Skip to main content

Harnessing stem cell-derived exosomes: a promising cell-free approach for spinal cord injury

Abstract

Spinal cord injury (SCI) is a severe injury to the central nervous system that often results in permanent neurological dysfunction. Current treatments have limited efficacy and face challenges in restoring neurological function after injury. Recently, stem cell-derived exosomes have gained attention as an experimental treatment for SCI due to their unique properties, including superior biocompatibility, minimal immunogenicity and non-tumorigenicity. With their potential as a cell-free therapy, exosomes promote SCI repair by enhancing nerve regeneration, reducing inflammation and stabilizing the blood-spinal cord barrier. This review summarizes advances in stem cell-derived exosome research for SCI over the past years, focusing on their mechanisms and future prospects. Despite their promising therapeutic potential, clinical translation remains challenging due to standardization of exosome isolation protocols, compositional consistency and long-term safety profiles that require further investigation.

Introduction

Spinal cord injury (SCI) is a devastating traumatic disease of the central nervous system (CNS), typically caused by mechanical damage resulting from traffic accidents, sports injuries, falls, and self-inflicted violence [1, 2]. The pathophysiology of the secondary injury after SCI is highly complex, involving, i.e., spinal cord ischemia, edema, oxidative stress, inflammatory responses, neuronal apoptosis and demyelination, and excitatory amino acid and ionic imbalances [3, 4]. SCI often results in permanent motor, sensory, and autonomic dysfunction, including paralysis, sensory abnormalities, spasticity, and pain, as well as cardiovascular, bowel, bladder, or sexual dysfunction. High-level spinal cord injuries can lead to life-threatening conditions such as respiratory and cardiac arrest [1, 3]. Current treatment strategies for SCI primarily encompass decompression and stabilization surgery, supportive medical care, and physical rehabilitation therapy [5]. However, their overall efficacy is quite limited, and reversing the neurological damage caused by the injury remains challenging [6]. This significantly diminishes patients’ quality of life and places a heavy economic and psychological burden on families, healthcare systems, and society [1]. As a result, there is a pressing need to discover new and more effective treatment options.

Stem cells—with their self-renewal capacity and ability to differentiate into multiple cell types—offer a promising avenue for SCI treatment by potentially repairing damaged tissues through differentiation into neurons or supportive cells. Consequently, researchers are increasingly focusing on stem cell therapies [7,8,9]. Among the various types of stem cells studied for transplantation, several have demonstrated early progress in cell replacement therapy for SCI [3, 6, 10,11,12]. Preclinical and phase I clinical studies suggest that stem cell therapies can promote recovery from neurodegenerative diseases and neurological injuries through neuroprotection, immunomodulation, neuronal relay formation, and remyelination [8]. However, the efficacy and safety of stem cell transplantation remain under-evaluated, with several challenges yet to be addressed [13]. In this context, exosomes, nano-sized vesicles secreted by stem cells, have emerged as a safer and more controllable alternative. Exosomes not only carry biologically active molecules like proteins, microRNAs (miRNA, miR), and messenger RNAs (mRNA) but also exhibit enhanced circulatory stability, blood-spinal cord barrier (BSCB) penetrability, and reduced immunogenicity compared to parental cells [14, 15]. Recent studies demonstrate that stem cell-derived exosomes promote SCI repair through neural regeneration [16], anti-inflammation [17], immune modulation [18], and BSCB repair [19]. These findings suggest that exosomes have great potential in SCI treatment. This narrative review categorizes stem cell-derived exosomes by their cellular origin and systematically analyzes their repair mechanisms, providing a framework for future translational research.

Mechanisms of SCI

The pathophysiologic mechanisms of SCI are typically divided into two phases: the initial injury and secondary injury cascades, each involving complex biological processes (Fig. 1) [3].

Fig. 1
figure 1

Schematic diagram of spinal cord injury mechanisms

Initial injury

The initial injury is usually directly caused by external trauma (e.g., compression, contusion, or puncture), which results in mechanical damage to the spinal cord tissue. The main effects include neuronal death by mechanical force, which extends to the axons within the spinal cord and leads to loss of conduction function [20, 21]. Damage to blood vessels at the site of injury, resulting in local hemorrhage and disrupting blood supply, thus triggering edema and ischemia [22]. Mechanical compression of the spinal cord due to spinal instability or fractures results in localized edema and hematoma formation, which further aggravates the injury [3].

Secondary injury

Secondary injury cascades are more complex pathological processes that occur after the initial trauma, usually within hours to weeks, leading to further neuronal death and loss of function. The mechanisms involved include an inflammatory response triggered by immune cells such as neutrophils, macrophages, and T cells that release inflammatory factors [e.g., Tumor necrosis factor-α (TNF-α), Interleukin-1β (IL-1β), IL-6], which may help clear necrotic tissue, but also exacerbates nerve damage [23]. Oxidative stress is mediated by free radicals and reactive oxygen species (ROS) that are generated after the injury and might damage neurons and cell membranes, which further exacerbates spinal cord degradation by destroying cellular structures and inducing apoptosis [21]. Apoptosis itself results in programmed cell death of neurons and supportive cells such as oligodendrocytes, further reducing the repair and regenerative capacity of the spinal cord [20]. Calcium overload is a result of dysregulated intracellular calcium levels that activate calcium-dependent enzymes, leading to the breakdown of structural proteins in neurons, further compromising neuronal function and cell membrane integrity [21]. Vascular damage and decreased blood supply after injury cause an ischemic and hypoxic environment, affecting the normal function of the spinal cord and impeding the regenerative process [22]. Glial scarring at the injury site, mediated by proliferating astrocytes that physically block axonal regeneration and repair, hinders nerve recovery after injury [24].

Given the combination of primary and secondary injuries, there is a significant loss of neurons and glial cells in the spinal cord, leading to long-term sensory, motor, and autonomic dysfunction. These impairments are irreversible and severely impact the quality of life of affected patients [1, 3, 21]. In summary, the pathophysiological mechanisms of SCI are highly complex, involving mechanical injury, inflammation, oxidative stress, cell death, and apoptosis. This complexity poses a significant challenge for effective treatment. Notably, the temporal-spatial dynamics of secondary injury create therapeutic windows that are often underutilized in current approaches. For instance, the early inflammatory phase (0–72 h) may require different intervention strategies compared to the subacute remodeling phase (1–4 weeks). This temporal specificity highlights the need for therapeutic agents with dynamic modulatory capacities—a feature potentially achievable through engineered exosomes carrying stage-specific cargo [90].

Stem cell and exosome characterization

Properties and limitations of stem cells

Stem cells are classified into various types, with the stem cells currently used in therapeutic research for SCI including neural stem cells (NSC), mesenchymal stem cells (MSC), embryonic stem cells (ESC), induced pluripotent stem cells (iPSC), and dental pulp stem cell (DPSC), each with distinct regenerative properties [3, 6, 10,11,12, 25, 26]. However, stem cell therapy faces several significant challenges, such as immune rejection, tumorigenic risks, the low survival rate of transplanted cells, and the complexity of the technique itself [7, 9, 10, 12, 21, 25]. As a result, there is an urgent need to explore safer, more controllable, and widely applicable therapeutic strategies.

Advantages of exosomes

Exosomes are membrane-bound vesicles, ranging from 30 to 150 nm in diameter, released by cells through exocytosis; exosome biogenesis is highly intricate (see Fig. 2 for details). Exosomes are rich in proteins, lipids, mRNAs, miRNAs, and many other biologically active molecules [27]. Compared with the direct use of stem cells, exosomes offer several advantages for treating SCI, including high biocompatibility, circulatory stability, low immunogenicity, non-carcinogenicity, editability, capacity to pass the BSCB, and ease of storage [8, 28, 29]. Stem cell-derived exosomes have gradually become a hotspot for SCI therapy research due to their efficiency, safety, and user-friendly properties.

Fig. 2
figure 2

The biogenesis process of exosomes

Exosomes derived from different types of stem cells for SCI treatment

To systematically evaluate the therapeutic potential of stem cell-derived exosomes in SCI, we categorize them based on their cellular origin (NSC, MSC, iPSC, and DPSC) and analyze their distinct repair mechanisms (Fig. 3).

