; Li, J.; Wang, W.; Xue, J.; Wang, Y.; Fang, M.; Zhou, N. Endothelial cell-derived exosomes boost and maintain repair-related phenotypes of Schwann cells via miR199-5p to promote nerve regeneration. J. Nanobiotechnol. 2023, 21, 10. [Google Scholar] [CrossRef]
  97. Sun, Y.; Zhao, J.; Liu, Q.; Xu, Y.; Qin, Y.; He, R.; Zheng, L.; Xie, Y.; Li, C.; Wu, T.; et al. Intranasal delivery of small extracellular vesicles from specific subpopulation of mesenchymal stem cells mitigates traumatic spinal cord injury. J. Control Release 2024, 369, 335–350. [Google Scholar] [CrossRef]
  98. Deng, M.; Xie, P.; Xue, H.; Chen, Q.; Zhou, Y.; Ming, J.; Ma, Y.; Liu, J.; Huang, H. Decellularized tissue matrices hydrogels functionalized with extracellular vesicles promote macrophage reprogramming and neural stem cell differentiation for spinal cord injury repair. J. Nanobiotechnol. 2025, 23, 139. [Google Scholar] [CrossRef]
  99. Chen, G.; Li, S.; Tong, K.; Huang, Z.; Liu, S.; Zhu, H.; Zhong, Y.; Zhou, Z.; Jiao, G.; Wei, F.; et al. Extracellular vesicles released by transforming growth factor-beta 1-preconditional mesenchymal stem cells promote recovery in mice with spinal cord injury. Bioact. Mater. 2024, 35, 135–149. [Google Scholar] [CrossRef]
  100. Xu, Y.; Wang, X.; Zhou, X.; Zeng, W.; Yuan, J.; Ye, J. Multiple strategies enhance the efficacy of MSC-Exos transplantation for spinal cord injury. Exp. Neurol. 2024, 383, 115038. [Google Scholar] [CrossRef]
  101. Wang, D.; Lyu, Y.; Yang, Y.; Zhang, S.; Chen, G.; Pan, J.; Tian, W. Schwann cell-derived EVs facilitate dental pulp regeneration through endogenous stem cell recruitment via SDF-1/CXCR4 axis. Acta. Biomater. 2022, 140, 610–624. [Google Scholar] [CrossRef]
  102. Ghosh, S.; Roy, R.; Mukherjee, N.; Ghosh, S.; Jash, M.; Jana, A.; Ghosh, S. EphA4 targeting peptide-conjugated extracellular vesicles rejuvenates adult neural stem cells and exerts therapeutic benefits in aging rats. ACS Chem. Neurosci. 2024, 15, 3482–3495. [Google Scholar] [CrossRef] [PubMed]
  103. Wan, Y.; Li, L.; Chen, R.; Han, J.; Lei, Q.; Chen, Z.; Tang, X.; Wu, W.; Liu, S.; Yao, X. Engineered extracellular vesicles efficiently deliver CRISPR-Cas9 ribonucleoprotein (RNP) to inhibit herpes simplex virus1 infection in vitro and in vivo. Acta Pharm. Sin. B 2024, 14, 1362–1379. [Google Scholar] [CrossRef] [PubMed]
  104. Rong, Y.; Wang, Z.; Tang, P.; Wang, J.; Ji, C.; Chang, J.; Zhu, Y.; Ye, W.; Bai, J.; Liu, W.; et al. Engineered extracellular vesicles for delivery of siRNA promoting targeted repair of traumatic spinal cord injury. Bioact. Mater. 2023, 23, 328–342. [Google Scholar] [CrossRef] [PubMed]
  105. Zhang, C.; Li, D.; Hu, H.; Wang, Z.; An, J.; Gao, Z.; Zhang, K.; Mei, X.; Wu, C.; Tian, H. Engineered extracellular vesicles derived from primary M2 macrophages with anti-inflammatory and neuroprotective properties for the treatment of spinal cord injury. J. Nanobiotechnol. 2021, 19, 373. [Google Scholar] [CrossRef] [PubMed]
  106. Liu, Z.; Guo, S.; Dong, L.; Wu, P.; Li, K.; Li, X.; Li, X.; Qian, H.; Fu, Q. A tannic acid doped hydrogel with small extracellular vesicles derived from mesenchymal stem cells promotes spinal cord repair by regulating reactive oxygen species microenvironment. Mater. Today Bio 2022, 16, 100425. [Google Scholar] [CrossRef]
