Skip to main content
American Journal of Translational Research logoLink to American Journal of Translational Research
. 2023 Jul 15;15(7):4425–4438.

Mesenchymal stem cell-derived extracellular vesicles: emerging concepts in the treatment of spinal cord injury

Shujun Wang 1, Chengzhe Du 1, Guilan Li 2
PMCID: PMC10408507  PMID: 37560238

Abstract

Spinal cord injury (SCI) is a prevalent central nervous system disease with a high disability rate, leading to the loss of motor and sensory nerve function. Due to the complex pathophysiology of SCI, more effective clinical treatment strategies are needed. Research has indicated the considerable potential of extracellular vesicles (EVs) derived from mesenchymal stem cells (MSC-EVs) as a cell-free therapy in SCI repair and regeneration due to their ability to regulate immune cell activity and stimulate damaged neuron regeneration. Moreover, applying MSCs and engineered EVs can fully exploit the potential of MSC-EVs in spinal cord repair. Here, we outline the pathological process of SCI and its current clinical treatment status, summarize the latest MSC-EVs research and its pretreatment and engineering strategies in SCI treatment, and explore MSC-EVs application prospects.

Keywords: Spinal cord injury, mesenchymal stem cells, extracellular vesicles, engineering, treatment

Introduction

Spinal cord injury (SCI) is a severe central nervous system (CNS) disorder with high disability and mortality rates and is recognized as one of the greatest threats to human health [1,2]. SCI is estimated to permanently disable more than 27 million people worldwide, with approximately 77,000 new cases each year [1,3,4]. Current clinical treatments for SCI include pharmacotherapy, surgical decompression, hemodynamic therapy, and electrical stimulation. However, these methods do not completely slow SCI’s pathological progression [3-5]. Thus, an urgent need exists for a novel SCI treatment approach [6].

Transplantation of mesenchymal stem cells (MSCs) is a promising therapeutic approach for SCI treatment, but direct transplantation of stem cells brings safety and ethical concerns [10,11]. MSCs, or multipotent stromal cells, are a class of pluripotent stem cells belonging to the mesoderm, mainly found in connective tissue and organ mesenchyme [7]. MSCs have a strong proliferative capacity with multiple differentiation potentials and immunomodulatory functions and are also “seed cells” for tissue damage repair [8,9]. Notably, growing evidence suggests that MSCs’ therapeutic properties are more likely to derive from paracrine effects, particularly from extracellular vesicles (EVs) [12,13]. EVs are essential mediators of intercellular communication and are involved in many pathological processes [14,15]. The therapeutic potential of MSCs derived from EVs (MSC-EVs) in SCI has received increasing attention in recent years [16-18].

Here, we review the pathophysiology and current status of SCI treatment. Then, we describe EV biogenesis and summarize the roles and mechanisms of MSC-EVs in SCI repair. Furthermore, we review the application and challenges of pretreatment methods and engineering strategies being used to improve the therapeutic potential of MSC-EVs in SCI.

The pathophysiology of SCI

Most spinal cord injuries are caused by contusion or impingement [6]. Changes in spinal canal shape or volume lead to physical deformation of spinal cord tissue, compressing blood vessels and axons and triggering a cascade of pathological processes [1,19,20]. In the acute phase of SCI (typically within hours to days after injury), bleeding from ruptured blood vessels causes tissue ischemia and localized spinal cord swelling [21,22]. Damaged cells release adenosine triphosphate (ATP) that act on purinergic receptors of various immune cells, inducing the immune cells to converge on the damaged area [23,24]. Microglia (innate immune cells of the CNS) exert phagocytosis in the injured tissue to prevent injury spread. However, microglia also mediate excessive immune responses. Subsequently, tissue reperfusion-induced oxidative stress and glutamate release lead to the death of neighboring neurons and glial cells [25,26].

Furthermore, a dramatic imbalance in the ionic homeostasis of the tissue microenvironment activates enzymes such as lipid oxidase and lipid accumulation, mediating the occurrence of cellular lipid peroxidation and expanding areas of tissue necrosis [19,22,26].

Later in the acute phase of SCI, the hemostatic response also brings about a series of cascade reactions to stop hemorrhage from broken vessels while also bringing about a potent inflammatory response stimulus (e.g., chemokines and eicosanoids released by platelets) to glial cells [27,28]. The subacute phase occurs days to weeks after injury, where the incomplete pathological tissue and inflammatory microenvironment activate a variety of cells (including astrocytes, fibroblasts, oligodendrocytes, pericytes, and macrophages), triggering a cascade of secondary damage that further exacerbates injury and inflammation [21,29-32]. Later in the subacute phase, cross-linked interactions between reactive glial cells and non-neural cells form scar tissue [33,34]. Simultaneously, extracellular matrix (ECM) molecules, such as chondroitin sulfate proteoglycans (CSPGs), block the outgrowth and regeneration of damaged axons [20,35,36].

SCI’s chronic phase exhibits a gradual decrease in the local acute inflammatory response and a diminishing of glial cell proliferation. Additionally, the spinal cord tissue at the injury site may become atrophied, softened, cystic, or even spinal cavernous [21,30,36]. At the periphery of the damaged area, distinct scar tissue or spinal cord compression forms by glial cells, fibroblasts, and ECM molecules. While limiting inflammation, scar tissue can prevent axonal lengthening [31,35]. Lack of axonal lengthening results in long-term loss of spinal cord function and affects the physiological function of the trunk and extremities below the site of spinal cord damage [29,37-39].

Current status of SCI treatment

According to the Clinical Neurorestorative Therapeutic Guidelines for Spinal Cord Injury (version 2019) proposed by the International Association of Neurorestoratology and the Chinese Association of Neurorestoratology, the primary strategies for SCI neurorepair are pharmacotherapy, surgical intervention, and electrical stimulation therapy [3]:

(a) Pharmacotherapy: Early high-dose methylprednisolone (MP) therapy was once considered to benefit neurorepair in SCI’s acute phase. However, various clinical trials have shown that MP therapy may have serious side effects, such as pneumonia and wound infections [40,41]. Accordingly, high-dose MP therapy is no longer recommended for routine use for acute SCI [4,42]. Pharmacological treatments, such as gangliosides and acidic fibroblast growth factor, may benefit SCI, but additional clinical data are needed to confirm their use [43,44].

(b) Surgical intervention: Spinal cord decompression and internal fixation surgery can be immediately performed post-injury, within an appropriate window, to reduce secondary injury and prevent further destruction of spinal cord tissue. However, many patients are usually unable to undergo surgery promptly due to transportation, preoperative examination, and preparation [5,45].

(c) Electrical stimulation therapy: In SCI’s chronic phase, local neuromuscular or peripheral nerve electrical stimulation may improve and induce axonal regeneration. However, this therapy is still being further explored for therapeutic effects [46-48].

The pathological changes of SCI are incredibly complex. Clinical treatment focuses on alleviating the imbalance of the microenvironment caused by secondary injury in the acute and/or subacute phase and protecting the surviving neurons. However, the current treatment options are ineffective in changing the underlying pathology of SCI [49]. Therefore, searching for novel, more effective, and safe therapies is an urgent clinical need.

Overview of MSC-EVs

With strong immunomodulatory and tissue regenerative potential, MSCs effectively treat various refractory diseases, including SCI [50,51]. MSCs influence tissue damage repair primarily through paracrine effects. One of the most essential paracrine effectors of MSCs is EVs. EVs are nanoscale-sized vesicles composed of phospholipid bilayer membranes that deliver bioactive components [7,12,16].

Depending on their size and biogenesis, EVs can be classified into three major categories: apoptotic bodies [52], microvesicles, and small EVs, known as exosomes [53]. Apoptotic bodies are the largest subpopulation of EVs and are released by cells undergoing programmed cell death. Microvesicles are 200-1000 nm in diameter and arise from the plasma membrane surface by means of outgrowth [54]. Exosomes are produced by the endocytic pathway and have an average diameter of 30-200 nm.

Exosome formation is a complex process involving the formation of endosomes and intracellular multivessel bodies (MVBs) and ends in their release [55]. First, the cytoplasmic membrane forms early endosomes via the endocytic pathway. Early endosomes subsequently fuse and exchange with other organelle material to form late endosomes. Late endosomes further develop into MVBs [56]. During this process, intraluminal vesicles form and accumulate in MVBs with the help of Golgi complexes. Finally, intraluminal vesicles form exosomes after fusing with the plasma membrane; exosomes are then released into the extracellular environment [57]. When exosomes are released, they can be internalized into recipient cells by three potential mechanisms: endocytosis, direct fusion (with plasma membrane), or receptor-ligand interactions [53,58]. Exosomes may contain many components depending on the parental cells’ specific physiological or pathological state, including DNA, RNA, lipids, proteins, and metabolites [59,60]. Thus, exosomes reflect the parental cells’ metabolic state and function. Exosomes then reach recipient cells and exchange molecular information [15] (Figure 1).