Fig. 3
figure 3

Comparison of the mechanisms of exosomes derived from different cell types for spinal cord injury

NSC-derived exosomes

NSC can differentiate into neurons, astrocytes, and oligodendrocytes [30]. Localized transplantation of NSCs has also been shown to have a therapeutic effect on SCI, the effect that may be related to cytokines such as brain-derived neurotrophic factor (BDNF), vascular endothelial growth factor (VEGF), or nerve growth factor (NGF); however, given that implantation efficiencies are very low (< 5%) and that transplants typically result in a severe inflammatory immune microenvironment, exogenous NSC transplantation exists controversial [31]。 NSC-derived exosomes are enriched in NGF, BDNF, and various miRNAs [31]. These exosomes promote neuronal regeneration and improve functional recovery after SCI by preventing neuronal apoptosis and regulating autophagy, reducing inflammation, and promoting angiogenesis (Fig. 4 & Table 1), are emerging as a new research field.

Fig. 4
figure 4

Origin of exosomes and their related mechanisms in the treatment of spinal cord injury

Table 1 Research on the treatment of SCI with exosomes derived from stem cells

Apoptosis and autophagy regulation

Recent studies have focused on NSC-derived exosomes’ role in regulating apoptosis and autophagy. For instance, NSC-derived exosomes can activate autophagy by targeting the miR-374-5p/STK-4 axis, which inhibits neuronal apoptosis and aids in SCI repair. miR-374-5p regulates autophagy-related genes by modulating STK-4 activity, enhancing autophagy to clear damaged cells and maintain neuron survival [32]. However, these findings are largely based on preclinical models, and their clinical relevance remains uncertain. Moreover, NSC-derived exosomes can increase the expression of autophagy markers like LC3B and beclin-1, facilitating autophagosome formation, reducing SCI damage, and decreasing pro-apoptotic proteins like Bax and caspase-3, while increasing anti-apoptotic proteins like Bcl-2, further mitigating neuronal apoptosis [33]. A recent study utilizing single-cell RNA sequencing and transcriptomic analysis verified that NSC originates from ependymal cells and identified increased expression of Tsc2(compared with the sham group, p = 0.0298), a hub gene in the competing endogenous RNAs (ceRNA) network. The study further explained that following necrotic apoptosis, NSC-derived exosomes can reduce the damage of spinal cord tissue by inhibiting mTORC1 expression, thereby increasing autophagic flux [34]. The balance between autophagy activation and apoptosis inhibition requires further investigation. Future studies should clarify the precise mechanisms, potential side effects, and feasibility of clinical application.

Neuroinflammation suppression

NSC-derived exosomes also reduce pro-inflammatory cytokines such as TNF-α, IL-1β, and IL-6 and inhibit activation of microglia, thus reducing the inflammatory response [33]. Additionally, Ma et al. [35] found that cell viability was higher and axons were longer in IGF-Exo group's PC12 cells than in the injury model group or Nor-Exo group (p < 0.05), which demonstrated that NSC-derived exosomes exposed to IGF-1 could partially suppress neuroinflammation and apoptosis while enhancing pro-neuroregenerative capacities via a miR-219a-2-3p/YY1/NK-kB-dependent mechanism. However, while this finding is promising, the mechanisms involved remain complex and context-dependent, and the long-term effects of exosome-based therapies on neuroinflammation in humans are still unknown. Additionally, the potential off-target effects of targets inhibition and the incomplete understanding of pathway warrant further investigation before clinical applications can be considered.

Angiogenesis promotion

A groundbreaking study, for the first time, discovered that NSC-derived exosomes are enriched with vascular endothelial growth factor (VEGF) A, which promotes microvascular regeneration and tissue healing and reduces tissue cavity formation thus restoring neurological functions following SCI (BMS score in the NSCsCon shRNA-Exos groups and control groups on 28 day = 4.30 ± 0.95 vs. 6.70 ± 1.25, P < 0.01) [36]. This indicated that NSC-derived exosomes may enhance SCI repair by promoting angiogenesis and fostering neural regeneration.

In conclusion, although NSC-derived exosomes show promising potential in SCI repair, their application remains primarily in basic research. The underlying mechanisms are diverse and vary from study to study, with no consensus yet on their therapeutic pathways or standardized use. Future research should focus on unravelling the precise molecular mechanisms of NSC-derived exosomes, identifying key bioactive components and exploring their interactions within the SCI microenvironment. Standardized protocols for exosome isolation, characterization and delivery need to be established to ensure reproducibility and efficacy. In addition, preclinical and clinical studies are essential to evaluate their safety, optimal dosage and therapeutic efficacy, paving the way for their eventual translation into clinical applications [37,38,39].

MSC-derived exosomes

MSC-derived exosomes are one of the most widely studied exosome types due to their diverse sources (e.g., bone marrow, umbilical cord, adipose tissue, etc.) and robust immunomodulatory capabilities [40,41,42]. They promote SCI repair through three core mechanisms: anti-inflammation, angiogenesis/BSCB stabilization, and apoptosis inhibition/nerve regeneration (Fig. 4 & Table 1).

Anti-inflammatory effects

Inflammation plays a critical role in SCI, and the balance between pro-inflammatory and anti-inflammatory components within the local microenvironment of SCI is closely associated with post-injury prognosis. Modulating the microenvironment to suppress pro-inflammatory responses while promoting anti-inflammatory processes represents a key therapeutic strategy for SCI treatment.

The inflammatory cascade following SCI involves a complex interplay of signaling pathways and immune cells. MSC-derived exosomes counteract the inflammatory process through three main synergistic strategies:

Targeting key inflammatory pathways

The TLR4/NF-κB axis is a central driver of neuroinflammation. Bone marrow mesenchymal stem cell (BMSC)-derived exosomes loaded with miR-146a disrupt this pathway by inhibiting IRAK1/TRAF6/NF-κB p65, significantly reducing inflammatory factors in SCI rats (the levels of TNF-α, IL-1β and IL-6 in spinal cord tissues of the miR-146a-EXO group were lower than those of the control group at 1d, 3d, 1, 2 and 4 weeks after surgery, p < 0.05) [43]. This mechanism is corroborated by Wang et al., who employed miR-146a-5p-engineered human mbilical cord mesenchymal stem cell (hUCMSC)-exosomes to achieve similar TLR4/NF-κB suppression [44]. Other studies also found that miR-216a-5p or miR-145-5p carried by BMSC-derived exosomes inactivated the TLR4/NF-κB pathway in rats after SCI, which attenuated the inflammatory response and protected the spinal cord [45,46,47]. Beyond TLR4, UCMSC derived exosomes modified with miR-138-5p can reduce the inflammatory reaction of BV-2 cells through the NLRP3-caspase1 signaling pathway [48]. Notably, UCMSC-derived exosomes also can modulate inflammatory responses through the NF-κB/MAPK signaling pathway [49]. Another recent study demonstrated that hypoxia-preconditioned adipose-derived mesenchymal stem cells (ADSC) derived exosomes reduced the levels of ROS and inflammatory cytokines through the circ-Wdfy3/miR-423-3p/GPX4 signaling pathway [50]. BMSC derived exosomes loaded with miR-9-5p can inhibit LPS-induced inflammation by promoting FGF2 expression by inhibiting HDAC5-mediated deacetylation [51]. Another study found ADSC derived exosomes transfected with LRRC75A-AS1 upregulated the mRNA expression of FDFT1 and inhibited the inflammatory response after SCI via ceRNAs- and RNA-binding proteins-dependent pathways [17]. Although extensive research has focused on the anti-inflammatory effects mediated by the TLR4/NF-κB pathway, with additional studies exploring other pathways such as NLRP3-caspase1 and NF-κB/MAPK, these findings predominantly originate from animal-based models and require further validation for clinical translation.