  107. Roh, E.J.; Kim, D.S.; Kim, J.H.; Lim, C.S.; Choi, H.; Kwon, S.Y.; Park, S.Y.; Kim, J.Y.; Kim, H.M.; Hwang, D.Y.; et al. Multimodal therapy strategy based on a bioactive hydrogel for repair of spinal cord injury. Biomaterials 2023, 299, 122160. [Google Scholar] [CrossRef]
  108. King, V.; Alovskaya, A.; Wei, D.; Brown, R.; Priestley, J. The use of injectable forms of fibrin and fibronectin to support axonal ingrowth after spinal cord injury. Biomaterials 2010, 31, 4447–4456. [Google Scholar] [CrossRef]
  109. Wang, S.; Wang, R.; Chen, J.; Yang, B.; Shu, J.; Cheng, F.; Tao, Y.; Shi, K.; Wang, C.; Wang, J.; et al. Controlled extracellular vesicles release from aminoguanidine nanoparticle-loaded polylysine hydrogel for synergistic treatment of spinal cord injury. J. Control. Release 2023, 363, 27–42. [Google Scholar] [CrossRef]
  110. Mu, J.; Wu, J.; Cao, J.; Ma, T.; Li, L.; Feng, S.; Gao, J. Rapid and effective treatment of traumatic spinal cord injury using stem cell derived exosomes. Asian. J. Pharm. Sci. 2021, 16, 806–815. [Google Scholar] [CrossRef]
  111. He, X.; Yang, L.; Dong, K.; Zhang, F.; Liu, Y.; Ma, B.; Chen, Y.; Hai, J.; Zhu, R.; Cheng, L. Biocompatible exosome-modified fibrin gel accelerates the recovery of spinal cord injury by VGF-mediated oligodendrogenesis. J. Nanobiotechnol. 2022, 20, 360. [Google Scholar] [CrossRef]
  112. Liu, X.; Zhang, L.; Xu, Z.; Xiong, X.; Yu, Y.; Wu, H.; Qiao, H.; Zhong, J.; Zhao, Z.; Dai, J.; et al. A functionalized collagen-I scaffold delivers microRNA 21-loaded exosomes for spinal cord injury repair. Acta Biomater. 2022, 154, 385–400. [Google Scholar] [CrossRef]
  113. Zhang, L.; Fan, C.; Hao, W.; Zhuang, Y.; Liu, X.; Zhao, Y.; Chen, B.; Xiao, Z.; Chen, Y.; Dai, J. NSCs migration promoted and drug delivered exosomes-collagen scaffold via a bio-specific peptide for one-step spinal cord injury repair. Adv. Healthc. Mater. 2021, 10, e2001896. [Google Scholar] [CrossRef]
  114. Han, M.; Yang, H.; Lu, X.; Li, Y.; Liu, Z.; Li, F.; Shang, Z.; Wang, X.; Li, X.; Li, J.; et al. Three-dimensional-cultured MSC-derived exosome-hydrogel hybrid microneedle array patch for spinal cord repair. Nano Lett. 2022, 22, 6391–6401. [Google Scholar] [CrossRef]
  115. Wang, C.; Wang, M.; Xia, K.; Wang, J.; Cheng, F.; Shi, K.; Ying, L.; Yu, C.; Xu, H.; Xiao, S.; et al. A bioactive injectable self-healing anti-inflammatory hydrogel with ultralong extracellular vesicles release synergistically enhances motor functional recovery of spinal cord injury. Bioact. Mater. 2021, 6, 2523–2534. [Google Scholar] [CrossRef]
  116. Mu, J.; Li, L.; Wu, J.; Huang, T.; Zhang, Y.; Cao, J.; Ma, T.; Chen, J.; Zhang, C.; Zhang, X.; et al. Hypoxia-stimulated mesenchymal stem cell-derived exosomes loaded by adhesive hydrogel for effective angiogenic treatment of spinal cord injury. Biomater. Sci. 2022, 10, 1803–1811. [Google Scholar] [CrossRef] [PubMed]
  117. Li, L.; Zhang, Y.; Mu, J.; Chen, J.; Zhang, C.; Cao, H.; Gao, J. Transplantation of human mesenchymal stem-cell-derived exosomes immobilized in an adhesive hydrogel for effective treatment of spinal cord injury. Nano Lett. 2020, 20, 4298–4305. [Google Scholar] [CrossRef]