Figure 1.

Figure 1

The biogenesis and uptake of exosomes. Exosomes are generated via the endocytic pathway and are released into the extracellular space. Cellular contents such as proteins, lipids, metabolites, small molecules, DNA, RNA, and cell surface proteins can be internalized into extracellular vesicles (EVs) via endocytosis and membrane invagination. The EVs released by donor cells can interact with recipient cells and induce biological responses within the recipient cells. The process occurs via an interaction with cell surface proteins or receptors or through internalization via endocytosis or membrane fusion.

Many studies have found that MSC-EVs can repair a wide range of tissue damage and may be a novel “non-cellular” therapeutic strategy superior to cell therapy [12,61]. Additionally, the expression profiles of MSC-EV components from multi-omics applications of active RNA, protein, and lipid components have been gradually revealed. We now know that MSC-EVs play essential therapeutic roles in diabetes, tissue recovery, immunomodulation, and neuroprotection by regulating gene expression of receptor cells [62-66]. Mechanically, enriched proteins of MSC-EVs play irreplaceable roles in tissue repair and regeneration, including in Wnt4 [67], 14-3-3ζ [68,69], glutathione peroxidase 1 [70], casein kinase 1δ (CK1δ), β-transducin repeats-containing proteins [71], E3 ubiquitin-protein ligase NEDD4 [72] and Beclin-1 [73], among others. In summary, MSC-EVs have promising applications in various refractory diseases and tissue damage repair. The following section will focus on the latest advances in MSC-EVs in treating SCI.

Therapeutic role of MSC-EVs in SCI

Natural MSC-EVs in SCI

The use of MSC-EVs in SCI repair has been gaining increased attention, both independently and in conjunction with bioactive molecules [74]. Typically, these treatments focus on three key areas: (1) the inhibition of inflammation in the microenvironment of the spinal cord to decrease barriers of axonal germination [75], (2) the activation of the axonal regeneration potential of impaired neurons or endogenous neural stem cells to reconstruct damaged neural circuits and facilitate functional recovery [76], and (3) the use of MSC-EVs with biomaterials to supply both graft and nutritional support for guided axonal regeneration [77] (Figure 2).

Figure 2.

Figure 2

Natural mesenchymal stem cell-derived extracellular vesicles (MSC-EVs) in spinal cord injury (SCI) repair. The utilization of MSC-EVs in SCI restoration is achieved through the transportation of active molecules like miRNA, which suppresses the detrimental microenvironment, activates the regenerative potential of surviving neurons, and guides axonal regeneration to alleviate SCI.

Inhibition of the damaged microenvironment

The first step in regenerating damaged CNS neurons is early growth cone generation that guides axonal regeneration [78]. However, damaged neurons form structures called retraction bulbs, and axonal outgrowth ceases due to the poor microenvironment of the damaged spinal cord [34]. The spinal cord microenvironment comprises glial cells, immune cells, pericytes, endothelial cells, extracellular matrix, and neurotransmitters [39].

It has been well established that MSC-EVs can modulate various cell types of the spinal cord microenvironment to improve microenvironments that otherwise are not conducive to neural regeneration [79]. Zhao et al. found that MSC-EVs suppressed inflammation in the spinal cord microenvironment by blocking the activation of the nuclear factor-κB signaling pathway and the microglia complement system. Microglia are the first immune cells to activate and mediate inflammation after SCI [80]. Also, various miRNAs of MSC-EVs origin (e.g., miRNA-21-5p, miRNA-182, and let-7b) have been found to promote macrophage polarization toward the anti-inflammatory phenotype M2 alleviating SCI by targeting and inhibiting toll-like receptor signaling pathway [81-83]. Macrophages in the peripheral circulation are recruited in response to vascular rupture and inflammation caused by spinal cord damage. They further exacerbate the inflammatory response of the spinal cord microenvironment and are detrimental to axonal regeneration [81]. Lastly, MSC-EVs were found to protect pericytes, critical components of the neurovascular unit, thus promoting neuronal survival and nerve fiber extension by inhibiting cell scorching and improving blood-spinal cord barrier integrity; MSC-EVs ultimately improved motor function in spinal cord injured rats [84,85].

Activation of axonal regeneration potential

Multiple transcriptional programs are repressed in most highly differentiated neurons in mammals, thus limiting their regenerative potential [86]. The mammalian target of the rapamycin (mTOR) signaling pathway activates neonatal CNS neurons to maintain cell growth, metabolism, and protein synthesis. However, in adult CNS neurons, enhanced expression of phosphatase and tensin homolog deleted on chromosome ten (PTEN) activity, a negative regulator of mTOR, considerably reduces mTOR signaling levels, leading to diminished neural regeneration [17].

MSC-EVs have been demonstrated to inhibit PTEN by delivering miRNA-21 and miRNA-19b promoting axonal growth and neuronal survival [86,87]. Conversely, activation of endogenous neural stem cells in the spinal cord to differentiate toward mature neurons can reconnect parts of the neural circuits and restore neural function. Additionally, MSC-EVs have been suggested to promote the activation of proliferating endogenous neural stem cells by activating the ERK pathway, as evidenced by a considerable increase in activation of spinal SOX2 + GFAP + and SOX1 + KI67 + cells, among others [88-90].

Combining biomaterials to guide axonal extension

MSC-EVs can also be combined with biomaterials such as hydrogels for SCI repair applications [91]. Gel biomaterials can guide axonal extension toward the lesion region to slow-release carriers and enhance the utilization efficiency of MSC-EVs [92]. For example, Wang et al. encapsulated MSC-EVs in an injectable (FE@EVs) for in situ drug delivery in SCI rats. FE@EVs achieved up to 56 days of EV release in vivo and considerably improved motor function recovery in rats after SCI [77]. Additionally, to mimic the conductive properties of neural tissue, Fan et al. developed a conductive hydrogel composed of gelatin methacrylate and polypyrrole loaded with MSC-derived exosomes (GMPE). GMPE induced the differentiation in a mouse spinal cord hemisection model of neural stem cells to mature neurons in vitro and showed considerable neuroregenerative benefits after implantation [93]. Accordingly, the combined application of MSC-EVs and biomaterials may serve as an in vivo treatment for SCI (Figure 3).

Figure 3.

Figure 3

Pretreating and engineering strategies to enhance the therapeutic potential of mesenchymal stem cell-derived extracellular vesicles (MSC-EVs) for spinal cord injury (SCI) repair. Hypoxia and melatonin pretreatment alter the contents of MSC-EVs and thus improve their SCI efficacy. cystine-alanine-glutamine-lysine short peptide (CAQK) and extracellular-5-nucleotidase (CD73) engineering strategies can improve the expected efficacy of MSC-EVs.

Pretreating and engineering strategies to enhance the therapeutic potential of MSC-EVs for SCI

Despite the tremendous therapeutic potential of natural MSC-EVs for SCI, many inherent limitations still limit their large-scale application in SCI therapy. For example, the decreased stemness and replicative senescence of MSCs under conventional culture conditions lead to a decrease in the number and therapeutic efficacy of their secreted EVs [94,95]. Additionally, due to the inherent properties of natural EVs, MSC-EVs are poorly targeted to SCI injury sites in vivo after intravenous administration [96,97]. Therefore, improving the production of MSC-EVs and enhancing their targeting ability is essential for their clinical application [98].

Pretreating methods

The therapeutic potential of MSC-EVs is closely related to their cellular status. Changing culture conditions or drug pretreatment are effective strategies to improve MSC-EV yield and therapeutic efficacy [99]. For example, hypoxic culture was found to mimic the “stem cell niche” environment of MSCs in vivo, maintaining the stemness of MSCs and enhancing their proliferation capacity [100]. Also, MSC-EVs from a hypoxic culture delivered microRNA molecules involved in neuroregeneration and inflammation regulation (e.g., miRNA-511-3p and miRNA-499a-5p) and effectively promoted SCI repair [101,102]. Liu et al. found that melatonin (MT) pretreatment (an amine-like hormone secreted by the brain’s pineal gland with free radical scavenging and antioxidant effects [103]) decreased the expression of methyltransferase 3 in MSCs, inhibited m6A methylation, and maintained the stability of ubiquitin-specific protease 29 (USP29) mRNA in the cells. They also found that MT-pretreated MSC-EVs stabilized nuclear-like factors by delivering USP29, which regulated microglia/macrophage polarization and promoted recovery of motor behavior in SCI mice [103]. These studies demonstrate the benefits of diverse pretreatment methods for MSCs and MSC-EVs. Designing novel combinations of MSC pretreatment methods for different pathological stages or degrees of injury in SCI may provide more effective treatment strategies for SCI in the future [98].