Reprogramming the immune landscape

Microglia and macrophages dictate inflammatory progression following SCI. Hypoxia-preconditioned BMSC exosomes pivot microglia from pro-inflammatory M1 to anti-inflammatory M2 phenotypes via miR-216a-5p—a single miRNA that concurrently inhibits TLR4/NF-κB and activates PI3K/AKT pathways, in which alleviated the inflammation induced by M1 following SCI [52]. It was found that BMSC-derived exosomes mediate enhanced autophagy and focal death inhibition in macrophages/microglia through the miR-21a-5p/PELI1 axis and attenuate the immune cells related inflammatory response after SCI [53]. Beyond central immunity, peripheral neutrophils exacerbate secondary injury. Exosomes derived from human amniotic MSC (AMSC) carrying miR-125a-3p dismantle neutrophil extracellular traps, reducing myeloperoxidase activity and curbing neutrophil infiltration in the lesion immune micro-landscape [54]. AMSC-derived exosomes significantly decreased ROS, myeloperoxidase activity, and inflammatory cytokines (e.g., TNF-α, IL-6, and IL-1β) in the microenvironment, all of which contribute to neuronal protection [55]. After acute SCI, early intravenous application of hUCMSCs or hUCMSC-extracellular vesicles (EV) significantly dampened the accumulation of Iba1+ cells by ~ 25–30% in both gray and white matter, which significantly reduced the expression of pro-inflammatory cytokines in the spinal cord parenchyma and promoted the recovery of neurological function [56]. These studies collectively demonstrate exosomes’ capacity to modulate and reprogram the immune micro-landscape at injury sites. However, it is critical to note that these findings are derived from rodent models, and significant divergences may exist in human immune environments.

Other effects

BMSC-derived exosomes enriched with miR-17-92 significantly inhibited pro-inflammatory factors, including TNF-α, IL-1β, and IL-6 in spinal cord tissues after SCI and promoted nerve repair [57]. Other studies have also confirmed that miR-146a or miR-137 loaded in BMSC-derived exosomes similarly inhibited the expression of pro-inflammatory cytokines in the injured spinal cord tissues and enhanced locomotor activity and neuronal viability in SCI rats [58, 59]. All above demonstrate some anti-inflammatory effects through RNA-loaded exosomes, but the lack of clearly defined mechanisms limits their translational relevance for future clinical applications.

While most current research on MSC-derived exosomes for anti-inflammation are still in preclinical stages and lacks large-scale clinical validation, these findings highlight the significant potential of MSC-derived exosomes to mitigate SCI-induced inflammation and improve neurological recovery. Future research should prioritize clinical safety and efficacy studies to facilitate the translation of these therapies into clinical practice.

Angiogenesis and BSCB repair

Following SCI, vascular damage leads to ischemia, hemorrhage, and immune cell infiltration, which not only exacerbates the secondary injury but also further worsens neurological function. Thus, promoting angiogenesis is crucial for tissue survival and regeneration [60]. Research shows that MSC-derived exosomes significantly enhance both angiogenesis and functional recovery. For instance, exosomes derived from placental mesenchymal stem cells (PMSC) promoted endothelial cells formatted capillary-like structures in vitro and stimulated neo-angiogenesis in vivo, improving neurological outcomes in SCI mice (compared with the control group, BMS scores improved significantly in the hPMSCs-Exos group beginning 21 days post SCI, p < 0.05) [61]. Hypoxia-pretreated MSC-derived exosomes could significantly enhance angiogenesis and functional recovery by increasing VEGF expression via HIF-1α upregulation [62]. Additionally, Huang et al. [63] found that exosomes derived from miR-126-modified MSC significantly promoted angiogenesis post-SCI by inhibiting SPRED1 and PIK3R2. Another recent study demonstrated that ADSC-derived exosomes inhibit ferroptosis through the NRF2/SLC7A11/GPX4 pathway, promoting angiogenesis after SCI [64].

Preserving the integrity of the BSCB is critical to prevent secondary injury processes like edema, oxidative stress, and inflammation [60]. One study suggested that exosomes derived from BMSC improve the integrity of the BSCB by activating the NF-κB p65 signaling pathway, inhibiting pericyte migration, and thus promoting functional recovery after SCI [65]. On the other hand, BMSC-derived exosomes restored tight junctions among cells through the TIMP2/MMP pathway, mitigating the disruption of the BSCB and improving functional recovery after SCI [66]. Another recent study reported that RGD-CD146CD271-modified UCMSC-derived exosomes specifically target vascular endothelial cells at the site of injury, effectively inhibiting the expression of MLCK through miR-501-5p, and thus reducing the disruption of tight junctions, stabilizing the BSCB and ultimately promoting the recovery of neurological functions in SCI mice [19].

In summary, the above findings highlight the therapeutic potential of MSC-derived exosomes in promoting angiogenesis and maintaining BSCB integrity. However, a previous study has revealed differences in post-SCI angiogenesis rates even between rats and mice [67], suggesting potentially greater interspecies divergence in humans. Thus, future work should prioritize human-relevant models (e.g. primates), multi-omics analysis of endothelial junction dynamics, and clinically translatable exosome engineering strategies that integrate biomechanical cues.

Inhibition of apoptosis and promotion of nerve regeneration

Apoptosis, a critical mechanism for neuron and supporting cell loss following SCI, can be effectively minimized by inhibiting the cell death signaling pathway. This not only reduces neuron loss but also promotes nerve and axon regeneration, offering a promising strategy for SCI treatment. One study has shown that exosomes secreted by miR-126-modified BMSC can significantly reduce the expression of pro-apoptotic proteins Bax and cleaved caspase-3, while increasing anti-apoptotic Bcl-2 levels, effectively inhibiting apoptosis. These exosomes also enhance nerve regeneration by upregulating Nestin and SOX2 expression [63]. Another study confirmed that BMSC-derived exosomes could inhibit neuronal apoptosis by stimulating the Wnt/β-catenin pathway [92]. Several studies have also revealed that miR-21, contained in MSC-derived exosomes, exerts anti-apoptotic effects through various mechanisms [93,94,95]. While the anti-apoptotic effect of miR-21 has been increasingly recognized, further investigation and validation into its specific molecular pathways is urgently needed to advance our understanding. Moreover, exosomes from hypoxia-induced ADSC, enriched with miR-499-5p, have been found to target JNK3, thereby regulating the JNK3/c-Jun apoptosis pathway and reducing neuronal apoptosis after SCI [96]. This underscores the need for continued research into the specific molecular pathways of exosomes. In addition to preventing cell apoptosis, MSC-derived exosomes are key in promoting neural regeneration. For example, human PMSC-derived exosomes improve motor function after SCI by activating endogenous neural progenitor cells and neurogenesis, likely involving the activation of the MEK/ERK/CREB pathway [68]. Wang et al. [69] reported that miR-199a-3p/145-5p in UCMSC-derived exosomes targets the NGF/TrkA signaling pathway, inducing axon growth by increasing the level of TrkA ubiquitination. Besides, it was shown that let-7a-5p in MSC-derived exosomes promotes neuronal regeneration and improves neurological recovery in SCI rats by modulating the HMGA2/SMAD2 axis [70].

Although MSC-derived exosomes are the most extensively studied in the field of SCI and show significant therapeutic potential, particularly in inhibiting neuronal apoptosis, promoting neural regeneration and enhancing functional recovery, they face similar challenges as NPC-derived exosomes. Despite some preliminary guidance [37,38,39], the complex mechanisms underlying the effects of MSC-derived exosomes, coupled with the fact that researchers have often focused on narrow, specific areas of interest, have led to inconsistent results and a lack of replicability across studies.

iPSC-derived exosomes

iPSCs, derived from reprogrammed somatic cells, have a differentiation potential similar to ESC without the associated ethical concerns, making them particularly promising for clinical applications. Recent research has increasingly focused on the use of iPSC-derived exosomes for treating SCI (Fig. 4 & Table 1). For example, miR-199b-5p in iPSC-derived exosomes has been found to activate the PI3K signaling pathway by regulating hepatocyte growth factor, facilitating macrophage polarization from M1 to M2, and thereby reducing inflammation and supporting SCI recovery [71]. Recent finding suggested that engineered iPSC-derived exosomes containing miR-23b, miR-21-5p, and miR-199b-5p could suppress neuronal inflammation triggered by LPS and IFN γ, significantly improving recovery after SCI [72].Another study highlighted that let-7b-5p, enriched in iPSC-derived exosomes, effectively reduces pyroptosis of microglia/macrophages, promoting axonal growth while also inhibiting the expression of the let-7b-5p target gene LRIG3 and modulating the PI3K/Akt pathway to mitigate oxidative stress and inflammatory damage [73].