  118. Song, P.; Han, T.; Wu, Z.; Fang, H.; Liu, Y.; Ying, W.; Wang, X.; Shen, C. Transplantation of neural stem cells loaded in an IGF-1 bioactive supramolecular nanofiber hydrogel for the effective treatment of spinal cord injury. Adv. Sci. 2024, 11, e2306577. [Google Scholar] [CrossRef]
  119. Imamura, H.; Tomimaru, Y.; Kobayashi, S.; Harada, A.; Kita, S.; Sasaki, K.; Iwagami, Y.; Yamada, D.; Noda, T.; Takahashi, H.; et al. Adipose-derived stem cells using fibrin gel as a scaffold enhances post-hepatectomy liver regeneration. Sci. Rep. 2025, 15, 6334. [Google Scholar] [CrossRef]
  120. Ahmann, K.A.; Weinbaum, J.S.; Johnson, S.L.; Tranquillo, R.T. Fibrin degradation enhances vascular smooth muscle cell proliferation and matrix deposition in fibrin-based tissue constructs fabricated in vitro. Tissue Eng. Part A 2010, 16, 3261–3270. [Google Scholar] [CrossRef] [PubMed]
  121. Bayat, N.; Ebrahimi-Barough, S.; Ardakan, M.; Ai, A.; Kamyab, A.; Babaloo, H.; Ai, J. Differentiation of human endometrial stem cells into Schwann cells in fibrin hydrogel as 3D culture. Mol. Neurobiol. 2016, 53, 7170–7176. [Google Scholar] [CrossRef] [PubMed]
  122. Chen, Z.; Zhang, H.; Fan, C.; Zhuang, Y.; Yang, W.; Chen, Y.; Shen, H.; Xiao, Z.; Zhao, Y.; Li, X.; et al. Adhesive, stretchable, and spatiotemporal delivery fibrous hydrogels harness endogenous neural stem/progenitor cells for spinal cord injury repair. ACS Nano 2022, 16, 1986–1998. [Google Scholar] [CrossRef] [PubMed]
  123. Jiao, G.; Lou, G.; Mo, Y.; Pan, Y.; Zhang, Z.; Guo, R.; Li, Z. A combination of GDNF and hUCMSC transplantation loaded on SF/AGs composite scaffolds for spinal cord injury repair. Mater. Sci. Eng. C Mater. Biol. Appl. 2017, 74, 230–237. [Google Scholar] [CrossRef]
  124. Yeh, J.; Wang, D.; Cherng, J.; Wang, Y.; Fan, G.; Liou, N.; Liu, J.; Chou, C. A collagen-based scaffold for promoting neural plasticity in a rat model of spinal cord injury. Polymers 2020, 12, 2245. [Google Scholar] [CrossRef]
  125. Reinehr, P.; Diel, L.F.; Diz, F.M.; Balbinot, G.S.; Monteiro, W.F.; Ligabue, R.A.; Lamers, M.L.; Kunrath, M.F. Three-dimensional bioactive collagen scaffolds incorporated with titanate nanotubes for tissue regeneration. Colloids Surf. B Biointerfaces 2025, 252, 114638. [Google Scholar] [CrossRef]
  126. Li, X.; Zhu, X.; Liu, X.; Xu, H.; Jiang, W.; Wang, J.; Chen, F.; Zhang, S.; Li, R.; Chen, X.; et al. The corticospinal tract structure of collagen/silk fibroin scaffold implants using 3D printing promotes functional recovery after complete spinal cord transection in rats. J. Mater. Sci. Mater. Med. 2021, 32, 31. [Google Scholar] [CrossRef]
  127. Liu, S.; Xie, Y.; Wang, L.; Tai, C.; Chen, D.; Mu, D.; Cui, Y.; Wang, B. A multi-channel collagen scaffold loaded with neural stem cells for the repair of spinal cord injury. Neural. Regen. Res. 2021, 16, 2284–2292. [Google Scholar] [CrossRef]
  128. Fan, C.; Yang, W.; Zhang, L.; Cai, H.; Zhuang, Y.; Chen, Y.; Zhao, Y.; Dai, J. Restoration of spinal cord biophysical microenvironment for enhancing tissue repair by injury-responsive smart hydrogel. Biomaterials 2022, 288, 121689. [Google Scholar] [CrossRef] [PubMed]