Engineering strategy

MSC-EVs have also been modified by engineering means such as content piggybacking or membrane modification to enhance spinal cord targeting and SCI therapeutic efficacy [104,105]. Recently, it has been found that cystine-alanine-glutamine-lysine short peptide (CAQK) can specifically target CSPGs at SCI-damaged sites [106]. Accordingly, Wang et al. constructed CAQK peptide-modified MSC-derived exosomes (EXO-C@P) loaded with a TNF-α-responsive self-feedback CRISPR/Cas9 system. The team successfully delivered EXO-C@P by intravenous injection to the SCI-impaired site. EXO-C@P was phagocytosed at the site by activated immune cells (e.g., macrophages and neutrophils) and carried out cellular-level gene editing, resulting in the secretion of soluble mTNFR1 to neutralize TNF-α and reduce inflammation; the system facilitated recovery from SCI [106].

Additionally, as extracellular-5-nucleotidase (CD73) can catabolize excess ATP to adenosine after SCI and alleviate the damaged microenvironment, Zhai et al. engineered MSC-EVs overexpressing CD73 (CD73+ MSC-EVs). CD73+ MSC-EVs improved SCI by reducing extracellular ATP in spinal cord tissue and activating the A2bR/cAMP/PKA pathway [107]. As more therapeutic molecules for spinal cord and neuronal cells are uncovered, engineering technologies will load multiple combinations of molecules into MSC-EVs. The combined application of membrane modification and content piggybacking technologies is expected to address MSC-EV neural targeting while improving the enrichment of active molecules [108-110]. Thus, pretreatment and engineering strategies are expected to be the propellers of MSC-EVs in SCI treatment.

Conclusions and outlook

Due to the weak regenerative capacity of the CNS in adult mammals, recovery of neurological function after SCI is rather limited [1,5]. The pathological process of SCI is also complex and lengthy, with the recovery of spinal cord tissue subject to a combination of factors. Despite the availability of therapeutic tools to reduce mortality in SCI patients, functional recovery after SCI remains a great challenge for current medicine [4,28]. A series of experiments have demonstrated that MSC-EVs can easily cross the blood-spinal cord barrier, improve the poor injury microenvironment of the spinal cord, activate the regenerative potential of damaged neurons, and combine with biomaterials to guide axonal extension at the injured site [18,87,111]. These conclusions suggest that MSC-EVs are an expected novel, non-cellular therapy for SCI treatment. Additionally, pretreatment methods like hypoxia and engineering strategies like membrane modification could considerably optimize MSC-EV therapeutic potential in SCI [98] (Table 1).

Table 1.

Therapeutic role of MSC-EVs in SCI

Origin of EVs Target cells Cargos in EVs Mechanism Ref.
Natural Mesenchymal stromal cells-derived extracellular vesicles (MSC-EVs) Microglia / Inhibite activation of the microglia complement system and NF-κB signaling pathway [80]
Macrophages miRNA-21-5p, miRNA-182, or let-7b Promote macrophage polarization toward M2 (anti-inflammatory phenotype) [81-83]
Pericytes / Inhibite cell scorching and improving blood-spinal cord barrier integrity [84,85]
Neurons miRNA-21 or miRNA-19b Promote axonal growth and neuronal survival [86,87]
Neural stem cells / Promote the activation of proliferating endogenous neural stem cells by activating the ERK pathway [88-90]
F127-polycitrate-polyethyleneimine hydrogel (FE@EVs) Neurons / Improve motor function recovery in rats after SCI [77]
Gelatin methacrylate (GM) and polypyrrole (PPy) loaded with MSC-derived exosomes (GMPE) Neurons / Induce the differentiation of neural stem cells [93]
MSC-EVs from a hypoxic culture Neurons miRNA-511-3p or miRNA-499a-5p Improve neuroregeneration and inflammation regulation [101,102]
Melatonin (MT) pretreated MSC-EVs Microglia/macrophage / Regulate microglia/macrophage polarization and promoted recovery of motor behavior in SCI mice [98]
Cystine-alanine-glutamine-lysine short peptide (CAQK) modified MSC-derived exosomes (EXO-C@P) Macrophages and neutrophils TNF-α self-feedback CRISPR/Cas9 system Carry out cellular-level gene editing, resulting in the secretion of soluble mTNFR1 to neutralize TNF-α and reduce inflammation [106]
Engineered MSC-EVs overexpressing CD73 (CD73+ MSC-EVs) Microglia CD73 Reduce extracellular ATP in spinal cord tissue and activating the A2bR/cAMP/PKA pathway [107]

Certainly, MSC-EVs or modified MSC-EVs in SCI treatment need more study to elucidate their mechanisms [13,112]. Also, further clinical trials are required to validate the efficacy and safety of MSC-EVs in humans [111,113]. Likewise, establishing a set of standardized processes for the isolation and purification of MSC-EVs and the concentration and mode of administration are the keys to upscaling applications [114,115]. Future studies that address these issues will provide a comprehensive theoretical basis for the clinical translation of MSC-EVs in SCI treatment, providing direction and hope for SCI clinical treatment [116,117].

Disclosure of conflict of interest

None.