Although research on iPSC-derived exosomes for SCI repair remains relatively limited, their potentially great application value has been preliminarily demonstrated. Such iPSC-derived exosomes not only possess strong neuroprotective effects but also carry growth-promoting factors and various miRNAs that support neuronal survival and regeneration. Thus, iPSC-derived exosomes are poised to play an increasing role in SCI treatment, warranting further exploration and deeper investigation.

DPSC-derived exosomes

Dental-derived stem cell transplantation has expanded to using DPSC-derived exosomes, showing their potential in treating SCI. One study demonstrated that exosomes derived from DPSC could target the HMGB1/TLR4/MyD88/NF-κB pathway, effectively inhibiting TLR4, MyD88, and NF-κB expression during cerebral ischemia/reperfusion injury. This inhibition reduced pro-inflammatory cytokines such as IL-6, IL-1β, and TNF-α, highlighting DPSC-derived exosome’s protective role in brain injury repair and anti-inflammation [74]. Liu et al. [26] were the first to explore the therapeutic potential of DPSC-derived exosomes for SCI repair. Their findings showed that DPSC-derived exosomes reduce macrophage M1 polarization by inhibiting the ROS-MAPK-NFκB P65 signaling pathway, thereby decreasing inflammation and nerve damage while promoting functional recovery (LPS group vs. LPS + EXO group: 33.67 ± 1.79 vs. 21.43 ± 1.922, p < 0.05). This suggested that DPSC-derived exosomes can effectively modulate inflammation and mitigate nerve injury following SCI. What makes DPSC particularly attractive is their noninvasive extraction from discarded teeth without ethical concerns. Given these advantages, DPSC-derived exosomes possess promise as a safe and effective therapeutic strategy for clinical applications in treating SCI. Future studies may be able to turn to the use of DPSC-derived exosomes.

From bench to bedside: bridging the translational gap

To date, only one clinical trial has been registered and reported for exosome-based SCI therapies [75, 76]. This trial marks a critical milestone in the translational journey, providing the first clinical evidence for the safety and potential efficacy of intrathecal allogeneic HUC-MSC-exosomes in complete subacute SCI patients. Hower, in this single-center phase I, small-sample, uncontrolled study, the patients received other interventions, such as rehabilitation, which precluded an accurate assessment of the clinical efficacy of exosomes, reflecting the nascent stage of the field [77]. While preclinical studies and the above phase I clinical trial have demonstrated the therapeutic potential of stem cell-derived exosomes in SCI models and patients, there are still many challenges ahead, and a long way to go in the future [39].

Clinical challenges in exosome translation

Standardization of production and characterization

Current exosome isolation methods—ultracentrifugation (UC), size-exclusion chromatography (SEC), and polymer-based precipitation—vary significantly in yield, purity, and scalability [39, 78,79,80]. For instance, UC, the widely used method, often co-isolates protein aggregates, while SEC requires specialized equipment. These inconsistencies hinder reproducibility across studies and complicate regulatory approval. Emerging technologies, such as microfluidic sorting and immunoaffinity capture, show promise for high-purity exosome isolation but remain cost-prohibitive for large-scale clinical use. All of these issues need to be addressed to allow for repeatability and comparability between studies.

Optimization and solvent compatibility

Preclinical studies report exosome doses ranging from 20 μg to 1 mg/kg, with no consensus on optimal dosing. For example, intravenous doses of 100–200 μg in rodent models improved functional recovery, but translating these findings to humans requires careful scaling. Additionally, exosomes are often suspended in phosphate-buffered saline or plasma-mimicking solutions, yet the ideal solvent-to-exosome ratio remains undefined (Table 1), which is also a question that deserves to be explored in depth [6, 31].

Delivery route and bioavailability

Intravenous administration, though minimally invasive, results in low exosome accumulation at the injury site due to systemic distribution and rapid clearance [81]. Intranasal administration may be an effective option for the treatment of CNS disorders that require crossing the blood–brain barrier or BSCB. However, it is important to note that the bioavailability of intranasally administered drugs may be affected by a number of factors, including the physicochemical properties of the drug and its ability to be absorbed within the nasal cavity [82]. Localized approaches, such as intrathecal or intralesional injection, enhance bioavailability but require invasive procedures and precise dosing. For instance, studies using intraspinal exosome delivery in rats achieved significant neuroprotection, but local anatomical abnormalities in clinically injured patients highlighted technical complexities in clinical translation.

Dose target specificity and species differences

Preclinical models predominantly target pathways like NF-κB, TLR4, STK-4, GPX4 and so on (Fig. 4 & Table 1), yet human SCI pathology involves pathways or targets that may differ. Species-specific differences in exosome biodistribution and target engagement further complicate extrapolation to humans. For example, miR-146a-5p, effective in rodent microglial modulation, may exhibit altered efficacy in human immune cells. And single-cell sequencing and others may be used more often in the future in conjunction with exosomes for target selection in SCI therapy [83].

Future directions

To bridge the translational gap, future efforts should focus on standardized protocols, targeted delivery systems or routines, personalized medicine, collaborative clinical trials, such as establishing consensus guidelines for exosome isolation, characterization, and dosing.

At the same time, it may be difficult to achieve good efficacy with exosomes alone. Rather, the combination of stem cell-derived exosomes with other approaches for the treatment of SCI may become a more favorable solution. Several studies have been conducted to deliver stem-cell derived exosomes for SCI treatment by using biomaterial scaffolds with favorable results. Localized delivery of an adhesive hydrogel to SCI lesions resulted in significant recovery of neurological function in SCI rats after 28 days (the treatment groups’ BBB score of 6.44 ± 1.64 was significantly superior to that of the blank groups’ 3.11 ± 2.13 (p < 0.05)) [84]. In another study, the preservation and controlled release of EV at the site of SCI for up to 56 days was effectively facilitated by the development of a special hydrogel that inhibited reactive fibrotic scarring, reduced inflammatory response, and facilitated myelin sheath regeneration and axonal regeneration, all of which synergistically significantly enhanced tissue repair and recovery of motor function after SCI [81]. A further study used methacrylate hydrogel (GelMA) as a supportive material and loads it with 3D-cultured MSC-derived exosomes to autonomously construct a micro-needle array patch (GelMA-MN@3D-Exo) for SCI repair. The patch significantly improved the retention and release of 3D-Exo at the injury site, reducing SCI-induced neuroinflammation and glial scar formation. [85]. In the latest study, GelMA hydrogel was used to encapsulate and load MSC-derived small extracellular vesicles (sEV) carrying berberine for SCI treatment. This formulation promoted nerve regeneration and axon growth and improved functional recovery by inhibiting local inflammation as well as fibroblast proliferation and migration [86]. In addition, exosomes derived from human menstrual blood stem cells combined with hyperbaric oxygen therapy significantly reduced pro-inflammatory cytokine levels [87]. Therefore, in the future, it may be possible to combine stem cell-derived exosomes with other biomaterials, drugs or hyperbaric oxygen therapy for the treatment of spinal cord injuries for optimal efficacy.

Moreover, tailoring exosome sources (e.g., BMSCs vs. iPSCs) and cargo (e.g., miRNA or protein enrichment) based on injury severity and patient-specific biomarkers [42, 88, 89] and expanding multi-center trials with standardized endpoints are also important factors to further advance the field.

Summary and prospects

SCI remains a formidable challenge in regenerative medicine, but stem cell-derived exosomes offer a cell-free therapeutic strategy with multifaceted benefits, including neuroprotection, immunomodulation, and angiogenesis. Despite promising preclinical outcomes, clinical adoption is hindered by methodological variability in exosome production, incomplete mechanistic understanding, and unresolved safety concerns. Recent advances in exosome engineering and delivery systems, such as CRISPR-modified exosomes [91] or hydrogel encapsulation [81, 84, 85], hold potential to enhance therapeutic precision. However, large-scale clinical validation and regulatory alignment are imperative to transform these innovations into viable therapies. Collaborative efforts between researchers, clinicians, and regulatory bodies will be pivotal in overcoming these barriers, ultimately delivering hope for SCI patients worldwide.

Data availability

Not applicable.