  129. Chocarro-Wrona, C.; Pleguezuelos-Beltrán, P.; López de Andrés, J.; Antich, C.; de Vicente, J.; Jiménez, G.; Arias-Santiago, S.; Gálvez-Martín, P.; López-Ruiz, E.; Marchal, J.A. A bioactive three-layered skin substitute based on ECM components effectively promotes skin wound healing and regeneration. Mater. Today Bio 2025, 31, 101592. [Google Scholar] [CrossRef]
  130. Tran, K.; Jin, Y.; Bouyer, J.; DeOre, B.; Suprewicz, Ł.; Figel, A.; Walens, H.; Fischer, I.; Galie, P. Magnetic alignment of injectable hydrogel scaffolds for spinal cord injury repair. Biomater. Sci. 2022, 10, 2237–2247. [Google Scholar] [CrossRef]
  131. Zheng, Q.; Yao, J.; Sun, Z.; Li, R.; Zhang, Y.; Jiang, P.; Xie, Y.; Song, X.; Sun, H.; Zhu, D.; et al. Carboxymethyl chitosan/oxidized hyaluronic acid hydrogel-encapsulated hucMSC-derived exosomes for enhanced hepatic regeneration and post-hepatectomy applications. Carbohydr. Polym. 2025, 353, 123248. [Google Scholar] [CrossRef] [PubMed]
  132. Wu, T.; Song, H.; Wang, R.; Wang, W.; Xing, J. A hyaluronic acid nanogels based exosome production factory for tumor photothermal therapy and angiogenesis inhibition. Int. J. Biol. Macromol. 2024, 293, 139363. [Google Scholar] [CrossRef] [PubMed]
  133. Fang, A.; Wang, Y.; Guan, N.; Zuo, Y.; Lin, L.; Guo, B.; Mo, A.; Wu, Y.; Lin, X.; Cai, W.; et al. Porous microneedle patch with sustained delivery of extracellular vesicles mitigates severe spinal cord injury. Nat. Commun. 2023, 14, 4011. [Google Scholar] [CrossRef] [PubMed]
  134. Liang, C.; Dudko, V.; Khoruzhenko, O.; Hong, X.; Lv, Z.P.; Tunn, I.; Umer, M.; Timonen, J.V.I.; Linder, M.B.; Breu, J.; et al. Stiff and self-healing hydrogels by polymer entanglements in co-planar nanoconfinement. Nat. Mater. 2025, 24, 599–606. [Google Scholar] [CrossRef]
  135. Zhang, H.; Yu, R.; Xia, Y.; Liu, J.; Tu, R.; Shi, J.; Dai, H. Effect of magnesium and calcium ions on the strength and biofunctionality of GelMA/SAMA composite hydrogels. J. Mater. Chem. B 2024, 12, 10692–10704. [Google Scholar] [CrossRef]
  136. Galbiati, M.; Maiullari, F.; Ceraolo, M.G.; Bousselmi, S.; Fratini, N.; Gega, K.; Recchia, S.; Ferretti, A.M.; Scala, G.; Costantini, M.; et al. Bioactive hydrogel supplemented with stromal cell-derived extracellular vesicles enhance wound healing. Pharmaceutics 2025, 17, 162. [Google Scholar] [CrossRef] [PubMed]
  137. Maiullari, F.; Milan, M.; Chirivì, M.; Ceraolo, M.G.; Bousselmi, S.; Fratini, N.; Galbiati, M.; Fortunato, O.; Costantini, M.; Brambilla, F.; et al. Enhancing neovascularization post-myocardial infarction through injectable hydrogel functionalized with endothelial-derived EVs. Biofabrication 2024, 16, 045009. [Google Scholar] [CrossRef]
  138. Chen, Z.; Sun, Z.; Fan, Y.; Yin, M.; Jin, C.; Guo, B.; Yin, Y.; Quan, R.; Zhao, S.; Han, S.; et al. Mimicked spinal cord fibers trigger axonal regeneration and remyelination after injury. ACS Nano 2023, 17, 25591–25613. [Google Scholar] [CrossRef]
  139. Azizi, M.; Farahmandghavi, F.; Joghataei, M.T.; Zandi, M.; Imani, M.; Bakhtiari, M.; Omidian, H. ChABC-loaded PLGA nanoparticles: A comprehensive study on biocompatibility, functional recovery, and axonal regeneration in animal model of spinal cord injury. Int. J. Pharm. 2020, 577, 119037. [Google Scholar] [CrossRef]