References

  • 1.McDonald JW, Sadowsky C. Spinal-cord injury. Lancet. 2002;359:417–425. doi: 10.1016/S0140-6736(02)07603-1. [DOI] [PubMed] [Google Scholar]
  • 2.Lee BB, Cripps RA, Fitzharris M, Wing PC. The global map for traumatic spinal cord injury epidemiology: update 2011, global incidence rate. Spinal Cord. 2014;52:110–116. doi: 10.1038/sc.2012.158. [DOI] [PubMed] [Google Scholar]
  • 3.Huang H, Young W, Skaper S, Chen L, Moviglia G, Saberi H, Al-Zoubi Z, Sharma HS, Muresanu D, Sharma A, El Masry W, Feng S International Association of Neurorestoratology and The Chinese Association of Neurorestoratology. Clinical neurorestorative therapeutic guidelines for spinal cord injury (IANR/CANR version 2019) J Orthop Translat. 2019;20:14–24. doi: 10.1016/j.jot.2019.10.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Karsy M, Hawryluk G. Modern medical management of spinal cord injury. Curr Neurol Neurosci Rep. 2019;19:65. doi: 10.1007/s11910-019-0984-1. [DOI] [PubMed] [Google Scholar]
  • 5.O’Toole JE, Kaiser MG, Anderson PA, Arnold PM, Chi JH, Dailey AT, Dhall SS, Eichholz KM, Harrop JS, Hoh DJ, Qureshi S, Rabb CH, Raksin PB. Congress of neurological surgeons systematic review and evidence-based guidelines on the evaluation and treatment of patients with thoracolumbar spine trauma: executive summary. Neurosurgery. 2019;84:2–6. doi: 10.1093/neuros/nyy394. [DOI] [PubMed] [Google Scholar]
  • 6.Ahuja CS, Nori S, Tetreault L, Wilson J, Kwon B, Harrop J, Choi D, Fehlings MG. Traumatic spinal cord injury-repair and regeneration. Neurosurgery. 2017;80:S9–S22. doi: 10.1093/neuros/nyw080. [DOI] [PubMed] [Google Scholar]
  • 7.Galipeau J, Sensébé L. Mesenchymal stromal cells: clinical challenges and therapeutic opportunities. Cell Stem Cell. 2018;22:824–833. doi: 10.1016/j.stem.2018.05.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Uccelli A, Moretta L, Pistoia V. Mesenchymal stem cells in health and disease. Nat Rev Immunol. 2008;8:726–736. doi: 10.1038/nri2395. [DOI] [PubMed] [Google Scholar]
  • 9.Uder C, Brückner S, Winkler S, Tautenhahn HM, Christ B. Mammalian MSC from selected species: features and applications. Cytometry A. 2018;93:32–49. doi: 10.1002/cyto.a.23239. [DOI] [PubMed] [Google Scholar]
  • 10.Coppin L, Sokal E, Stéphenne X. Thrombogenic risk induced by intravascular mesenchymal stem cell therapy: current status and future perspectives. Cells. 2019;8:1160. doi: 10.3390/cells8101160. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Tan TT, Toh WS, Lai RC, Lim SK. Practical considerations in transforming MSC therapy for neurological diseases from cell to EV. Exp Neurol. 2022;349:113953. doi: 10.1016/j.expneurol.2021.113953. [DOI] [PubMed] [Google Scholar]
  • 12.Witwer KW, Van Balkom BWM, Bruno S, Choo A, Dominici M, Gimona M, Hill AF, De Kleijn D, Koh M, Lai RC, Mitsialis SA, Ortiz LA, Rohde E, Asada T, Toh WS, Weiss DJ, Zheng L, Giebel B, Lim SK. Defining mesenchymal stromal cell (MSC)-derived small extracellular vesicles for therapeutic applications. J Extracell Vesicles. 2019;8:1609206. doi: 10.1080/20013078.2019.1609206. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Rohde E, Pachler K, Gimona M. Manufacturing and characterization of extracellular vesicles from umbilical cord-derived mesenchymal stromal cells for clinical testing. Cytotherapy. 2019;21:581–592. doi: 10.1016/j.jcyt.2018.12.006. [DOI] [PubMed] [Google Scholar]
  • 14.Cocucci E, Meldolesi J. Ectosomes and exosomes: shedding the confusion between extracellular vesicles. Trends Cell Biol. 2015;25:364–372. doi: 10.1016/j.tcb.2015.01.004. [DOI] [PubMed] [Google Scholar]
  • 15.Tkach M, Théry C. Communication by extracellular vesicles: where we are and where we need to go. Cell. 2016;164:1226–1232. doi: 10.1016/j.cell.2016.01.043. [DOI] [PubMed] [Google Scholar]
  • 16.Cheng L, Hill AF. Therapeutically harnessing extracellular vesicles. Nat Rev Drug Discov. 2022;21:379–399. doi: 10.1038/s41573-022-00410-w. [DOI] [PubMed] [Google Scholar]
  • 17.Xiao X, Li W, Xu Z, Sun Z, Ye H, Wu Y, Zhang Y, Xie L, Jiang D, Jia R, Wang X. Extracellular vesicles from human umbilical cord mesenchymal stem cells reduce lipopolysaccharide-induced spinal cord injury neuronal apoptosis by mediating miR-29b-3p/PTEN. Connect Tissue Res. 2022;63:634–649. doi: 10.1080/03008207.2022.2060826. [DOI] [PubMed] [Google Scholar]
  • 18.Jia Y, Yang J, Lu T, Pu X, Chen Q, Ji L, Luo C. Repair of spinal cord injury in rats via exosomes from bone mesenchymal stem cells requires sonic hedgehog. Regen Ther. 2021;18:309–315. doi: 10.1016/j.reth.2021.08.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Badhiwala JH, Ahuja CS, Fehlings MG. Time is spine: a review of translational advances in spinal cord injury. J Neurosurg Spine. 2018;30:1–18. doi: 10.3171/2018.9.SPINE18682. [DOI] [PubMed] [Google Scholar]
  • 20.Bradbury EJ, Burnside ER. Moving beyond the glial scar for spinal cord repair. Nat Commun. 2019;10:3879. doi: 10.1038/s41467-019-11707-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Dias DO, Kalkitsas J, Kelahmetoglu Y, Estrada CP, Tatarishvili J, Holl D, Jansson L, Banitalebi S, Amiry-Moghaddam M, Ernst A, Huttner HB, Kokaia Z, Lindvall O, Brundin L, Frisén J, Göritz C. Pericyte-derived fibrotic scarring is conserved across diverse central nervous system lesions. Nat Commun. 2021;12:5501. doi: 10.1038/s41467-021-25585-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Munteanu C, Rotariu M, Turnea M, Ionescu AM, Popescu C, Spinu A, Ionescu EV, Oprea C, Tucmeanu RE, Tătăranu LG, Silisteanu SC, Onose G. Main cations and cellular biology of traumatic spinal cord injury. Cells. 2022;11:2503. doi: 10.3390/cells11162503. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.O’Shea TM, Burda JE, Sofroniew MV. Cell biology of spinal cord injury and repair. J Clin Invest. 2017;127:3259–3270. doi: 10.1172/JCI90608. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Pineau I, Lacroix S. Proinflammatory cytokine synthesis in the injured mouse spinal cord: multiphasic expression pattern and identification of the cell types involved. J Comp Neurol. 2007;500:267–285. doi: 10.1002/cne.21149. [DOI] [PubMed] [Google Scholar]
  • 25.Bellver-Landete V, Bretheau F, Mailhot B, Vallières N, Lessard M, Janelle ME, Vernoux N, Tremblay ME, Fuehrmann T, Shoichet MS, Lacroix S. Microglia are an essential component of the neuroprotective scar that forms after spinal cord injury. Nat Commun. 2019;10:518. doi: 10.1038/s41467-019-08446-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Haynes SE, Hollopeter G, Yang G, Kurpius D, Dailey ME, Gan WB, Julius D. The P2Y12 receptor regulates microglial activation by extracellular nucleotides. Nat Neurosci. 2006;9:1512–1519. doi: 10.1038/nn1805. [DOI] [PubMed] [Google Scholar]
  • 27.Goodman JH, Bingham WG Jr, Hunt WE. Platelet aggregation in experimental spinal cord injury. Ultrastructural observations. Arch Neurol. 1979;36:197–201. doi: 10.1001/archneur.1979.00500400051006. [DOI] [PubMed] [Google Scholar]
  • 28.Tator CH. Update on the pathophysiology and pathology of acute spinal cord injury. Brain Pathol. 1995;5:407–413. doi: 10.1111/j.1750-3639.1995.tb00619.x. [DOI] [PubMed] [Google Scholar]
  • 29.Hara M, Kobayakawa K, Ohkawa Y, Kumamaru H, Yokota K, Saito T, Kijima K, Yoshizaki S, Harimaya K, Nakashima Y, Okada S. Interaction of reactive astrocytes with type I collagen induces astrocytic scar formation through the integrin-N-cadherin pathway after spinal cord injury. Nat Med. 2017;23:818–828. doi: 10.1038/nm.4354. [DOI] [PubMed] [Google Scholar]