Abbreviations

SCI:

Spinal cord injury

CNS:

Central nervous system

ESC:

Embryonic stem cells

iPSC:

Induced pluripotent stem cells

MSC:

Mesenchymal stem cells

NSC:

Neural stem cells

miRNA, miR:

MicroRNAs

mRNA:

Messenger RNAs

BSCB:

Blood-spinal cord barrier

TNF-α:

Tumor necrosis factor-α

IL:

Interleukin

ROS:

Free radicals and reactive oxygen species

DPSC:

Dental pulp stem cell

NGF:

Nerve growth factor

Bcl-2:

B cell lymphoma-2

ceRNA:

Competing endogenous RNAs

VEGF:

Vascular endothelial growth factor

AMSC:

Amniotic mesenchymal stem cells

EV:

Extracellular vesicles

ADSC:

Adipose-derived mesenchymal stem cells

BMSC:

Bone marrow mesenchymal stem cells

UCMSC:

Umbilical cord mesenchymal stem cells

LPS:

Lipopolysaccharide

PMSC:

Placental mesenchymal stem cells

References

  1. Badhiwala JH, Wilson JR, Fehlings MG. Global burden of traumatic brain and spinal cord injury. Lancet Neurol. 2019;18:24–5. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/S1474-4422(18)30444-7.

    Article  PubMed  Google Scholar 

  2. Hwang J, Jang S, Kim C, Lee S, Jeong H. Role of stem cell-derived exosomes and microRNAs in spinal cord injury. Int J Mol Sci. 2023. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/ijms241813849.

    Article  PubMed  PubMed Central  Google Scholar 

  3. Quadri SA, Farooqui M, Ikram A, Zafar A, Khan MA, Suriya SS, et al. Recent update on basic mechanisms of spinal cord injury. Neurosurg Rev. 2020;43:425–41. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s10143-018-1008-3.

    Article  PubMed  Google Scholar 

  4. Khalatbary A. Stem cell-derived exosomes as a cell free therapy against spinal cord injury. Tissue Cell. 2021;71:101559. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.tice.2021.101559.

    Article  CAS  PubMed  Google Scholar 

  5. Kwon B, Tetreault L, Evaniew N, Skelly A, Fehlings M. AO spine/praxis clinical practice guidelines for the management of acute spinal cord injury: an introduction to a focus issue. Global Spine J. 2024;14:5s–9s. https://doiorg.publicaciones.saludcastillayleon.es/10.1177/21925682231189928.

    Article  PubMed  PubMed Central  Google Scholar 

  6. Shang Z, Wanyan P, Wang M, Zhang B, Cui X, Wang X. Stem cell-derived exosomes for traumatic spinal cord injury: a systematic review and network meta-analysis based on a rat model. Cytotherapy. 2024;26:1–10. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.jcyt.2023.09.002.

    Article  PubMed  Google Scholar 

  7. Assinck P, Duncan GJ, Hilton BJ, Plemel JR, Tetzlaff W. Cell transplantation therapy for spinal cord injury. Nat Neurosci. 2017;20:637–47. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/nn.4541.

    Article  CAS  PubMed  Google Scholar 

  8. Jabermoradi S, Paridari P, Ramawad H, Gharin P, Roshdi S, Toloui A, et al. Stem cell-derived exosomes as a therapeutic option for spinal cord injuries a systematic review and meta-analysis. Arch Acad Emerg Med. 2025;13:e2. https://doiorg.publicaciones.saludcastillayleon.es/10.22037/aaem.v12i1.2261.

    Article  PubMed  Google Scholar 

  9. Mukhamedshina Y, Shulman I, Ogurcov S, Kostennikov A, Zakirova E, Akhmetzyanova E, et al. Mesenchymal stem cell therapy for spinal cord contusion: a comparative study on small and large animal models. Biomolecules. 2019;9:811.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Maldonado-Soto AR, Oakley DH, Wichterle H, Stein J, Doetsch FK, Henderson CE. Stem cells in the nervous system. Am J Phys Med Rehabil. 2014;93:S132–44. https://doiorg.publicaciones.saludcastillayleon.es/10.1097/phm.0000000000000111.

    Article  PubMed  PubMed Central  Google Scholar 

  11. Aboul-Soud M, Alzahrani A, Mahmoud A. Induced pluripotent stem cells (iPSCs)-roles in regenerative therapies disease modelling and drug screening. Cells. 2021. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/cells10092319.

    Article  PubMed  PubMed Central  Google Scholar 

  12. Goel A. Stem cell therapy in spinal cord injury: hollow promise or promising science? J Craniovertebral Junction Spine. 2016;7:121–6. https://doiorg.publicaciones.saludcastillayleon.es/10.4103/0974-8237.181880.

    Article  Google Scholar 

  13. Fan X, Wang J, Lin X, Zhang L. Stem cell transplantation for spinal cord injury: a meta-analysis of treatment effectiveness and safety. Neural Regen Res. 2017;12:815–25. https://doiorg.publicaciones.saludcastillayleon.es/10.4103/1673-5374.206653.

    Article  PubMed  PubMed Central  Google Scholar 

  14. Huang W, Lin M, Yang C, Wang F, Zhang M, Gao J, et al. Rat bone mesenchymal stem cell-derived exosomes loaded with miR-494 promoting neurofilament regeneration and behavioral function recovery after spinal cord injury. Oxid Med Cell Longev. 2021;2021:1634917. https://doiorg.publicaciones.saludcastillayleon.es/10.1155/2021/1634917.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Ke W, Afonin K. Exosomes as natural delivery carriers for programmable therapeutic nucleic acid nanoparticles (NANPs). Adv Drug Deliv Rev. 2021;176:113835. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.addr.2021.113835.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Soleimani A, Oraee YS, Pedram M, Saadinam F, Rasaee M, Soleimani M. Intrathecal injection of human placental mesenchymal stem cells derived exosomes significantly improves functional recovery in spinal cord injured rats. Mol Biol Rep. 2024;51:193. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s11033-023-08972-7.

    Article  CAS  PubMed  Google Scholar 

  17. Xing X, Xu P, Xing X, Xu Z, Huang Z, Li Z, et al. Effects of ADSC-derived exosome LRRC75A-AS1 on anti-inflammatory function after SCI. Appl Biochem Biotechnol. 2024. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s12010-023-04836-9.

    Article  PubMed  Google Scholar 

  18. Guan P, Fan L, Zhu Z, Yang Q, Kang X, Li J, et al. M2 microglia-derived exosome-loaded electroconductive hydrogel for enhancing neurological recovery after spinal cord injury. J Nanobiotechnology. 2024;22:8. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12951-023-02255-w.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Xie Y, Sun Y, Liu Y, Zhao J, Liu Q, Xu J, 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;17:18008–24. https://doiorg.publicaciones.saludcastillayleon.es/10.1021/acsnano.3c04423.

    Article  CAS  PubMed  Google Scholar 

  20. Hachem LD, Fehlings MG. Pathophysiology of spinal cord injury. Neurosurg Clin N Am. 2021;32:305–13. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.nec.2021.03.002.

    Article  PubMed  Google Scholar 

  21. Lima R, Monteiro A, Salgado AJ, Monteiro S, Silva NA. Pathophysiology and therapeutic approaches for spinal cord injury. Int J Mol Sci. 2022;23:13833.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Popa C, Popa F, Grigorean VT, Onose G, Sandu AM, Popescu M, et al. Vascular dysfunctions following spinal cord injury. J Med Life. 2010;3:275–85.

    PubMed  PubMed Central  Google Scholar 

  23. Ulndreaj A, Chio J, Ahuja C, Fehlings M. Modulating the immune response in spinal cord injury. Expert Rev Neurother. 2016;16:1127–9. https://doiorg.publicaciones.saludcastillayleon.es/10.1080/14737175.2016.1207532.

    Article  CAS  PubMed  Google Scholar 

  24. Tran A, Warren P, Silver J. New insights into glial scar formation after spinal cord injury. Cell Tissue Res. 2022;387:319–36. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s00441-021-03477-w.

    Article  PubMed  Google Scholar 

  25. Wen X, Jiang W, Li X, Liu Q, Kang Y, Song B. Advancements in spinal cord injury repair: insights from dental-derived stem cells. Biomedicines. 2024. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/biomedicines12030683.

    Article  PubMed  PubMed Central  Google Scholar 

  26. Liu C, Hu F, Jiao G, Guo Y, Zhou P, Zhang Y, et al. Dental pulp stem cell-derived exosomes suppress M1 macrophage polarization through the ROS-MAPK-NFκB P65 signaling pathway after spinal cord injury. J Nanobiotechnology. 2022;20:65. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12951-022-01273-4.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Peng H, Ji W, Zhao R, Yang J, Lu Z, Li Y, et al. Exosome: a significant nano-scale drug delivery carrier. J Mater Chem B. 2020;8:7591–608. https://doiorg.publicaciones.saludcastillayleon.es/10.1039/d0tb01499k.

    Article  CAS  PubMed  Google Scholar 

  28. Zhang C. Exosomes derived from mesenchymal stem cells: therapeutic opportunities for spinal cord injury. Bull Exp Biol Med. 2024;176:716–21. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s10517-024-06095-y.

    Article  CAS  PubMed  Google Scholar 

  29. Ye Z, Zheng Y, Li N, Zhang H, Li Q, Wang X. Repair of spinal cord injury by bone marrow mesenchymal stem cell-derived exosomes: a systematic review and meta-analysis based on rat models. Front Mol Neurosci. 2024;17:1448777. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fnmol.2024.1448777.

    Article  PubMed  PubMed Central  Google Scholar 

  30. Bond Allison M, Ming G-l, Song H. Adult mammalian neural stem cells and neurogenesis: five decades later. Cell Stem Cell. 2015;17:385–95. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.stem.2015.09.003.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Li Y, Fang B. Neural stem cell-derived extracellular vesicles: the light of central nervous system diseases. Biomed Pharmacother. 2023;165:115092. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.biopha.2023.115092.

    Article  CAS  PubMed  Google Scholar 

  32. 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;22:411–21.

    CAS  PubMed  PubMed Central  Google Scholar 

  33. Rong Y, Liu W, Wang J, Fan J, Luo Y, Li L, et al. 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. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41419-019-1571-8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Li S, Li J, Chen G, Lin T, Zhang P, Tong K, et al. Exosomes originating from neural stem cells undergoing necroptosis participate in cellular communication by inducing TSC2 upregulation of recipient cells following spinal cord injury. Neural Regen Res. 2024. https://doiorg.publicaciones.saludcastillayleon.es/10.4103/nrr.Nrr-d-24-00068.

    Article  PubMed  PubMed Central  Google Scholar 

  35. Ma K, Xu H, Zhang J, Zhao F, Liang H, Sun H, et al. Insulin-like growth factor-1 enhances neuroprotective effects of neural stem cell exosomes after spinal cord injury via an miR-219a-2-3p/YY1 mechanism. Aging (Albany NY). 2019;11:12278–94. https://doiorg.publicaciones.saludcastillayleon.es/10.18632/aging.102568.

    Article  CAS  PubMed  Google Scholar 

  36. Zhong D, Cao Y, Li C, Li M, Rong Z, Jiang L, et al. Neural stem cell-derived exosomes facilitate spinal cord functional recovery after injury by promoting angiogenesis. Exp Biol Med (Maywood). 2020;245:54–65. https://doiorg.publicaciones.saludcastillayleon.es/10.1177/1535370219895491.

    Article  CAS  PubMed  Google Scholar 

  37. Lotvall J, Hill AF, Hochberg F, Buzas EI, Di Vizio D, Gardiner C, et al. Minimal experimental requirements for definition of extracellular vesicles and their functions: a position statement from the International Society for Extracellular Vesicles. J Extracell Vesicles. 2014;3:26913. https://doiorg.publicaciones.saludcastillayleon.es/10.3402/jev.v3.26913.

    Article  PubMed  Google Scholar 

  38. Thery C, Witwer KW, Aikawa E, Alcaraz MJ, Anderson JD, Andriantsitohaina R, et al. Minimal information for studies of extracellular vesicles 2018 (MISEV2018): a position statement of the International Society for Extracellular Vesicles and update of the MISEV2014 guidelines. J Extracell Vesicles. 2018;7:1535750. https://doiorg.publicaciones.saludcastillayleon.es/10.1080/20013078.2018.1535750.

    Article  PubMed  PubMed Central  Google Scholar 

  39. Welsh J, Goberdhan D, O’Driscoll L, Buzas E, Blenkiron C, Bussolati B, et al. Minimal information for studies of extracellular vesicles (MISEV2023): from basic to advanced approaches. J Extracell Vesicles. 2024;13:e12404. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/jev2.12404.

    Article  PubMed  PubMed Central  Google Scholar 

  40. Ren Z, Qi Y, Sun S, Tao Y, Shi R. Mesenchymal stem cell-derived exosomes: hope for spinal cord injury repair. Stem Cells Dev. 2020;29:1467–78. https://doiorg.publicaciones.saludcastillayleon.es/10.1089/scd.2020.0133.

    Article  CAS  PubMed  Google Scholar 

  41. Li Y, Luo W, Meng C, Shi K, Gu R, Cui S. Exosomes as promising bioactive materials in the treatment of spinal cord injury. Stem Cell Res Ther. 2024;15:335. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s13287-024-03952-5.

    Article  PubMed  PubMed Central  Google Scholar 

  42. Cheng L, You C, Peng C, Ren J, Guo K, Liu T. Mesenchymal stem cell-derived exosomes as a new drug carrier for the treatment of spinal cord injury: a review. Chin J Traumatol. 2024;27:134–46. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.cjtee.2024.03.009.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Tang Q, Chen W, Ke H, Lan C. Optical imaging detection of extracellular vesicles of miR-146 modified bone marrow mesenchymal stem cells promoting spinal cord injury repair. SLAS Technol. 2024;29:100172. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.slast.2024.100172.

    Article  CAS  PubMed  Google Scholar 

  44. Wang X, Yang Y, Li W, Hao M, Xu Y. Umbilical mesenchymal stem cell-derived exosomes promote spinal cord functional recovery through the miR-146b/TLR4 -mediated NF-κB p65 signaling pathway in rats. Biochem Biophys Rep. 2023;35:101497. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.bbrep.2023.101497.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Xue H, Ran B, Li J, Wang G, Chen B, Mao H. Bone marrow mesenchymal stem cell exosomes-derived microRNA-216a-5p on locomotor performance, neuronal injury, and microglia inflammation in spinal cord injury. Front Cell Dev Biol. 2023;11:1227440. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fcell.2023.1227440.

    Article  PubMed  PubMed Central  Google Scholar 

  46. Jiang Z, Zhang J. Mesenchymal stem cell-derived exosomes containing miR-145-5p reduce inflammation in spinal cord injury by regulating the TLR4/NF-κB signaling pathway. Cell Cycle (Georgetown, Tex). 2021;20:993–1009. https://doiorg.publicaciones.saludcastillayleon.es/10.1080/15384101.2021.1919825.

    Article  CAS  PubMed  Google Scholar 

  47. Fan L, Dong J, He X, Zhang C, Zhang T. Bone marrow mesenchymal stem cells-derived exosomes reduce apoptosis and inflammatory response during spinal cord injury by inhibiting the TLR4/MyD88/NF-κB signaling pathway. Hum Exp Toxicol. 2021;40:1612–23. https://doiorg.publicaciones.saludcastillayleon.es/10.1177/09603271211003311.

    Article  CAS  PubMed  Google Scholar 

  48. Xiao Y, Hu X, Jiang P, Qi Z. Thermos-responsive hydrogel system encapsulated engineered exosomes attenuate inflammation and oxidative damage in acute spinal cord injury. Front Bioeng Biotechnol. 2023;11:1216878. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fbioe.2023.1216878.

    Article  PubMed  PubMed Central  Google Scholar 

  49. Luan Z, Liu J, Li M, Wang Y, Wang Y. Exosomes derived from umbilical cord-mesenchymal stem cells inhibit the NF-κB/MAPK signaling pathway and reduce the inflammatory response to promote recovery from spinal cord injury. J Orthop Surg Res. 2024;19:184. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s13018-024-04651-w.

    Article  PubMed  PubMed Central  Google Scholar 

  50. Shao M, Ye S, Chen Y, Yu C, Zhu W. Exosomes from hypoxic ADSCs ameliorate neuronal damage post spinal cord injury through circ-Wdfy3 delivery and inhibition of ferroptosis. Neurochem Int. 2024;177:105759. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.neuint.2024.105759.

    Article  CAS  PubMed  Google Scholar 

  51. He X, Zhang J, Guo Y, Yang X, Huang Y, Hao D. Exosomal miR-9-5p derived from BMSCs alleviates apoptosis, inflammation and endoplasmic reticulum stress in spinal cord injury by regulating the HDAC5/FGF2 axis. Mol Immunol. 2022;145:97–108. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.molimm.2022.03.007.

    Article  CAS  PubMed  Google Scholar 

  52. Liu W, Rong Y, Wang J, Zhou Z, Ge X, Ji C, et al. Exosome-shuttled miR-216a-5p from hypoxic preconditioned mesenchymal stem cells repair traumatic spinal cord injury by shifting microglial M1/M2 polarization. J Neuroinflamm. 2020;17:47. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12974-020-1726-7.

    Article  CAS  Google Scholar 

  53. Gu J, Wu J, Wang C, Xu Z, Jin Z, Yan D, et al. BMSCs-derived exosomes inhibit macrophage/microglia pyroptosis by increasing autophagy through the miR-21a-5p/PELI1 axis in spinal cord injury. Aging (Albany NY). 2024;16:5184–206. https://doiorg.publicaciones.saludcastillayleon.es/10.18632/aging.205638.

    Article  CAS  PubMed  Google Scholar 

  54. Morishima Y, Kawabori M, Yamazaki K, Takamiya S, Yamaguchi S, Nakahara Y, et al. Intravenous administration of mesenchymal stem cell-derived exosome alleviates spinal cord injury by regulating neutrophil Extracellular trap formation through exosomal miR-125a-3p. Int J Mol Sci. 2024. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/ijms25042406.

    Article  PubMed  PubMed Central  Google Scholar 

  55. Zhou H, Wang J, Zhao P, Le D, Cai S, Mao G. Intravenous administration exosomes derived from human amniotic mesenchymal stem cells improves neurological recovery after acute traumatic spinal cord injury in rats. Iran J Basic Med Sci. 2024;27:1284–92. https://doiorg.publicaciones.saludcastillayleon.es/10.22038/ijbms.2024.76532.16576.

    Article  PubMed  PubMed Central  Google Scholar 

  56. Romanelli P, Bieler L, Scharler C, Pachler K, Kreutzer C, Zaunmair P, et al. Extracellular vesicles can deliver anti-inflammatory and anti-scarring activities of mesenchymal stromal cells after spinal cord injury. Front Neurol. 2019;10:1225. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fneur.2019.01225.

    Article  PubMed  PubMed Central  Google Scholar 

  57. Wang W, Yao F, Xing H, Yang F, Yan L. Exosomal miR-17-92 cluster from BMSCs alleviates apoptosis and inflammation in spinal cord injury. Biochem Genet. 2024. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s10528-024-10876-5.

    Article  PubMed  PubMed Central  Google Scholar 

  58. Shao Y, Wang Q, Liu L, Wang J, Wu M. Exosomes from microRNA 146a overexpressed bone marrow mesenchymal stem cells protect against spinal cord injury in rats. J Orthop Sci. 2023;28:1149–56. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.jos.2022.07.013.

    Article  PubMed  Google Scholar 

  59. Shao Y, Wang Q, Liu L, Wang J, Wu M. Alleviation of spinal cord injury by MicroRNA 137-overexpressing bone marrow mesenchymal stem cell-derived exosomes. Tohoku J Exp Med. 2023;259:237–46. https://doiorg.publicaciones.saludcastillayleon.es/10.1620/tjem.2022.J118.

    Article  CAS  PubMed  Google Scholar 

  60. Wang H, Zhao C, Rong Q, Cao J, Chen H, Li R, et al. The role of exosomes from mesenchymal stem cells in spinal cord injury: a systematic review. Int J Stem Cells. 2024;17:236–52. https://doiorg.publicaciones.saludcastillayleon.es/10.15283/ijsc23092.

    Article  PubMed  Google Scholar 

  61. Zhang C, Zhang C, Xu Y, Li C, Cao Y, Li P. Exosomes derived from human placenta-derived mesenchymal stem cells improve neurologic function by promoting angiogenesis after spinal cord injury. Neurosci Lett. 2020;739:135399. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.neulet.2020.135399.

    Article  CAS  PubMed  Google Scholar 

  62. Mu J, Li L, Wu J, Huang T, Zhang Y, Cao J, et al. Hypoxia-stimulated mesenchymal stem cell-derived exosomes loaded by adhesive hydrogel for effective angiogenic treatment of spinal cord injury. Biomater Sci. 2022;10:1803–11. https://doiorg.publicaciones.saludcastillayleon.es/10.1039/d1bm01722e.

    Article  CAS  PubMed  Google Scholar 

  63. Huang J, Xu Y, Yin X, Lin F. Exosomes derived from miR-126-modified MSCs promote angiogenesis and neurogenesis and attenuate apoptosis after spinal cord injury in rats. Neuroscience. 2020;424:133–45. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.neuroscience.2019.10.043.

    Article  CAS  PubMed  Google Scholar 

  64. Wu S, Chen Z, Wu Y, Shi Q, Yang E, Zhang B, et al. ADSC-Exos enhance functional recovery after spinal cord injury by inhibiting ferroptosis and promoting the survival and function of endothelial cells through the NRF2/SLC7A11/GPX4 pathway. Biomed Pharmacother. 2024;172:116225. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.biopha.2024.116225.

    Article  CAS  PubMed  Google Scholar 

  65. Lu Y, Zhou Y, Zhang R, Wen L, Wu K, Li Y, et al. Bone mesenchymal stem cell-derived extracellular vesicles promote recovery following spinal cord injury via improvement of the integrity of the blood-spinal cord barrier. Front Neurosci. 2019;13:209. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fnins.2019.00209.

    Article  PubMed  PubMed Central  Google Scholar 

  66. Xin W, Qiang S, Jianing D, Jiaming L, Fangqi L, Bin C, et al. Human bone marrow mesenchymal stem cell-derived exosomes attenuate blood-spinal cord barrier disruption via the TIMP2/MMP pathway after acute spinal cord injury. Mol Neurobiol. 2021;58:6490–504. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s12035-021-02565-w.

    Article  CAS  PubMed  Google Scholar 

  67. Surey S, Berry M, Logan A, Bicknell R, Ahmed Z. Differential cavitation, angiogenesis and wound-healing responses in injured mouse and rat spinal cords. Neuroscience. 2014;275:62–80. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.neuroscience.2014.06.003.

    Article  CAS  PubMed  Google Scholar 

  68. Zhou W, Silva M, Feng C, Zhao S, Liu L, Li S, et al. Exosomes derived from human placental mesenchymal stem cells enhanced the recovery of spinal cord injury by activating endogenous neurogenesis. Stem Cell Res Ther. 2021;12:174. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s13287-021-02248-2.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. 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. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s13287-021-02148-5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Wang Y, Han T, Guo R, Song P, Liu Y, Wu Z, et al. Micro-RNA let-7a-5p derived from mesenchymal stem cell-derived extracellular vesicles promotes the regrowth of neurons in Spinal-cord-injured rats by targeting the HMGA2/SMAD2 axis. Front Mol Neurosci. 2022;15:850364. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fnmol.2022.850364.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Li J, Jing Y, Bai F, Wu Y, Wang L, Yan Y, et al. Induced pluripotent stem cells as natural biofactories for exosomes carrying miR-199b-5p in the treatment of spinal cord injury. Front Pharmacol. 2022;13:1078761. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fphar.2022.1078761.

    Article  CAS  PubMed  Google Scholar 

  72. Abbas A, Huang X, Ullah A, Luo L, Xi W, Qiao Y, et al. Enhanced spinal cord repair using bioengineered induced pluripotent stem cell-derived exosomes loaded with miRNA. Mol Med (Cambridge, Mass). 2024;30:168. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s10020-024-00940-6.

    Article  CAS  Google Scholar 

  73. Liu J, Kong G, Lu C, Wang J, Li W, Lv Z, et al. IPSC-NSCs-derived exosomal let-7b-5p improves motor function after spinal cord Injury by modulating microglial/macrophage pyroptosis. J Nanobiotechnology. 2024;22:403. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12951-024-02697-w.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Li S, Luo L, He Y, Li R, Xiang Y, Xing Z, et al. Dental pulp stem cell-derived exosomes alleviate cerebral ischaemia-reperfusion injury through suppressing inflammatory response. Cell Prolif. 2021;54:e13093. https://doiorg.publicaciones.saludcastillayleon.es/10.1111/cpr.13093.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. ClinicalTrials.gov. https://clinicaltrials.gov/.

  76. International Clinical Trials Registry Platform (ICTRP). https://www.who.int/clinical-trials-registry-platform.

  77. Akhlaghpasand M, Tavanaei R, Hosseinpoor M, Yazdani K, Soleimani A, Zoshk 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. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s13287-024-03868-0.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Omrani M, Beyrampour-Basmenj H, Jahanban-Esfahlan R, Talebi M, Raeisi M, Serej ZA, et al. Global trend in exosome isolation and application: an update concept in management of diseases. Mol Cell Biochem. 2024;479:679–91. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s11010-023-04756-6.

    Article  CAS  PubMed  Google Scholar 

  79. Dilsiz N. A comprehensive review on recent advances in exosome isolation and characterization: toward clinical applications. Transl Oncol. 2024;50:102121. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.tranon.2024.102121.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. De Sousa KP, Rossi I, Abdullahi M, Ramirez MI, Stratton D, Inal JM. Isolation and characterization of extracellular vesicles and future directions in diagnosis and therapy. Wiley Interdiscip Rev Nanomed Nanobiotechnol. 2023;15:e1835. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/wnan.1835.

    Article  CAS  PubMed  Google Scholar 

  81. Wang C, Wang M, Xia K, Wang J, Cheng F, Shi K, 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;6:2523–34. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.bioactmat.2021.01.029.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  82. Tolomeo AM, Zuccolotto G, Malvicini R, De Lazzari G, Penna A, Franco C, et al. Biodistribution of intratracheal, intranasal, and intravenous injections of human mesenchymal stromal cell-derived extracellular vesicles in a mouse model for drug delivery studies. Pharmaceutics. 2023;15:548.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. Sun Y, Liu Q, Qin Y, Xu Y, Zhao J, Xie Y, et al. Exosomes derived from CD271(+)CD56(+) bone marrow mesenchymal stem cell subpopoulation identified by single-cell RNA sequencing promote axon regeneration after spinal cord injury. Theranostics. 2024;14:510–27. https://doiorg.publicaciones.saludcastillayleon.es/10.7150/thno.89008.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  84. Li L, Zhang Y, Mu J, Chen J, Zhang C, Cao H, et al. Transplantation of human mesenchymal stem-cell-derived exosomes immobilized in an adhesive hydrogel for effective treatment of spinal cord injury. Nano Lett. 2020;20:4298–305. https://doiorg.publicaciones.saludcastillayleon.es/10.1021/acs.nanolett.0c00929.

    Article  CAS  PubMed  Google Scholar 

  85. Han M, Yang H, Lu X, Li Y, Liu Z, Li F, et al. Three-dimensional-cultured MSC-derived exosome-hydrogel hybrid microneedle array patch for spinal cord repair. Nano Lett. 2022;22:6391–401. https://doiorg.publicaciones.saludcastillayleon.es/10.1021/acs.nanolett.2c02259.

    Article  CAS  PubMed  Google Scholar 

  86. Wang H, Tang Q, Lu Y, Chen C, Zhao Y-l, Xu T, et al. Berberine-loaded MSC-derived sEVs encapsulated in injectable GelMA hydrogel for spinal cord injury repair. Int J Pharm. 2023;643:123283. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.ijpharm.2023.123283.

    Article  CAS  PubMed  Google Scholar 

  87. Cheshmi H, Mohammadi H, Akbari M, Nasiry D, Rezapour-Nasrabad R, Bagheri M, et al. Human placental mesenchymal stem cell-derived exosomes in combination with hyperbaric oxygen synergistically promote recovery after spinal cord injury in rats. Neurotox Res. 2023;41:431–45. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s12640-023-00649-0.

    Article  CAS  PubMed  Google Scholar 

  88. Kalluri R, LeBleu V. The biology, function, and biomedical applications of exosomes. Science. 2020. https://doiorg.publicaciones.saludcastillayleon.es/10.1126/science.aau6977.

    Article  PubMed  PubMed Central  Google Scholar 

  89. Staufer O, Dietrich F, Rimal R, Schroter M, Fabritz S, Boehm H, et al. Bottom-up assembly of biomedical relevant fully synthetic extracellular vesicles. Sci Adv. 2021;7:6666. https://doiorg.publicaciones.saludcastillayleon.es/10.1126/sciadv.abg6666.

    Article  CAS  Google Scholar 

  90. Lu X, Xu G, Lin Z, Zou F, Liu S, Zhang Y, et al. Engineered exosomes enriched in netrin-1 modRNA promote axonal growth in spinal cord injury by attenuating inflammation and pyroptosis. Biomater Res. 2023;27:3. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s40824-023-00339-0.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  91. Wang B, Chang M, Zhang R, Wo J, Wu B, Zhang H, et al. Spinal cord injury target-immunotherapy with TNF-α autoregulated and feedback-controlled human umbilical cord mesenchymal stem cell derived exosomes remodelled by CRISPR/Cas9 plasmid. Biomater Adv. 2022;133:112624. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.msec.2021.112624.

    Article  CAS  PubMed  Google Scholar 

  92. Kang J, Guo Y. Human umbilical cord mesenchymal stem cells derived exosomes promote neurological function recovery in a rat spinal cord injury model. Neurochem Res. 2022;47:1532–40. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s11064-022-03545-9.

    Article  CAS  PubMed  Google Scholar 

  93. Zhou X, Chu X, Yuan H, Qiu J, Zhao C, Xin D, et al. Mesenchymal stem cell derived EVs mediate neuroprotection after spinal cord injury in rats via the microRNA-21-5p/FasL gene axis. Biomed Pharmacother. 2019;115:108818. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.biopha.2019.108818.

    Article  CAS  PubMed  Google Scholar 

  94. Xu G, Ao R, Zhi Z, Jia J, Yu B. miR-21 and miR-19b delivered by hMSC-derived EVs regulate the apoptosis and differentiation of neurons in patients with spinal cord injury. J Cell Physiol. 2019;234:10205–17. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/jcp.27690.

    Article  CAS  PubMed  Google Scholar 

  95. Kang J, Li Z, Zhi Z, Wang S, Xu G. MiR-21 derived from the exosomes of MSCs regulates the death and differentiation of neurons in patients with spinal cord injury. Gene Ther. 2019;26:491–503. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41434-019-0101-8.

    Article  CAS  PubMed  Google Scholar 

  96. Liang Y, Wu J, Zhu J, Yang H. Exosomes secreted by hypoxia-pre-conditioned adipose-derived mesenchymal stem cells reduce neuronal apoptosis in rats with spinal cord injury. J Neurotrauma. 2022;39:701–14. https://doiorg.publicaciones.saludcastillayleon.es/10.1089/neu.2021.0290.

    Article  PubMed  Google Scholar 

Download references

Acknowledgements

Miaoman Lin acknowledges the support of the China Scholarship Council for funding his academic pursuits. The authors declare that they have used AI assisted copy editing but no further AI-generated work in this manuscript

Funding

Open Access funding enabled and organized by Projekt DEAL. None.

Author information

Authors and Affiliations

Authors

Contributions

Alexander Younsi and Miaoman Lin conceived and designed the article. Miaoman Lin searched literature, drafted the manuscript, and drew the figures. Alexander Younsi, Thomas Skutella, Farzaneh Alimerzaloo, Xingjin Wang, Obada Alhalabi and Sandro Krieg gave constructive comments and revised the manuscript. All authors reviewed and approved the final manuscript.

Corresponding author

Correspondence to Alexander Younsi.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

All authors declared that they have no conflict of interest.

Additional information

Publisher's Note

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

Rights and permissions

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

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Lin, M., Alimerzaloo, F., Wang, X. et al. Harnessing stem cell-derived exosomes: a promising cell-free approach for spinal cord injury. Stem Cell Res Ther 16, 182 (2025). https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s13287-025-04296-4

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s13287-025-04296-4