  140. Chen, L.; Wang, W.; Lin, Z.; Lu, Y.; Chen, H.; Li, B.; Li, Z.; Xia, H.; Li, L.; Zhang, T. Conducting molybdenum sulfide/graphene oxide/polyvinyl alcohol nanocomposite hydrogel for repairing spinal cord injury. J. Nanobiotechnol. 2022, 20, 210. [Google Scholar] [CrossRef]
  141. Nukolova, N.V.; Aleksashkin, A.D.; Abakumova, T.O.; Morozova, A.Y.; Gubskiy, I.L.; Kirzhanova, E.; Abakumov, M.A.; Chekhonin, V.P.; Klyachko, N.L.; Kabanov, A.V. Multilayer polyion complex nanoformulations of superoxide dismutase 1 for acute spinal cord injury. J. Control. Release 2018, 270, 226–236. [Google Scholar] [CrossRef] [PubMed]
  142. Nutt, K.; Dombros-Ryan, Z.; Birea, R.; Franks, E.V.; Eastham, S.; Godwin, M.; Adams, C.F.; Chari, D.M.; Jenkins, S.I. Electrospun polycaprolactone (PCL) nanofibers induce elongation and alignment of co-cultured primary cortical astrocytes and neurons. Micromachines 2025, 16, 256. [Google Scholar] [CrossRef] [PubMed]
  143. Geddes, L.; Themistou, E.; Burrows, J.F.; Buchanan, F.J.; Carson, L. Evaluation of the in vitro cytotoxicity and modulation of the inflammatory response by the bioresorbable polymers poly(D,L-lactide-co-glycolide) and poly(L-lactide-co-glycolide). Acta Biomater. 2021, 134, 261–275. [Google Scholar] [CrossRef]
  144. Wang, W.; Cheng, Z.; Yu, M.; Liu, K.; Duan, H.; Zhang, Y.; Huang, X.; Li, M.; Li, C.; Hu, Y.; et al. Injectable ECM-mimetic dynamic hydrogels abolish ferroptosis-induced post-discectomy herniation through delivering nucleus pulposus progenitor cell-derived exosomes. Nat. Commun. 2025, 16, 3131. [Google Scholar] [CrossRef]
  145. Gao, Z.; Zhang, C.; Xia, N.; Tian, H.; Li, D.; Lin, J.; Mei, X.; Wu, C. Berberine-loaded M2 macrophage-derived exosomes for spinal cord injury therapy. Acta Biomater. 2021, 126, 211–223. [Google Scholar] [CrossRef]
  146. Cao, J.; Zhang, X.; Guo, J.; Wu, J.; Lin, L.; Lin, X.; Mu, J.; Huang, T.; Zhu, M.; Ma, L.; et al. An engineering-reinforced extracellular vesicle-integrated hydrogel with an ROS-responsive release pattern mitigates spinal cord injury. Sci. Adv. 2025, 11, eads3398. [Google Scholar] [CrossRef]
  147. Xia, B.; Gao, X.; Qian, J.; Li, S.; Yu, B.; Hao, Y.; Wei, B.; Ma, T.; Wu, H.; Yang, S.; et al. A novel superparamagnetic multifunctional nerve scaffold: A remote actuation strategy to boost in situ extracellular vesicles production for enhanced peripheral nerve repair. Adv. Mater. 2024, 36, e2305374. [Google Scholar] [CrossRef]
  148. Nie, X.; Liu, Y.; Yuan, T.; Yu, T.; Yun, Z.; Xue, W.; Yu, T.; An, J.; Dai, A.; Wu, K.; et al. Platelet-rich plasma-derived exosomes promote blood-spinal cord barrier repair and attenuate neuroinflammation after spinal cord injury. J. Nanobiotechnol. 2024, 22, 456. [Google Scholar] [CrossRef]
  149. Kim, G.; Chen, X.; Yang, Y. Pathogenic extracellular vesicle (EV) signaling in amyotrophic lateral sclerosis (ALS). Neurotherapeutics 2022, 19, 1119–1132. [Google Scholar] [CrossRef]
  150. Pauwels, J.; Van de Steene, T.; Van de Velde, J.; De Muyer, F.; De Pauw, D.; Baeke, F.; Eyckerman, S.; Gevaert, K. Filter-aided extracellular vesicle enrichment (FAEVEr) for proteomics. Mol. Cell. Proteom. 2025, 24, 100907. [Google Scholar] [CrossRef]