  • 30.Dias DO, Göritz C. Fibrotic scarring following lesions to the central nervous system. Matrix Biol. 2018;68-69:561–570. doi: 10.1016/j.matbio.2018.02.009. [DOI] [PubMed] [Google Scholar]
  • 31.Francos-Quijorna I, Sánchez-Petidier M, Burnside ER, Badea SR, Torres-Espin A, Marshall L, de Winter F, Verhaagen J, Moreno-Manzano V, Bradbury EJ. Chondroitin sulfate proteoglycans prevent immune cell phenotypic conversion and inflammation resolution via TLR4 in rodent models of spinal cord injury. Nat Commun. 2022;13:2933. doi: 10.1038/s41467-022-30467-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Van Broeckhoven J, Sommer D, Dooley D, Hendrix S, Franssen AJPM. Macrophage phagocytosis after spinal cord injury: when friends become foes. Brain. 2021;144:2933–2945. doi: 10.1093/brain/awab250. [DOI] [PubMed] [Google Scholar]
  • 33.Okada M, Miyamoto O, Shibuya S, Zhang X, Yamamoto T, Itano T. Expression and role of type I collagen in a rat spinal cord contusion injury model. Neurosci Res. 2007;58:371–377. doi: 10.1016/j.neures.2007.04.009. [DOI] [PubMed] [Google Scholar]
  • 34.Sofroniew MV. Dissecting spinal cord regeneration. Nature. 2018;557:343–350. doi: 10.1038/s41586-018-0068-4. [DOI] [PubMed] [Google Scholar]
  • 35.Gaudet AD, Popovich PG. Extracellular matrix regulation of inflammation in the healthy and injured spinal cord. Exp Neurol. 2014;258:24–34. doi: 10.1016/j.expneurol.2013.11.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Didangelos A, Puglia M, Iberl M, Sanchez-Bellot C, Roschitzki B, Bradbury EJ. High-throughput proteomics reveal alarmins as amplifiers of tissue pathology and inflammation after spinal cord injury. Sci Rep. 2016;6:21607. doi: 10.1038/srep21607. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Göritz C, Dias DO, Tomilin N, Barbacid M, Shupliakov O, Frisén J. A pericyte origin of spinal cord scar tissue. Science. 2011;333:238–242. doi: 10.1126/science.1203165. [DOI] [PubMed] [Google Scholar]
  • 38.Hutson TH, Di Giovanni S. The translational landscape in spinal cord injury: focus on neuroplasticity and regeneration. Nat Rev Neurol. 2019;15:732–745. doi: 10.1038/s41582-019-0280-3. [DOI] [PubMed] [Google Scholar]
  • 39.Tran AP, Warren PM, Silver J. The biology of regeneration failure and success after spinal cord injury. Physiol Rev. 2018;98:881–917. doi: 10.1152/physrev.00017.2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Liu Z, Yang Y, He L, Pang M, Luo C, Liu B, Rong L. High-dose methylprednisolone for acute traumatic spinal cord injury: a meta-analysis. Neurology. 2019;93:e841–e850. doi: 10.1212/WNL.0000000000007998. [DOI] [PubMed] [Google Scholar]
  • 41.Canseco JA, Karamian BA, Bowles DR, Markowitz MP, DiMaria SL, Semenza NC, Leibensperger MR, Smith ML, Vaccaro AR. Updated review: the steroid controversy for management of spinal cord injury. World Neurosurg. 2021;150:1–8. doi: 10.1016/j.wneu.2021.02.116. [DOI] [PubMed] [Google Scholar]
  • 42.Lee BJ, Jeong JH. Review: steroid use in patients with acute spinal cord injury and guideline update. Korean J Neurotrauma. 2022;18:22–30. doi: 10.13004/kjnt.2022.18.e21. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Torelli AG, Cristante AF, de Barros-Filho TEP, Dos Santos GB, Morena BC, Correia FF, Paschon V. Effects of ganglioside GM1 and erythropoietin on spinal cord injury in mice: functional and immunohistochemical assessments. Clinics (Sao Paulo) 2022;77:100006. doi: 10.1016/j.clinsp.2022.100006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Chen X, Jin X, Huang F, Wang J, Cao X, Wang PG, Feng Y, Jiang F, Yang G. Design, synthesis and neurite outgrowth activity of novel ganglioside GM1 derivatives by remodeling of the fatty acid moiety. Eur J Med Chem. 2022;241:114636. doi: 10.1016/j.ejmech.2022.114636. [DOI] [PubMed] [Google Scholar]
  • 45.Rowald A, Komi S, Demesmaeker R, Baaklini E, Hernandez-Charpak SD, Paoles E, Montanaro H, Cassara A, Becce F, Lloyd B, Newton T, Ravier J, Kinany N, D’Ercole M, Paley A, Hankov N, Varescon C, McCracken L, Vat M, Caban M, Watrin A, Jacquet C, Bole-Feysot L, Harte C, Lorach H, Galvez A, Tschopp M, Herrmann N, Wacker M, Geernaert L, Fodor I, Radevich V, Van Den Keybus K, Eberle G, Pralong E, Roulet M, Ledoux JB, Fornari E, Mandija S, Mattera L, Martuzzi R, Nazarian B, Benkler S, Callegari S, Greiner N, Fuhrer B, Froeling M, Buse N, Denison T, Buschman R, Wende C, Ganty D, Bakker J, Delattre V, Lambert H, Minassian K, van den Berg CAT, Kavounoudias A, Micera S, Van De Ville D, Barraud Q, Kurt E, Kuster N, Neufeld E, Capogrosso M, Asboth L, Wagner FB, Bloch J, Courtine G. Activity-dependent spinal cord neuromodulation rapidly restores trunk and leg motor functions after complete paralysis. Nat Med. 2022;28:260–271. doi: 10.1038/s41591-021-01663-5. [DOI] [PubMed] [Google Scholar]
  • 46.Wagner FB, Mignardot JB, Le Goff-Mignardot CG, Demesmaeker R, Komi S, Capogrosso M, Rowald A, Seáñez I, Caban M, Pirondini E, Vat M, McCracken LA, Heimgartner R, Fodor I, Watrin A, Seguin P, Paoles E, Van Den Keybus K, Eberle G, Schurch B, Pralong E, Becce F, Prior J, Buse N, Buschman R, Neufeld E, Kuster N, Carda S, von Zitzewitz J, Delattre V, Denison T, Lambert H, Minassian K, Bloch J, Courtine G. Targeted neurotechnology restores walking in humans with spinal cord injury. Nature. 2018;563:65–71. doi: 10.1038/s41586-018-0649-2. [DOI] [PubMed] [Google Scholar]
  • 47.Courtine G, Sofroniew MV. Spinal cord repair: advances in biology and technology. Nat Med. 2019;25:898–908. doi: 10.1038/s41591-019-0475-6. [DOI] [PubMed] [Google Scholar]
  • 48.Kathe C, Skinnider MA, Hutson TH, Regazzi N, Gautier M, Demesmaeker R, Komi S, Ceto S, James ND, Cho N, Baud L, Galan K, Matson KJE, Rowald A, Kim K, Wang R, Minassian K, Prior JO, Asboth L, Barraud Q, Lacour SP, Levine AJ, Wagner F, Bloch J, Squair JW, Courtine G. The neurons that restore walking after paralysis. Nature. 2022;611:540–547. doi: 10.1038/s41586-022-05385-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Guan B, Fan Y, Zheng R, Fu R, Yao L, Wang W, Li G, Chen L, Zhou H, Feng S. A critical appraisal of clinical practice guidelines on pharmacological treatments for spinal cord injury. Spine J. 2023;23:392–402. doi: 10.1016/j.spinee.2022.09.009. [DOI] [PubMed] [Google Scholar]
  • 50.Andrzejewska A, Dabrowska S, Lukomska B, Janowski M. Mesenchymal stem cells for neurological disorders. Adv Sci (Weinh) 2021;8:2002944. doi: 10.1002/advs.202002944. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Cao T, Chen H, Huang W, Xu S, Liu P, Zou W, Pang M, Xu Y, Bai X, Liu B, Rong L, Cui ZK, Li M. hUC-MSC-mediated recovery of subacute spinal cord injury through enhancing the pivotal subunits β3 and γ2 of the GABAA receptor. Theranostics. 2022;12:3057–3078. doi: 10.7150/thno.72015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Pegtel DM, Gould SJ. Exosomes. Annu Rev Biochem. 2019;88:487–514. doi: 10.1146/annurev-biochem-013118-111902. [DOI] [PubMed] [Google Scholar]
  • 53.van Niel G, D’Angelo G, Raposo G. Shedding light on the cell biology of extracellular vesicles. Nat Rev Mol Cell Biol. 2018;19:213–228. doi: 10.1038/nrm.2017.125. [DOI] [PubMed] [Google Scholar]
  • 54.Raposo G, Stoorvogel W. Extracellular vesicles: exosomes, microvesicles, and friends. J Cell Biol. 2013;200:373–383. doi: 10.1083/jcb.201211138. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Mathieu M, Martin-Jaular L, Lavieu G, Théry C. Specificities of secretion and uptake of exosomes and other extracellular vesicles for cell-to-cell communication. Nat Cell Biol. 2019;21:9–17. doi: 10.1038/s41556-018-0250-9. [DOI] [PubMed] [Google Scholar]
  • 56.Jeppesen DK, Fenix AM, Franklin JL, Higginbotham JN, Zhang Q, Zimmerman LJ, Liebler DC, Ping J, Liu Q, Evans R, Fissell WH, Patton JG, Rome LH, Burnette DT, Coffey RJ. Reassessment of exosome composition. Cell. 2019;177:428–445. e418. doi: 10.1016/j.cell.2019.02.029. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.van Niel G, Carter DRF, Clayton A, Lambert DW, Raposo G, Vader P. Challenges and directions in studying cell-cell communication by extracellular vesicles. Nat Rev Mol Cell Biol. 2022;23:369–382. doi: 10.1038/s41580-022-00460-3. [DOI] [PubMed] [Google Scholar]
  • 58.Colombo M, Raposo G, Théry C. Biogenesis, secretion, and intercellular interactions of exosomes and other extracellular vesicles. Annu Rev Cell Dev Biol. 2014;30:255–289. doi: 10.1146/annurev-cellbio-101512-122326. [DOI] [PubMed] [Google Scholar]
  • 59.Théry C, Witwer KW, Aikawa E, Alcaraz MJ, Anderson JD, Andriantsitohaina R, Antoniou A, Arab T, Archer F, Atkin-Smith GK, Ayre DC, Bach JM, Bachurski D, Baharvand H, Balaj L, Baldacchino S, Bauer NN, Baxter AA, Bebawy M, Beckham C, Bedina Zavec A, Benmoussa A, Berardi AC, Bergese P, Bielska E, Blenkiron C, Bobis-Wozowicz S, Boilard E, Boireau W, Bongiovanni A, Borràs FE, Bosch S, Boulanger CM, Breakefield X, Breglio AM, Brennan MÁ, Brigstock DR, Brisson A, Broekman ML, Bromberg JF, Bryl-Górecka P, Buch S, Buck AH, Burger D, Busatto S, Buschmann D, Bussolati B, Buzás EI, Byrd JB, Camussi G, Carter DR, Caruso S, Chamley LW, Chang YT, Chen C, Chen S, Cheng L, Chin AR, Clayton A, Clerici SP, Cocks A, Cocucci E, Coffey RJ, Cordeiro-da-Silva A, Couch Y, Coumans FA, Coyle B, Crescitelli R, Criado MF, D’Souza-Schorey C, Das S, Datta Chaudhuri A, de Candia P, De Santana EF, De Wever O, Del Portillo HA, Demaret T, Deville S, Devitt A, Dhondt B, Di Vizio D, Dieterich LC, Dolo V, Dominguez Rubio AP, Dominici M, Dourado MR, Driedonks TA, Duarte FV, Duncan HM, Eichenberger RM, Ekström K, El Andaloussi S, Elie-Caille C, Erdbrügger U, Falcón-Pérez JM, Fatima F, Fish JE, Flores-Bellver M, Försönits A, Frelet-Barrand A, Fricke F, Fuhrmann G, Gabrielsson S, Gámez-Valero A, Gardiner C, Gärtner K, Gaudin R, Gho YS, Giebel B, Gilbert C, Gimona M, Giusti I, Goberdhan DC, Görgens A, Gorski SM, Greening DW, Gross JC, Gualerzi A, Gupta GN, Gustafson D, Handberg A, Haraszti RA, Harrison P, Hegyesi H, Hendrix A, Hill AF, Hochberg FH, Hoffmann KF, Holder B, Holthofer H, Hosseinkhani B, Hu G, Huang Y, Huber V, Hunt S, Ibrahim AG, Ikezu T, Inal JM, Isin M, Ivanova A, Jackson HK, Jacobsen S, Jay SM, Jayachandran M, Jenster G, Jiang L, Johnson SM, Jones JC, Jong A, Jovanovic-Talisman T, Jung S, Kalluri R, Kano SI, Kaur S, Kawamura Y, Keller ET, Khamari D, Khomyakova E, Khvorova A, Kierulf P, Kim KP, Kislinger T, Klingeborn M, Klinke DJ, Kornek M, Kosanović MM, Kovács ÁF, Krämer-Albers EM, Krasemann S, Krause M, Kurochkin IV, Kusuma GD, Kuypers S, Laitinen S, Langevin SM, Languino LR, Lannigan J, Lässer C, Laurent LC, Lavieu G, Lázaro-Ibáñez E, Le Lay S, Lee MS, Lee YXF, Lemos DS, Lenassi M, Leszczynska A, Li IT, Liao K, Libregts SF, Ligeti E, Lim R, Lim SK, Linē A, Linnemannstöns K, Llorente A, Lombard CA, Lorenowicz MJ, Lörincz ÁM, Lötvall J, Lovett J, Lowry MC, Loyer X, Lu Q, Lukomska B, Lunavat TR, Maas SL, Malhi H, Marcilla A, Mariani J, Mariscal J, Martens-Uzunova ES, Martin-Jaular L, Martinez MC, Martins VR, Mathieu M, Mathivanan S, Maugeri M, McGinnis LK, McVey MJ, Meckes DG, Meehan KL, Mertens I, Minciacchi VR, Möller A, Møller Jørgensen M, Morales-Kastresana A, Morhayim J, Mullier F, Muraca M, Musante L, Mussack V, Muth DC, Myburgh KH, Najrana T, Nawaz M, Nazarenko I, Nejsum P, Neri C, Neri T, Nieuwland R, Nimrichter L, Nolan JP, Nolte-’t Hoen EN, Noren Hooten N, O’Driscoll L, O’Grady T, O’Loghlen A, Ochiya T, Olivier M, Ortiz A, Ortiz LA, Osteikoetxea X, Østergaard O, Ostrowski M, Park J, Pegtel DM, Peinado H, Perut F, Pfaffl MW, Phinney DG, Pieters BC, Pink RC, Pisetsky DS, Pogge von Strandmann E, Polakovicova I, Poon IK, Powell BH, Prada I, Pulliam L, Quesenberry P, Radeghieri A, Raffai RL, Raimondo S, Rak J, Ramirez MI, Raposo G, Rayyan MS, Regev-Rudzki N, Ricklefs FL, Robbins PD, Roberts DD, Rodrigues SC, Rohde E, Rome S, Rouschop KM, Rughetti A, Russell AE, Saá P, Sahoo S, Salas-Huenuleo E, Sánchez C, Saugstad JA, Saul MJ, Schiffelers RM, Schneider R, Schøyen TH, Scott A, Shahaj E, Sharma S, Shatnyeva O, Shekari F, Shelke GV, Shetty AK, Shiba K, Siljander PR, Silva AM, Skowronek A, Snyder OL, Soares RP, Sódar BW, Soekmadji C, Sotillo J, Stahl PD, Stoorvogel W, Stott SL, Strasser EF, Swift S, Tahara H, Tewari M, Timms K, Tiwari S, Tixeira R, Tkach M, Toh WS, Tomasini R, Torrecilhas AC, Tosar JP, Toxavidis V, Urbanelli L, Vader P, van Balkom BW, van der Grein SG, Van Deun J, van Herwijnen MJ, Van Keuren-Jensen K, van Niel G, van Royen ME, van Wijnen AJ, Vasconcelos MH, Vechetti IJ, Veit TD, Vella LJ, Velot É, Verweij FJ, Vestad B, Viñas JL, Visnovitz T, Vukman KV, Wahlgren J, Watson DC, Wauben MH, Weaver A, Webber JP, Weber V, Wehman AM, Weiss DJ, Welsh JA, Wendt S, Wheelock AM, Wiener Z, Witte L, Wolfram J, Xagorari A, Xander P, Xu J, Yan X, Yáñez-Mó M, Yin H, Yuana Y, Zappulli V, Zarubova J, Žėkas V, Zhang JY, Zhao Z, Zheng L, Zheutlin AR, Zickler AM, Zimmermann P, Zivkovic AM, Zocco D, Zuba-Surma EK. 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. doi: 10.1080/20013078.2018.1535750. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Mori MA, Ludwig RG, Garcia-Martin R, Brandão BB, Kahn CR. Extracellular miRNAs: from biomarkers to mediators of physiology and disease. Cell Metab. 2019;30:656–673. doi: 10.1016/j.cmet.2019.07.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Robbins PD, Morelli AE. Regulation of immune responses by extracellular vesicles. Nat Rev Immunol. 2014;14:195–208. doi: 10.1038/nri3622. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Cai X, Zhang ZY, Yuan JT, Ocansey DKW, Tu Q, Zhang X, Qian H, Xu WR, Qiu W, Mao F. hucMSC-derived exosomes attenuate colitis by regulating macrophage pyroptosis via the miR-378a-5p/NLRP3 axis. Stem Cell Res Ther. 2021;12:416. doi: 10.1186/s13287-021-02492-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Sun Y, Shi H, Yin S, Ji C, Zhang X, Zhang B, Wu P, Shi Y, Mao F, Yan Y, Xu W, Qian H. Human mesenchymal stem cell derived exosomes alleviate type 2 Diabetes Mellitus by reversing peripheral insulin resistance and relieving β-cell destruction. ACS Nano. 2018;12:7613–7628. doi: 10.1021/acsnano.7b07643. [DOI] [PubMed] [Google Scholar]
  • 64.Jin C, Wu P, Li L, Xu W, Qian H. Exosomes: emerging therapy delivery tools and biomarkers for kidney diseases. Stem Cells Int. 2021;2021:7844455. doi: 10.1155/2021/7844455. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Jin Q, Wu P, Zhou X, Qian H, Xu W. Extracellular vesicles: novel roles in neurological disorders. Stem Cells Int. 2021;2021:6640836. doi: 10.1155/2021/6640836. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Li T, Yan Y, Wang B, Qian H, Zhang X, Shen L, Wang M, Zhou Y, Zhu W, Li W, Xu W. Exosomes derived from human umbilical cord mesenchymal stem cells alleviate liver fibrosis. Stem Cells Dev. 2013;22:845–854. doi: 10.1089/scd.2012.0395. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Zhang B, Wang M, Gong A, Zhang X, Wu X, Zhu Y, Shi H, Wu L, Zhu W, Qian H, Xu W. HucMSC-exosome mediated-Wnt4 signaling is required for cutaneous wound healing. Stem Cells. 2015;33:2158–2168. doi: 10.1002/stem.1771. [DOI] [PubMed] [Google Scholar]
  • 68.Zhang B, Shi Y, Gong A, Pan Z, Shi H, Yang H, Fu H, Yan Y, Zhang X, Wang M, Zhu W, Qian H, Xu W. HucMSC exosome-delivered 14-3-3ζ orchestrates self-control of the Wnt response via modulation of YAP during cutaneous regeneration. Stem Cells. 2016;34:2485–2500. doi: 10.1002/stem.2432. [DOI] [PubMed] [Google Scholar]
  • 69.Wu P, Zhang B, Han X, Sun Y, Sun Z, Li L, Zhou X, Jin Q, Fu P, Xu W, Qian H. HucMSC exosome-delivered 14-3-3ζ alleviates ultraviolet radiation-induced photodamage via SIRT1 pathway modulation. Aging (Albany NY) 2021;13:11542–11563. doi: 10.18632/aging.202851. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Yan Y, Jiang W, Tan Y, Zou S, Zhang H, Mao F, Gong A, Qian H, Xu W. hucMSC exosome-derived GPX1 Is required for the recovery of hepatic oxidant injury. Mol Ther. 2017;25:465–479. doi: 10.1016/j.ymthe.2016.11.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Ji C, Zhang J, Zhu Y, Shi H, Yin S, Sun F, Wang Q, Zhang L, Yan Y, Zhang X, Xu W, Qian H. Exosomes derived from hucMSC attenuate renal fibrosis through CK1δ/β-TRCP-mediated YAP degradation. Cell Death Dis. 2020;11:327. doi: 10.1038/s41419-020-2510-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Sun F, Sun Y, Zhu J, Wang X, Ji C, Zhang J, Chen S, Yu Y, Xu W, Qian H. Mesenchymal stem cells-derived small extracellular vesicles alleviate diabetic retinopathy by delivering NEDD4. Stem Cell Res Ther. 2022;13:293. doi: 10.1186/s13287-022-02983-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Tan Y, Huang Y, Mei R, Mao F, Yang D, Liu J, Xu W, Qian H, Yan Y. HucMSC-derived exosomes delivered BECN1 induces ferroptosis of hepatic stellate cells via regulating the xCT/GPX4 axis. Cell Death Dis. 2022;13:319. doi: 10.1038/s41419-022-04764-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Sung SE, Seo MS, Kim YI, Kang KK, Choi JH, Lee S, Sung M, Yim SG, Lim JH, Seok HG, Yang SY, Lee GW. Human epidural AD-MSC exosomes improve function recovery after spinal cord injury in rats. Biomedicines. 2022;10:678. doi: 10.3390/biomedicines10030678. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Nakazaki M, Morita T, Lankford KL, Askenase PW, Kocsis JD. Small extracellular vesicles released by infused mesenchymal stromal cells target M2 macrophages and promote TGF-beta upregulation, microvascular stabilization and functional recovery in a rodent model of severe spinal cord injury. J Extracell Vesicles. 2021;10:e12137. doi: 10.1002/jev2.12137. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Jia X, Huang G, Wang S, Long M, Tang X, Feng D, Zhou Q. Extracellular vesicles derived from mesenchymal stem cells containing microRNA-381 protect against spinal cord injury in a rat model via the BRD4/WNT5A axis. Bone Joint Res. 2021;10:328–339. doi: 10.1302/2046-3758.105.BJR-2020-0020.R1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Wang C, Wang M, Xia K, Wang J, Cheng F, Shi K, Ying L, Yu C, Xu H, Xiao S, Liang C, Li F, Lei B, Chen Q. 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. doi: 10.1016/j.bioactmat.2021.01.029. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Varadarajan SG, Hunyara JL, Hamilton NR, Kolodkin AL, Huberman AD. Central nervous system regeneration. Cell. 2022;185:77–94. doi: 10.1016/j.cell.2021.10.029. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Huang JH, Fu CH, Xu Y, Yin XM, Cao Y, Lin FY. Extracellular Vesicles derived from epidural fat-mesenchymal stem cells attenuate NLRP3 inflammasome activation and improve functional recovery after spinal cord injury. Neurochem Res. 2020;45:760–771. doi: 10.1007/s11064-019-02950-x. [DOI] [PubMed] [Google Scholar]
  • 80.Zhao C, Zhou X, Qiu J, Xin D, Li T, Chu X, Yuan H, Wang H, Wang Z, Wang D. Exosomes derived from bone marrow mesenchymal stem cells inhibit complement activation in rats with spinal cord injury. Drug Des Devel Ther. 2019;13:3693–3704. doi: 10.2147/DDDT.S209636. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Liang ZY, Xu XJ, Rao J, Yang ZL, Wang CH, Chen CM. Mesenchymal stem cell-derived exosomal MiRNAs promote M2 macrophages polarization: therapeutic opportunities for spinal cord injury. Front Mol Neurosci. 2022;15:926928. doi: 10.3389/fnmol.2022.926928. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.An N, Yang J, Wang H, Sun S, Wu H, Li L, Li M. Mechanism of mesenchymal stem cells in spinal cord injury repair through macrophage polarization. Cell Biosci. 2021;11:41. doi: 10.1186/s13578-021-00554-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Nakazaki M, Morita T, Lankford KL, Askenase PW, Kocsis JD. Small extracellular vesicles released by infused mesenchymal stromal cells target M2 macrophages and promote TGF-β upregulation, microvascular stabilization and functional recovery in a rodent model of severe spinal cord injury. J Extracell Vesicles. 2021;10:e12137. doi: 10.1002/jev2.12137. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Lu Y, Zhou Y, Zhang R, Wen L, Wu K, Li Y, Yao Y, Duan R, Jia Y. 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. doi: 10.3389/fnins.2019.00209. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Zhou Y, Wen LL, Li YF, Wu KM, Duan RR, Yao YB, Jing LJ, Gong Z, Teng JF, Jia YJ. Exosomes derived from bone marrow mesenchymal stem cells protect the injured spinal cord by inhibiting pericyte pyroptosis. Neural Regen Res. 2022;17:194–202. doi: 10.4103/1673-5374.314323. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Chen Y, Tian Z, He L, Liu C, Wang N, Rong L, Liu B. Exosomes derived from miR-26a-modified MSCs promote axonal regeneration via the PTEN/AKT/mTOR pathway following spinal cord injury. Stem Cell Res Ther. 2021;12:224. doi: 10.1186/s13287-021-02282-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Xiao X, Li W, Rong D, Xu Z, Zhang Z, Ye H, Xie L, Wu Y, Zhang Y, Wang X. Human umbilical cord mesenchymal stem cells-derived extracellular vesicles facilitate the repair of spinal cord injury via the miR-29b-3p/PTEN/Akt/mTOR axis. Cell Death Discov. 2021;7:212. doi: 10.1038/s41420-021-00572-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Han T, Song P, Wu Z, Xiang X, Liu Y, Wang Y, Fang H, Niu Y, Shen C. MSC secreted extracellular vesicles carrying TGF-beta upregulate Smad 6 expression and promote the regrowth of neurons in spinal cord injured rats. Stem Cell Rev Rep. 2022;18:1078–1096. doi: 10.1007/s12015-021-10219-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Hu X, Liu Z, Zhou X, Jin Q, Xu W, Zhai X, Fu Q, Qian H. Small extracellular vesicles derived from mesenchymal stem cell facilitate functional recovery in spinal cord injury by activating neural stem cells via the ERK1/2 pathway. Front Cell Neurosci. 2022;16:954597. doi: 10.3389/fncel.2022.954597. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Zhou W, Silva M, Feng C, Zhao S, Liu L, Li S, Zhong J, Zheng W. 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. doi: 10.1186/s13287-021-02248-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Oliveira E, Assuncao-Silva RC, Ziv-Polat O, Gomes ED, Teixeira FG, Silva NA, Shahar A, Salgado AJ. Influence of different ECM-Like hydrogels on neurite outgrowth induced by adipose tissue-derived stem cells. Stem Cells Int. 2017;2017:6319129. doi: 10.1155/2017/6319129. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Wechsler ME, Rao VV, Borelli AN, Anseth KS. Engineering the MSC secretome: a hydrogel focused approach. Adv Healthc Mater. 2021;10:e2001948. doi: 10.1002/adhm.202001948. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Fan L, Liu C, Chen X, Zheng L, Zou Y, Wen H, Guan P, Lu F, Luo Y, Tan G, Yu P, Chen D, Deng C, Sun Y, Zhou L, Ning C. Exosomes-loaded electroconductive hydrogel synergistically promotes tissue repair after spinal cord injury via immunoregulation and enhancement of myelinated axon growth. Adv Sci (Weinh) 2022;9:e2105586. doi: 10.1002/advs.202105586. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Samal JRK, Rangasami VK, Samanta S, Varghese OP, Oommen OP. Discrepancies on the role of oxygen gradient and culture condition on mesenchymal stem cell fate. Adv Healthc Mater. 2021;10:e2002058. doi: 10.1002/adhm.202002058. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Kasoju N, Wang H, Zhang B, George J, Gao S, Triffitt JT, Cui Z, Ye H. Transcriptomics of human multipotent mesenchymal stromal cells: Retrospective analysis and future prospects. Biotechnol Adv. 2017;35:407–418. doi: 10.1016/j.biotechadv.2017.04.005. [DOI] [PubMed] [Google Scholar]
  • 96.Peng H, Li Y, Ji W, Zhao R, Lu Z, Shen J, Wu Y, Wang J, Hao Q, Wang J, Wang W, Yang J, Zhang X. Intranasal administration of self-oriented nanocarriers based on therapeutic exosomes for synergistic treatment of parkinson’s disease. ACS Nano. 2022;16:869–884. doi: 10.1021/acsnano.1c08473. [DOI] [PubMed] [Google Scholar]
  • 97.Perets N, Betzer O, Shapira R, Brenstein S, Angel A, Sadan T, Ashery U, Popovtzer R, Offen D. Golden exosomes selectively target brain pathologies in neurodegenerative and neurodevelopmental disorders. Nano Lett. 2019;19:3422–3431. doi: 10.1021/acs.nanolett.8b04148. [DOI] [PubMed] [Google Scholar]
  • 98.Chen S, Sun F, Qian H, Xu W, Jiang J. Preconditioning and engineering strategies for improving the efficacy of mesenchymal stem cell-derived exosomes in cell-free therapy. Stem Cells Int. 2022;2022:1779346. doi: 10.1155/2022/1779346. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Ferreira JR, Teixeira GQ, Santos SG, Barbosa MA, Almeida-Porada G, Gonçalves RM. Mesenchymal stromal cell secretome: influencing therapeutic potential by cellular pre-conditioning. Front Immunol. 2018;9:2837. doi: 10.3389/fimmu.2018.02837. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Ahmed NE, Murakami M, Kaneko S, Nakashima M. The effects of hypoxia on the stemness properties of human dental pulp stem cells (DPSCs) Sci Rep. 2016;6:35476. doi: 10.1038/srep35476. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Huang T, Jia Z, Fang L, Cheng Z, Qian J, Xiong F, Tian F, He X. Extracellular vesicle-derived miR-511-3p from hypoxia preconditioned adipose mesenchymal stem cells ameliorates spinal cord injury through the TRAF6/S1P axis. Brain Res Bull. 2022;180:73–85. doi: 10.1016/j.brainresbull.2021.12.015. [DOI] [PubMed] [Google Scholar]
  • 102.Liang Y, Wu JH, Zhu JH, 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;39:701–714. doi: 10.1089/neu.2021.0290. [DOI] [PubMed] [Google Scholar]
  • 103.Liu W, Tang P, Wang J, Ye W, Ge X, Rong Y, Ji C, Wang Z, Bai J, Fan J, Yin G, Cai W. Extracellular vesicles derived from melatonin-preconditioned mesenchymal stem cells containing USP29 repair traumatic spinal cord injury by stabilizing NRF2. J Pineal Res. 2021;71:e12769. doi: 10.1111/jpi.12769. [DOI] [PubMed] [Google Scholar]
  • 104.Fu P, Zhang J, Li H, Mak M, Xu W, Tao Z. Extracellular vesicles as delivery systems at nano-/micro-scale. Adv Drug Deliv Rev. 2021;179:113910. doi: 10.1016/j.addr.2021.113910. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Wu P, Zhang B, Ocansey DKW, Xu W, Qian H. Extracellular vesicles: a bright star of nanomedicine. Biomaterials. 2021;269:120467. doi: 10.1016/j.biomaterials.2020.120467. [DOI] [PubMed] [Google Scholar]
  • 106.Wang B, Chang M, Zhang R, Wo J, Wu B, Zhang H, Zhou Z, Li Z, Zhang F, Zhong C, Tang S, Yang S, Sun G. Spinal cord injury target-immunotherapy with TNF-alpha autoregulated and feedback-controlled human umbilical cord mesenchymal stem cell derived exosomes remodelled by CRISPR/Cas9 plasmid. Biomater Adv. 2022;133:112624. doi: 10.1016/j.msec.2021.112624. [DOI] [PubMed] [Google Scholar]
  • 107.Zhai X, Chen K, Yang H, Li B, Zhou T, Wang H, Zhou H, Chen S, Zhou X, Wei X, Bai Y, Li M. Extracellular vesicles derived from CD73 modified human umbilical cord mesenchymal stem cells ameliorate inflammation after spinal cord injury. J Nanobiotechnology. 2021;19:274. doi: 10.1186/s12951-021-01022-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Yu T, Zhao C, Hou S, Zhou W, Wang B, Chen Y. Exosomes secreted from miRNA-29b-modified mesenchymal stem cells repaired spinal cord injury in rats. Braz J Med Biol Res. 2019;52:e8735. doi: 10.1590/1414-431X20198735. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Lee JR, Kyung JW, Kumar H, Kwon SP, Song SY, Han IB, Kim BS. Targeted delivery of mesenchymal stem cell-derived nanovesicles for spinal cord injury treatment. Int J Mol Sci. 2020;21:4185. doi: 10.3390/ijms21114185. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Kim HY, Kumar H, Jo MJ, Kim J, Yoon JK, Lee JR, Kang M, Choo YW, Song SY, Kwon SP, Hyeon T, Han IB, Kim BS. Therapeutic efficacy-potentiated and diseased organ-targeting nanovesicles derived from mesenchymal stem cells for spinal cord injury treatment. Nano Lett. 2018;18:4965–4975. doi: 10.1021/acs.nanolett.8b01816. [DOI] [PubMed] [Google Scholar]
  • 111.Guo S, Perets N, Betzer O, Ben-Shaul S, Sheinin A, Michaelevski I, Popovtzer R, Offen D, Levenberg S. Intranasal delivery of mesenchymal stem cell derived exosomes loaded with phosphatase and tensin homolog siRNA repairs complete spinal cord injury. ACS Nano. 2019;13:10015–10028. doi: 10.1021/acsnano.9b01892. [DOI] [PubMed] [Google Scholar]
  • 112.Liu WZ, Ma ZJ, Li JR, Kang XW. Mesenchymal stem cell-derived exosomes: therapeutic opportunities and challenges for spinal cord injury. Stem Cell Res Ther. 2021;12:102. doi: 10.1186/s13287-021-02153-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Lener T, Gimona M, Aigner L, Börger V, Buzas E, Camussi G, Chaput N, Chatterjee D, Court FA, Del Portillo HA, O’Driscoll L, Fais S, Falcon-Perez JM, Felderhoff-Mueser U, Fraile L, Gho YS, Görgens A, Gupta RC, Hendrix A, Hermann DM, Hill AF, Hochberg F, Horn PA, de Kleijn D, Kordelas L, Kramer BW, Krämer-Albers EM, Laner-Plamberger S, Laitinen S, Leonardi T, Lorenowicz MJ, Lim SK, Lötvall J, Maguire CA, Marcilla A, Nazarenko I, Ochiya T, Patel T, Pedersen S, Pocsfalvi G, Pluchino S, Quesenberry P, Reischl IG, Rivera FJ, Sanzenbacher R, Schallmoser K, Slaper-Cortenbach I, Strunk D, Tonn T, Vader P, van Balkom BW, Wauben M, Andaloussi SE, Théry C, Rohde E, Giebel B. Applying extracellular vesicles based therapeutics in clinical trials - an ISEV position paper. J Extracell Vesicles. 2015;4:30087. doi: 10.3402/jev.v4.30087. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Piffoux M, Volatron J, Cherukula K, Aubertin K, Wilhelm C, Silva AKA, Gazeau F. Engineering and loading therapeutic extracellular vesicles for clinical translation: a data reporting frame for comparability. Adv Drug Deliv Rev. 2021;178:113972. doi: 10.1016/j.addr.2021.113972. [DOI] [PubMed] [Google Scholar]
  • 115.Chen J, Li P, Zhang T, Xu Z, Huang X, Wang R, Du L. Review on strategies and technologies for exosome isolation and purification. Front Bioeng Biotechnol. 2022;9:811971. doi: 10.3389/fbioe.2021.811971. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Flack JA, Sharma KD, Xie JY. Delving into the recent advancements of spinal cord injury treatment: a review of recent progress. Neural Regen Res. 2022;17:283–291. doi: 10.4103/1673-5374.317961. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Kim GU, Sung SE, Kang KK, Choi JH, Lee S, Sung M, Yang SY, Kim SK, Kim YI, Lim JH, Seo MS, Lee GW. Therapeutic potential of mesenchymal stem cells (MSCs) and MSC-derived extracellular vesicles for the treatment of spinal cord injury. Int J Mol Sci. 2021;22:13672. doi: 10.3390/ijms222413672. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from American Journal of Translational Research are provided here courtesy of e-Century Publishing Corporation

RESOURCES