  151. Zhang, J.; Song, H.; Dong, Y.; Li, G.; Li, J.; Cai, Q.; Yuan, S.; Wang, Y.; Song, H. Surface engineering of HEK293 cell-derived extracellular vesicles for improved pharmacokinetic profile and targeted delivery of IL-12 for the treatment of hepatocellular carcinoma. Int. J. Nanomed. 2023, 18, 209–223. [Google Scholar] [CrossRef] [PubMed]
  152. Sabani, B.; Brand, M.; Albert, I.; Inderbitzin, J.; Eichenseher, F.; Schmelcher, M.; Rohrer, J.; Riedl, R.; Lehmann, S. A novel surface functionalization platform to prime extracellular vesicles for targeted therapy and diagnostic imaging. Nanomed 2023, 47, 102607. [Google Scholar] [CrossRef] [PubMed]
  153. Larsen, J.H.; Jensen, I.S.; Svenningsen, P. Benchmarking transcriptome deconvolution methods for estimating tissue- and cell-type-specific extracellular vesicle abundances. J. Extracell. Vesicles 2024, 13, e12511. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Pathological physiology and fundamental mechanisms of SCI at different stages of the injury. (All figures were created with BioRender.com).
Figure 1. Pathological physiology and fundamental mechanisms of SCI at different stages of the injury. (All figures were created with BioRender.com).
Biomedicines 13 01262 g001
Figure 2. The reparative effects of EVs derived from various cell types on SCI. The diagram depicts different cell sources of EVs, including NSCs, hucMSCs, ADSCs, SCs, astrocytes, and macrophages. EVs, administrated with the tail vein injection or lesions injection, exhibit varying levels of efficacy in generating new neurons to restore neural pathways, promoting angiogenesis, axon regeneration, and tissue repair, preventing apoptosis, reducing inflammation, and excessive growth of glial cells.
Figure 2. The reparative effects of EVs derived from various cell types on SCI. The diagram depicts different cell sources of EVs, including NSCs, hucMSCs, ADSCs, SCs, astrocytes, and macrophages. EVs, administrated with the tail vein injection or lesions injection, exhibit varying levels of efficacy in generating new neurons to restore neural pathways, promoting angiogenesis, axon regeneration, and tissue repair, preventing apoptosis, reducing inflammation, and excessive growth of glial cells.
Biomedicines 13 01262 g002
Figure 3. Schematic illustration of EVs binded with biomaterials for SCI. Synthesized composite materials, containing EVs with fibrin gels, collagen scaffolds, and bioactive hydrogels (which are administrated through transplantation or patch), inhibit apoptosis, reduce inflammation, decrease ROS, and promote angiogenesis and axonal regeneration.
Figure 3. Schematic illustration of EVs binded with biomaterials for SCI. Synthesized composite materials, containing EVs with fibrin gels, collagen scaffolds, and bioactive hydrogels (which are administrated through transplantation or patch), inhibit apoptosis, reduce inflammation, decrease ROS, and promote angiogenesis and axonal regeneration.
Biomedicines 13 01262 g003
Table 1. Biomaterials for promoting functional recovery of SCI.
Table 1. Biomaterials for promoting functional recovery of SCI.
MaterialMethod of ModificationSCI ModelOutcomeReference
Fibrin gelHuman umbilical cord MSC-sEV and in situ gelate-encapsulated fibrin glueSpinal cord injury (T9–T11)Alleviate inflammatory and oxidative microenvironment
Induce effective nerve tissue repair and functional recovery
[110]
Fibrin gelEncapsulation of sEVSpinal cord injury (T8–T9)Enhance neurogenesis
Remyelination
[111]
Collagen-I (Col-I) scaffoldImmobilization of CBD peptide fused in Lamp2b on the sEV surfaceSpinal cord injury (T10)Inhibit pro-apoptotic genes
Provide enough space for cell growth or migration
[112]
Collagen BSP specific binding to MSC-sEVsSpinal cord injury (T8)Enhance neural regeneration
Reduce scar deposition
[113]
Gelatin methacryloyl hydrogel3D-sEV hybrid cultureSpinal cord injury (T9–T11)Induce inflammation and glial scar[114]
F127-polycitrate-polyethyleneimine hydrogel (FE)Encapsulation of EVSpinal cord injury (T10)Suppress fibrotic scar formation
Promote remyelination and axonal regeneration
[115]
Hyaluronic acid-hydrogelEncapsulation of MSC-derived sEV, DBM, PDRN, et al.Spinal cord injury (T10)Increase regenerative capacity
Reduce pro-inflammatory responses and restore BSCB disrupted by SCI
Enhance remyelination
[107]
Hyaluronic acid-hydrogelEncapsulation of hypo-sEVSpinal cord injury (T9–T10)Reduce pro-inflammatory responses
Increase angiogenesis capacity and nerve regeneration
[116]
Nanofiber scaffold and hyaluronic acid hydrogel composite patchLoaded with MP and SCs-sEVsSpinal cord injury (T10)Reduce neuronal apoptosis
Inhibit inflammatory reaction
[35]
Peptide-modified adhesive hydrogel (pGel)Encapsulation of hMSC-derived sEVSpinal cord injury (T9–T10)Mitigate inflammation and oxidation
Improve nerve recovery and urinary tissue preservation
[117]
Table 2. Clinical trials investigating the use of diverse biomaterials for SCI treatment.
Table 2. Clinical trials investigating the use of diverse biomaterials for SCI treatment.
NCT NumberStudy TypeStudy PhaseRecruiting StatusBiomaterialsSCI ModelAgesEnrollment
ChiCTR2100043838Interventional0RecruitingNerve regeneration collagen scaffoldSpinal cord injury (C6–T12)18 years to 60 years30
ChiCTR-INR-17012152InterventionalNew treatment measure clinical studyActive, not recruitingFunctional scaffoldComplete spinal cord injury (T1–T11)18 years to 60 years6
NCT03762655InterventionalNot applicableActive, not recruitingNeuro-spinal scaffoldComplete spinal cord injury (T2–T12)16 years to 70 years20
NCT02352077InterventionalPhase 1Unknown statusNeural regeneration collagen scaffoldComplete spinal cord injury (C5–T12)18 years to 65 years30
NCT03966794InterventionalPhase 2Unknown statusFunctional neural regeneration scaffoldComplete spinal cord injury (C4–T12/L1)18 years to 60 years9
NCT03933072InterventionalPhase 2Unknown statusCollagen scaffoldComplete spinal cord injury (C5–T10)16 years to 65 years2
NCT05967325InterventionalNot applicableRecruitingFunctional self-assembling peptide nanofiber hydrogelsComplete spinal cord injury (T1–T12)18 years to 60 years15
NCT04132596InterventionalNot applicableCompletedTranscutaneous hydrogel electrodesSpinal cord injury (C4–T12)18 years and older12
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Jiang, J.; Wang, Z.; Bao, Q.; Chen, S.; Xu, W.; Jiang, J. Extracellular Vesicles as Emerging Therapeutic Strategies in Spinal Cord Injury: Ready to Go. Biomedicines 2025, 13, 1262. https://doi.org/10.3390/biomedicines13051262

AMA Style

Jiang J, Wang Z, Bao Q, Chen S, Xu W, Jiang J. Extracellular Vesicles as Emerging Therapeutic Strategies in Spinal Cord Injury: Ready to Go. Biomedicines. 2025; 13(5):1262. https://doi.org/10.3390/biomedicines13051262

Chicago/Turabian Style

Jiang, Jiali, Ziyi Wang, Qinghua Bao, Shenyuan Chen, Wenrong Xu, and Jiajia Jiang. 2025. "Extracellular Vesicles as Emerging Therapeutic Strategies in Spinal Cord Injury: Ready to Go" Biomedicines 13, no. 5: 1262. https://doi.org/10.3390/biomedicines13051262

APA Style

Jiang, J., Wang, Z., Bao, Q., Chen, S., Xu, W., & Jiang, J. (2025). Extracellular Vesicles as Emerging Therapeutic Strategies in Spinal Cord Injury: Ready to Go. Biomedicines, 13(5), 1262. https://doi.org/10.3390/biomedicines13051262

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop