Skip to main content

Extracellular vesicle therapy in neurological disorders

Abstract

Extracellular vesicles (EVs) are vital for cell-to-cell communication, transferring proteins, lipids, and nucleic acids in various physiological and pathological processes. They play crucial roles in immune modulation and tissue regeneration but are also involved in pathogenic conditions like inflammation and degenerative disorders. EVs have heterogeneous populations and cargo, with numerous subpopulations currently under investigations. EV therapy shows promise in stimulating tissue repair and serving as a drug delivery vehicle, offering advantages over cell therapy, such as ease of engineering and minimal risk of tumorigenesis. However, challenges remain, including inconsistent nomenclature, complex characterization, and underdeveloped large-scale production protocols. This review highlights the recent advances and significance of EVs heterogeneity, emphasizing the need for a better understanding of their roles in disease pathologies to develop tailored EV therapies for clinical applications in neurological disorders.

Graphical abstract

Highlights

This paper reviews recent advances in EV subpopulation and characterization. We discuss the potential of EVs to address multiple aspects of neurological diseases, including neuroinflammation, mitochondrial dysfunction, apoptosis, and blood-brain barrier leakage. The review emphasizes the complexity and heterogeneity of EVs, highlighting the need for better characterization and classification to optimize therapeutic applications. By understanding EV subtypes and their roles, we can develop more effective and tailored EV therapies for clinical use in treating neurological conditions.

Introduction

Extracellular vesicles (EVs) play a vital role in cell-to-cell communication, facilitating the transfer of proteins, lipids, and nucleic acids across various physiological and pathological processes acids [1, 2]. EVs were previously categorized into three classes based on their biogenesis: exosomes, microvesicles (MVs), and apoptotic bodies [1]. While numerous subpopulations continue to be identified, a clear understanding of their distinct functions remains elusive. Some aspects of biogenesis and regulation overlap among these classes. Furthermore, the heterogeneity of EV populations and their cargo is influenced by various factors [3]. Thus, in this study, we opt to classify EVs based on their size, providing a more generalized framework while also leaving room for future investigations.

Since EVs play pivotal roles in immune modulation [4] and tissue regeneration [5, 6], EVs represent a promising avenue for therapy across various medical domains. Compared to cell therapy, EVs offer several advantages, including greater versatility in delivery routes, ease of engineering, more concentrated cargo, absence of ethical concerns, and minimal risk of tumorigenesis or alloimmunization [7]. Moreover, EVs serve as a promising drug delivery platform [8,9,10]. However, several limitations hinder the real-world application of EVs, especially the characterization of their heterogenicity and exact therapeutic mechanisms.

This review underscores the importance of studying EV heterogeneity for therapeutic purposes in neurological disorders. Initially, we will delineate the various classes of EVs and elucidate the factors influencing their heterogeneity. Subsequent sections will delve into studies concerning the clinical utilization of EVs across diverse neurological disorders (Fig. 1). A better understanding of EV subpopulations and functions will pave the way for more tailored EV therapies.

Extracellular vesicle subtypes

EVs are lipid bilayer-bound vesicles released by cells, varying from 30 nm to 2000 nm in diameter, and cannot replicate [1, 11]. Various classification systems exist for EVs, including their cellular origin, biological function, or biogenesis (Table 1). However, there is still not yet a consensus on EV classification. Although distinct mechanisms underlie the formation of each type of EV, there are notable overlaps between subpopulations. For example, all EV classes involve actin-myosin interactions [29,30,31] and translocation of phosphatidylserine [32, 33]. A combination of markers is commonly utilized to define EV subtypes [34].

Table 1 EV populations and subpopulations

Numerous factors contribute to the heterogeneity of EVs, including cellular source, physiological state, and biological environment. EVs isolated from mesenchymal stem cells (MSCs) originating from different tissues exhibit variations in composition and function [35]. EVs from higher passage MSCs demonstrate reduced efficacy compared to those from younger cells [36]. EVs secreted from basolateral epithelial cells utilize distinct pathways compared to those from apical cells [20, 37]. A myriad of cell culture paradigms, such as 2D and 3D scaffolds, also impact EV composition [38, 39]. Furthermore, external stimuli such as inflammatory signals, ATP, heat stress, intracellular calcium levels, hypoxia, and various others can alter the composition of EVs, leading to environmental modification in EV therapy optimization [40,41,42,43].

Due to the difficulty in EV characterization, EVs can be roughly divided into small (< 200 nm) and large (> 200 nm) sizes. Small EVs, such as exosomes and MVs, are generally found to be more therapeutic than large EVs. However, the mechanism of action of each class of small EVs is different. For example, exosomes carry anti-inflammatory microRNA (miRNA) and growth factor receptors while MVs transfer functional mitochondria. Small EVs are also frequently used as drug delivery vehicles more often than large EVs. Finding the optimal treatment regimens of EVs and MVs will advance their safe and effective therapeutic applications for neurological disorders.

Small EVs

Small EVs are the most studied group of EVs, especially exosomes and MVs. Exosomes arise via the inward budding of multivesicular bodies (MVBs), generating intraluminal vesicles (ILVs) [44]. MVBs may fuse with the cell membrane for exosome secretion or with lysosomes for degradation [45, 46]. There are different exosome subtypes, as indicated by variations in ILV formation and cargo-loading mechanisms [2]. Exosomes employ two primary cargo sorting mechanisms: the endosomal sorting complexes required for transport (ESCRT) pathway and the ceramide-dependent mechanism [47, 48]. Key regulators of the ESCRT pathway, such as programmed cell death 6-interacting protein (PDCD6IP or ALIX) and tumor susceptibility gene 101 protein (TSG101), are often used as exosome markers [49, 50]. Tetraspanins, including CD9, CD63, CD81, and CD82, also play a role in cargo selection [51, 52]. Other well-known markers for exosomes are LAMP1/2 and Syntenin [15].

Small ectosomes are released through plasma membrane budding [15]. Differentiating between small ectosomes and exosomes within the small EV population requires a combination of presenting plasma membrane molecules and lacking endosomal markers, such as CD63. Small ectosomes, albeit similar in size to exosomes, are more enriched in centrosomal, ribosomal, and mitochondrial proteins and contain fewer oncogenic genes compared to exosomes [53].

Protrusion-derived ectosomes are released from membrane protrusions such as filopodia, microvilli, and cilia during cellular movement [16]. Bin–Amphiphysin–Rvs (I-BAR) domain-containing proteins, including MIM and IRSp53, connect the plasma membrane to actin, GTPase, and phosphoinositides [54, 55] to facilitate protrusion forming and also ectosome release. CD133, also known as prominin, is necessary for the release of ectosomes from microvilli [56, 57]. There are three proposed scission processes: first, ESCRT machinery recruitment, as evidenced by viral-induced vesicle release [58]; second, actomyosin contractility causing plasma membrane scission through GTPase ADP-ribosylation factor, though its exact mechanism—whether mechanical contraction or molecular signaling—remains unclear; and third, shear friction from extracellular fluid flow contributing to ectosome release. Prominin-1 and I-BAR domain-containing proteins are potential markers for protrusion-derived ectosomes, but further research is needed for precise classification.

Arrestin domain-containing protein 1 (ARRDC1)-mediated microvesicles (ARMMs) are generated through plasma membrane budding, akin to virus-induced MVs. The PSAP motif of ARRDC1 on the plasma membrane recruits endosomal TSG101 to the cell membrane for budding. The VPS4 ATPase facilitates the final budding process of ARMMs, similar to exosome and viral budding [17]. ARRDC1 is specifically recruited in PPXY-mediated budding and interacts with HECT ubiquitin ligases such as WW1, WW2, and [18]. ARRDC1 serves as a marker for ARMMs, and due to their biogenesis, these MVs are negative for endosomal markers such as LAMP3 and CD63 [17].

Intracellular membrane-derived ectosomes exhibit two distinct release mechanisms: slow-releasing and fast-releasing. In the slow-releasing method, ectosomes are secreted via outward budding of the plasma membrane [19]. Conversely, the fast-releasing process involves intracellular vesicles being directly squeezed out through pores in the plasma membrane, resulting in ectosomes with different components compared to conventional plasma membrane-derived ectosomes. Ectosomes released in the fast phase possess negatively charged phospholipids, typically found in the inner membrane, whereas conventional ectosomes share similar plasma membrane components with the host cells.

Small EVs, particularly exosomes and small ectosomes, have been extensively studied for their critical roles in intercellular communication. Despite differences in their biogenesis, overlapping mechanisms highlight the necessity for combination markers in their characterization. However, the characterization of their subpopulation within the same classes is still not clear. The heterogenicity of their cargo leads to inconsistent results in treatment outcomes and mechanism of action. Better characterization and isolation techniques as well as a complete content profile are important for their utilization. Protrusion-derived ectosomes and ARMMs, though less understood, offer insights into cell movement-related and viral-induced communication, respectively. Intracellular membrane-derived ectosomes represent another subtype, and the impact of their negatively charged membranes on targeting ability remains unexplored. Although exosomes and small ectosomes from sources like stem cells have shown therapeutic potential, their optimal treatment regimen, as well as the utility of yet to be examined small EVs, remain unclear and warrant additional studies.

Small-to-large EVs

MVs, generally known as ectosomes or shedding vesicles, are generated via direct outward budding of the cell membrane [46, 59]. Despite ongoing research, the mechanisms underlying cargo sorting in MVs remain elusive, with various proposed pathways such as calcium-induced cytoskeletal remodeling [60, 61], protein kinase C and purinergic receptors P2X7/P2Y [62, 63]. ARF6, TSG101, ceramide, and lipid rafts regulate the formation of both MVs and exosomes, suggesting shared underlying mechanisms [64, 65]. Following shedding, MVs either degrade rapidly, releasing their contents into the extracellular space, or engage in communication with specific target cells through receptor signaling, direct fusion with the plasma membrane, or endocytosis [66].

Apoptotic bodies, also known as apoptosomes, are vesicles released during programmed cell death [34, 67]. These bodies typically exhibit a large size and contain organelles within their vesicular structure [68, 69]. Apoptotic bodies are expelled via a 'beads-on-a-string' formation, a screening procedure that selectively excludes nuclear content from these bodies [70, 71]. Concurrently, smaller vesicles are also released, potentially originating from membrane blebbing during apoptosis [70, 72]. Most apoptotic bodies are phagocytosed by local macrophages, which recognize them through Annexin V, thrombospondin, and C3b [73,74,75]. Apoptotic bodies also carry genetic cargo, potentially involved in tumor metastasis [76, 77].

Small-to-large EVs encompass a wide range of sizes and properties. MVs exemplify a pivotal class of EV that has garnered much interest. Initially, MVs were often confused with exosomes due to their similar size. However, discoveries in their distinct biogenesis, cargo, and markers have clarified the diversity of EVs. MVs deserve more attention due to varying reports on their therapeutic effects. Their larger size and direct membrane budding biogenesis may be advantageous for developing drug delivery vehicles, as MVs can carry more content and functional organelles such as mitochondria. On the other hand, apoptotic bodies are generally pathogenic. Enhancing the clearance of apoptotic bodies or inhibiting their uptake by peripheral tissues can be a promising area of study to suppress inflammation and tumor metastasis, especially in relation to brain function will advance EV use in neurological disorders.

Large EVs

Large oncosomes manifest as large ectosomes originating from tumor cells. Oncoproteins such as MyrAkt1, HB-EGF, and caveolin-1 (Cav-1), as well as EGFR overexpression resulting in plasma membrane blebbing. The cargo of oncosomes can induce tumor spreading and progression [22]. In prostate cancer, tumor cells release oncosomes containing AKT1 kinase. The internalization of oncosomes leads to fibroblast reprogramming, promoting tumor growth via MYC activation and environmental modulation. Inhibition of oncosome uptake can prevent tumor progression, offering a novel therapeutic approach for cancer [78]. In addition to oncoproteins, cytokeratin 18 (CK18) is proposed as a marker for oncosomes.

Migrasomes are formed during cell migration [23]. During this process, large vesicles develop at the tips of retracting fibers behind the cells, relying on actin polymerization. Migrasomes contain abundant small vesicles, with diameters ranging from 50 to 100 nm, resembling pomegranates. Tetraspanin-4 (TSPAN4) has been identified as the most prominent marker for migrasomes, along with TSPAN7, cholesterol, and integrin α5 [24, 25]. Although the precise function of migrasomes remains unclear, they are hypothesized to facilitate cell–cell communication in a specific direction related to cell migration [23].

Midbody remnants, another type of ectosome, are remnants of the intercellular bridge formed during cell division [26]. They are rich in cytoskeletal proteins such as microtubules, centralspindlin, and the chromosomal passenger complex. This structure can either retract into daughter cells or be released into the extracellular space, where they may be degraded or internalized by neighboring cells. Midbody remnants are primarily reported in cancer cells [26, 79]. Uptake of midbody remnants secreted from cancer cells can induce a malignant phenotype in fibroblasts [26]. Midbody derivatives selectively accumulate in stem cells, leading to loss of differentiation and autophagy evasion through the binding of the CEP55 midbody protein to the autophagic receptor NBR1 [80]. The precise mechanism of action, whether through mutated protein cargo or epigenetic dysregulation, remains unknown.

Exophers represent large ectosomes with ambiguous biogenesis, containing organelles, particularly mitochondria and lysosomes, as well as protein aggregates such as huntingtin and tau. Most of the secreted exophers are taken up by neighboring cells. Exophers have also been found in remote tissues, suggesting secondary release after uptake [27]. During stress, cardiomyocytes excrete dysfunctional mitochondria into exophers driven by autophagy machinery. Impaired autophagy causes the accumulation of anomalous mitochondria, leading to dysfunctional ventricles and metabolism [81]. The role of exophers may be the eradication of toxins and dysfunctional organelles during stress.

Taken together, large oncosomes, migrasomes, midbody remnants, and exophers represent diverse and specialized ectosomes with significant roles in cellular processes and disease progression. Large oncosomes and midbody remnants, which play crucial roles in tumor progression and spreading, are potential targets for cancer therapy through the inhibition of their biogenesis and uptake. The discovery of migrasomes suggests direction-specific communication, but more research is needed to understand their effects and control mechanisms fully. Exophers help alleviate cellular stress by removing dysfunctional components. Understanding their function will elucidate organelle transfer and cellular stress management. Enhancing exopher production in situations involving organelle dysfunction-related diseases may mitigate pathogenesis. Probing the pathological and treatment modalities of these large EVs presents new avenues for research and therapeutic development for brain diseases.

EV therapy in neurological disorders

EVs play crucial roles in disease pathogenesis, especially immunomodulation (Table 2). Inflammation is involved across a spectrum of diseases, including degeneration, cancer, infections, and trauma [112]. EV therapy can be categorized into two methods: inhibiting pathogenic EVs and promoting therapeutic EVs [1].

Table 2 Pathologic and therapeutic effects of EVs

Pathologic EVs carry pro-inflammatory factors and toxic proteins. Blocking phosphatidylserine, a surface component crucial for EV sorting and uptake, can reduce EV uptake, consequently diminishing tumor growth and angiogenesis [113]. Additionally, targeting FAS ligands on EVs with anti-FASL monoclonal antibodies has been shown to reduce tumor progression [114]. However, inhibiting the EV cascade lacks specificity and may disrupt physiological processes.

Therapeutic EVs suppress inflammation and promote tissue regeneration. Therapeutic EVs can be derived from various sources, with stem cells being the most extensively studied due to their versatility [1]. While stem cell transplantation has shown great results in treating neurological disorders, their mechanism of action is primarily through paracrine effects rather than cellular replacement [115, 116]. Culture media derived from healthy cells can alleviate inflammation, promote angiogenesis, and restore function in the same way as cell transplantation [91]. EVs mediating these paracrine effects contain miRNA, non-coding RNA, growth factors, receptors, proteins, and lipids [1]. miRNA is believed to be indispensable for the therapeutic effects. EVs contain components involved in RNA transportation and processing such as RNA-binding proteins Staufen homolog 1 (STAU1), STAU2, Argonaute 2 (AGO2), and trinucleotide repeat-containing gene 6A protein (TNRC6A; also known as GW182). Ago2 knockdown diminishes the therapeutic effects of MSC-EVs [117].

Neurological disorders are always notoriously challenging due to the limited regenerative capabilities of neurons and the selective blood–brain barrier (BBB) preventing CNS entry of many therapeutic agents. EV therapy has a great advantage as it can precisely target brain parenchyma and effectively cross BBB (Table 3). Systemic administration or invasive methods such as intranasal spray can deliver EVs into the central nervous system (CNS). Here we summarize advances of EV therapy in neurological disorders across various pathologies to give a complete view of EV application: ischemia, trauma, degeneration, autoimmune-induced inflammation, and genetic mutation. In each disease section, we cover pathogenesis, pathologic EV involvement, mechanisms of therapeutic action, and EV optimization.

Table 3 Cell-derived EV therapy in neurological disorders

Stroke

Stroke, including ischemic and hemorrhagic strokes, ranks second as a cause of death and disability worldwide [189, 190]. The pathogenesis of brain injury following ischemia involves oxidative stress, inflammation, excitotoxicity, and apoptosis [191]. On the other hand, hemorrhagic stroke injury is from hematoma compression and increased intracranial pressure, subsequently also causing inflammation, excitotoxicity, and impaired BBB [192]. Interestingly, the upregulation of CD63 exosomes closely approximates with endogenous neurovascular unit regenerative process [193]. Although stroke intervention methods like mechanical thrombectomy and surgical decompression have rapidly improved, few treatments effectively address neuronal death [194]. Novel anti-inflammatory therapies targeting inflammatory cell recruitment have failed in clinical trials, suggesting that additional modalities, such as regeneration, may be necessary.

EV therapy can address multiple aspects of stroke pathophysiology and improve neurological outcomes [195], while minimizing complications associated with cell-based therapy [196]. Overall, most EV therapies improve infarct size, hematoma clearance, brain edema, and neurological functions [108, 119, 120, 124, 134]. Mechanistically, EVs derived from MSCs, adipose-derived stem cells (ASCs), and astrocytes promote anti-inflammatory M2 microglia polarization, suppress inflammatory cytokines, and reduce oxidative stress [88, 197, 198]. EVs carrying miR-132 can suppress eukaryotic elongation factor 2 kinase (eef2k) and restore VE-cadherin, an endothelial adhesive junction component [110]. ASCs-EVs also decrease aquaporin-4 (AQP-4) levels [105]. Mitochondria transfer by MVs and mitochondrial DNA via exosomes also increase the integrity of brain endothelial cells (BECs) [100, 101]. EVs effectively reduce apoptosis of neurons, oligodendrocytes, and astrocytes. EVs containing miR-138-5p reduce Lipocalin 2 and metastasis-associated lung adenocarcinoma transcript 1 (MALAT1), downregulating bax, caspase-3, caspase-8, and inflammatory cytokines while upregulating Bcl-2 and Cyclin family proteins [95, 98]. EVs containing miR-199a-5p also reduce apoptosis by ameliorating endoplasmic reticulum stress [102]. EVs promote neurogenesis and angiogenesis through miR-126, miR-26a, miR-181b, and multiple growth factors [107,108,109]. They also benefit the microbiome-gut-brain axis damaged after ischemic stroke by downregulating Interleukin (IL) 17 and upregulating IL-10, which modulates microbiota diversity and intestinal immunity [111].

Preconditioning, source selection, and combination therapy improve EV therapy efficacy [199, 200]. Pretreatment with hypoxia in MSC restores the BBB more effectively [201]. EVs from macrophages and microglia pretreated with IL-4 ameliorate apoptosis and promote angiogenesis, while EVs from tumor necrosis factor alpha (TNF-α) pretreated endothelial progenitor cells have the opposite effect [202, 203]. Mitochondria-containing EVs derived from brain endothelial cells from the same species donor are more effective in mitochondria transfer [204]. Better outcomes are observed in combined therapies with exercise, enriched environments, acupuncture, brain stimulation, and hypothermia [199].

EV therapy for stroke offers multiple therapeutic benefits, including anti-neuroinflammation, anti-apoptosis, BBB restoration, neurogenesis, and angiogenesis. However, the diverse range of therapeutic cargo and molecular pathways reported, even from the same cell source, leads to confusion about which cargo acts as the primary regulator. It remains unclear whether these components coexist within the same vesicles or are distributed among different EV subtypes. Various cell sources have been studied, but their effects on different domains of pathogenesis have not yet been compared. For instance, EVs derived from stem cells and anti-inflammatory glia may primarily modulate inflammation, while those from endothelial cells may be more effective in promoting angiogenesis. Preconditioning parent cells with hypoxia, which mimics ischemic stroke conditions, induces the secretion of EVs suited for such situations. Given that neuronal death is a primary cause of disabilities in ischemic stroke, EV therapy should focus more on long-term neurogenesis and functional recovery. Moreover, while most preclinical studies deliver EVs within the first hour following the lesion induction, actual patients typically receive treatment at a much later stage, implying the need to modify the experiment design [205]. Finally, the safety profile of EVs is a concern. Overexpression of vascular endothelial growth factor (VEGF) and hypoxia-inducible factor-1 (HIF-1) can potentially lead to BBB leakage and brain edema [206, 207]. Therefore, recognizing the risks and benefits of these secreted growth factors should guide the timing of administration and expression of specific EV components and will require further investigations.

Traumatic brain injury (TBI)

TBI is the most common cause of morbidity and mortality in the young population, commonly resulting from falls and traffic accidents [208]. Neurological damage occurs both from the initial impact (primary brain injury) and subsequent ischemia due to brain swelling (secondary injury). Despite several neuroprotective strategies, such as antioxidants, N-methyl-d-aspartate (NMDA) receptor antagonists, and calcium channel blockers, there is only minimal improvement [209]. TBI brain-derived EVs induce multisystemic organ dysfunction. Lactadherin can eliminate brain-derived EVs and improve coagulopathy and inflammation [210,211,212].

Like ischemic stroke, therapeutic EVs modulate neuroinflammation by promoting M2 microglia polarization and reducing pro-inflammatory cytokines, subsequently decreasing neuronal apoptosis. Astrocytes-derived EVs carry miR-873a-5p, inhibiting the ERK/NF-κB signaling pathway [93]. Additionally, microglia-derived EVs contain miR-124–3p, which suppress mTOR signaling, autophagy-associated FIP200 gene, Rela/ApoE pathway, and toll-like receptor-4 (TLR4) signaling pathway [133,134,135]. MSC-EVs suppress TRAF6 in the TLR4 signaling pathway via miR-146a, the cAMP/PKA/CREB pathway via miR-140-5p, and the CysLT2R-ERK1/2 pathway mediating M1 polarization [128, 213,214,215]. MSC-EVs also decrease the pro-apoptotic factor Bax while increasing the anti-apoptotic factor Bcl-2 expression [99]. For long-term complications of TBI, MSC-EVs inhibit chronic activation of the NLRP3-p38/MAPK signaling pathway and improve long-term cognitive function [129]. EVs also increase neuron survival by stimulating myelination [103] and transferring neuroprotective agents such as Apo-lipoprotein D (ApoD) [216, 217]. EVs upregulate genes associated with neurogenesis, synaptogenesis, and neuroplasticity while downregulating non-neuronal differentiation genes [104]. Neural stem cell-derived EVs (NSC-EVs) increase neurogenesis through miR-320-5p, miR-210, miR-21a, and miR-9 [218,219,220,221].

EV therapy effectively addresses neuroinflammation in both the acute and chronic phases of TBI while also promoting neurogenesis. Various miRNAs and their associated pathways are integral to this mechanism. Tailoring specific EV properties to different phases of pathology can enhance treatment outcomes. For example, administering anti-inflammatory EVs in the hyperacute phase may prevent secondary brain injury, while using pro-neurogenesis and pro-angiogenesis EVs later can improve functional recovery. Besides therapeutic EVs, temporarily blocking pathological EVs that signal inflammation may also benefit patients by reducing secondary damage, though current techniques for blocking EV release, explored in models of neurodegenerative disorders, still lack precision. Furthermore, since TBI often coincides with systemic damage such as hemorrhagic shock or organ trauma, it is crucial to investigate the effects and systemic distribution of EVs in these contexts. The modification of EV sources in TBI has not been as thoroughly studied as in ischemic stroke, presenting an opportunity to apply existing stroke-relevant knowledge to this field.

Spinal cord injury (SCI)

SCI, often caused by trauma, is a chronic disability that imposes a significant healthcare burden [222]. Like TBI, SCI involves primary injury from traction and compression forces, followed by secondary hypoperfusion due to spinal cord swelling [223, 224]. Inflammation further exacerbates the condition, leading to excitotoxicity and reactive oxygen species (ROS)-induced apoptosis. Despite therapeutic advances over the past decade, treatments for SCI, such as surgical decompression, corticosteroids, and neuroprotective agents, remain controversial and largely ineffective. Novel therapeutic strategies focus on limiting cell death, promoting regeneration, and restoring myelination. Pathologic EVs inhibit axon regeneration, induce systemic inflammation, and cause multi-organ damage [225].

Neural stem cell/progenitor cell (NPSC)-derived EVs reduce inflammation by suppressing the NLRP3 inflammasome and increasing autophagy-regulating Beclin-1 expression through the 14-3-3τ protein [140, 141]. They also induce angiogenesis by transferring VEGF to endothelial cells [142]. Cortical neuron-derived EVs suppress pro-inflammatory microglia and astrocytes through miR-124-3p [143]. MSC-derived EVs promote A1-to-A2 astrocyte conversion via miR-21 [136], suppress cyclooxygenase 2 (COX2) mRNA via miR-211-5p [94], inhibit macrophage/microglial pyroptosis through the miR-21a-5p/PELI1 axis-mediated autophagy pathway [226], and reduce neutrophil extracellular trap (NET) formation in both the spinal cord and circulation via miR-125a-3p [167]. Additionally, miR-26b-5p-enriched MSC-EVs epigenetically regulate the KDM6A/NOX4 axis to suppress inflammation and ROS production [139].

Environmentally modulated EVs perform better than naïve EVs [225]. Microglia-derived EVs function differently under pro-inflammatory or pro-regenerative preconditioning [227]. Similarly, EVs isolated from hypoxic MSCs have significantly higher potency in miR-146a-5p-mediated immune modulation [228]. NPSCs primed with insulin-like growth factor 1 (IGF-1) secrete EVs highly enriched in miR-219a-2-3p, which induce oligodendrocyte progenitor cell (OPC) maturation and promote axonal regeneration [229].

Similar to TBI, EVs help prevent secondary injury in SCI by suppressing inflammation. To establish EVs as a viable immunosuppressant therapy, comparative studies with conventional corticosteroids are needed to evaluate their efficacy. Currently, EV sources in SCI are limited to MSCs, NPSCs, and neurons. Exploring additional sources, such as endothelial cells and immune cells, can provide a more comprehensive treatment approach for SCI. Additionally, assessing the extent of SCI severity and determining the optimal treatment regimen specific to the EV source in tandem with adjunct therapies will be crucial for maximizing the beneficial effects of EVs.

Peripheral nerve injury (PNI)

Despite advances in neurology, there is still no effective therapy for nerve regeneration [230]. Although axons can regrow after injury, the growth rate is extremely slow and often complicated by inflammation and scar formation [231]. Neurorrhaphy is feasible only for short-gap injuries, while autologous nerve grafts have limitations, including nerve source selection and donor site dysfunction. Nerve guide conduits and cell therapy are potential candidates for PNI treatment but still face several complications [230, 232]. Pathologic EVs play an important role in blocking nerve growth. Schwann cells (SCs) secrete miR-1, inhibiting brain-derived neurotrophic factor (BDNF) expression and blocking axonal regeneration. miR-1 inhibitors efficiently improve SC proliferation and migration [233]. Injured dorsal root ganglia (DRG) secrete miR-23a-enriched EVs, targeting the A20 gene and promoting M1 macrophage polarization. EV-miR-23a antagomir reduces M1 macrophages, pro-inflammatory cytokines, and pain hypersensitivity [234].

EVs effectively target injured neurons and peripheral axons [235]. SC-derived EVs are highly focused candidates for PNI treatment [230]. Following PNI, SCs dedifferentiate to a progenitor-like state, guiding axonal regeneration. Exosomes from dedifferentiated SCs significantly increase axonal growth by inhibiting GTPase RhoA [148] and downregulating PTEN by miR-21 [147]. MSC-exosomes enhance neurite outgrowth by expressing neural growth factors such as BDNF, fibroblast growth factor 1 (FGF-1), glial cell line-derived neurotrophic factor (GDNF), IGF-1, and nerve growth factor (NGF), while MSC-MVs have the opposite effect [3, 144]. However, MVs derived from M1 macrophages increase SC proliferation and migration compared to those from M0 macrophages [149]. Other beneficial cell sources for nerve growth include olfactory ensheathing cells (OECs), pericytes, dental pulp stem cells, and induced pluripotent stem cells (iPSCs) [150,151,152, 236, 237]. EVs also address complications of PNI, such as injury-induced neuropathic pain [235] and denervation-induced muscle atrophy [238]. EVs are also effective in treating non-traumatic peripheral neuropathy, such as diabetic peripheral neuropathy and chemotherapy-induced peripheral neuropathy [239, 240].

Multiple optimization techniques show promise in PNI treatment. Mechanical stimulation of SCs increases miR-23b-3p-enriched EVs, which promote DRG neuron survival and neurite outgrowth [241]. Platelet-rich plasma (PRP) supplementation upregulates c-Jun and GDNF in the EVs while also promoting parent cell viability [242, 243]. Hypoxic neural crest cells promote sensory neuron repair through miR-21-5p [244]. EVs combined with conduits offer more efficient PNI treatment [244,245,246,247]. Even more advanced, a superparamagnetic nanocomposite scaffold, which can mechanically stimulate encapsulated SCs to release EVs, optimizes noninvasive and remotely time-scheduled nerve repair [248].

PNI models demonstrate the potential of EV therapy in the peripheral nervous system (PNS). EVs facilitate nerve regeneration by restoring SCs and DRG neurons while simultaneously suppressing inflammation. However, different EV subtypes yield distinct outcomes: exosomes are therapeutic, whereas MVs can be pathological. Due to the size range overlap between exosomes and MVs, the purification and characterization of EVs are critical for effective treatment outcomes. In addition to conventional peripheral nerve models, exploring the effects of EV therapy on cranial nerve injuries is also needed. Advances in EV-conduit integration and noninvasive remote scheduling systems show great promise for the future of PNI treatment, offering more precise and effective therapeutic options.

Epilepsy

Epilepsy stands as a major debilitating brain disorder complicated by numerous factors and genetic predispositions [249]. While antiepileptic medication can suppress seizures, it does not improve long-term outcomes. Epilepsy surgery is the most effective treatment but is only suitable for selected patients. Prolonged seizures, such as status epilepticus (SE), lead to an inflammatory cytokine storm mediated by activated microglia and reactive astrocytes, causing neurodegeneration, particularly in the hippocampus.

EVs can alleviate brain damage from epilepsy by modulating neuroinflammation [250], similar to stem cell transplantation [251]. However, there are only a few studies on EVs in epileptic models. Intranasal administration of MSC-exosomes immediately after SE reduces SE-induced injury in the hippocampus and preserves glutamatergic and GABAergic neurons [153]. Loading exogenous GABA into EVs can significantly suppress seizures. EVs derived from GABAergic interneurons (INs) and medial ganglionic eminence (MGE) cells were particularly effective, while EVs from NPSCs showed limited efficacy [252].

EVs reduce brain damage by suppressing the inflammatory cytokine storm following a seizure. However, it remains unexplored whether EV therapy can reduce the occurrence of seizures. Current studies primarily utilize MSCs as the EV source, with little exploration of other cell sources such as neurons and glia. Additionally, there is a lack of research on the molecular pathways of EVs specific to epilepsy treatment and their effects on electrophysiological properties. With limited investigations on the application of EVs in epilepsy, more studies testing EVs in epileptic models are needed to enhance our understanding of the pathological and therapeutic roles of EVs in this disease.

Alzheimer’s disease (AD)

In the era of an aging society, AD has been routinely associated with dementia in the elderly [253]. AD pathology manifests as extracellular accumulation of amyloid β (Aβ) plaques and intracellular neurofibrillary tau tangles in cortical and limbic areas [254]. Aβ peptides are phagocytosed by microglia, subsequently triggering immune neuroinflammation. EVs are closely implicated in AD pathogenesis. Microglia spread tau protein through exosomes [85]. Additionally, EVs mediate systemic inflammation and multi-organ dysfunction in AD, such as osteoporosis and cardiovascular diseases [255, 256]. Inhibition of EV biogenesis can reduce Aβ and tau accumulation, subsequently delaying disease progression [257]. Inhibiting ceramide-dependent exosome formation with sphingomyelinase (SMase) silencing improves cognitive function [85, 258, 259]. Similarly, GW4869, an exosome synthesis inhibitor, alleviates neurological deficits in AD [260]. Inhibiting P2X purinoceptor 7 (P2RX7), an ATP-gated cation channel important for microglia’s exosome release, improves memory in animal models [261].

EVs tackle multiple aspects of AD pathogenesis [262]. EVs themselves act therapeutically, functioning as a Trojan horse for Aβ accumulation. Exogenous exosomes markedly reduce Aβ levels in mouse models by binding Aβ to glycosphingolipids (GSLs) and subsequently taken up by microglia [168, 263]. EVs have excellent targeting ability. MSC-EVs are specifically taken up by neurons in pathological regions, suggesting inflammation-driven uptake [264]. However, other studies conversely report that the majority of EVs are internalized by microglia and astrocytes [154]. MSCs are the most popular source of EVs in AD therapy. The proposed mechanisms of MSC-EVs include activation of autophagy through the catalase enzyme, Nrf2 signaling pathway, miR-146a-inhibited NF-κB pathway, and miR-223 targeting the PTEN-PI3K/Akt pathway [155, 156, 159, 265]. Cerebrospinal fluid (CSF) exchange therapy using artificial CSF enriched with MSCs promotes neurogenesis and decreases gliosis in the hippocampus [266]. ASC-EVs similarly decrease Aβ accumulation, neuronal apoptosis, and energy consumption activated by glutamate [161,162,163], even more effective than MSC-EVs [267]. EVs from other sources, such as NPSCs, neurons, CSF, and human brain microvascular endothelial cells (HBMVECs), also promote neuronal restoration and cognitive recovery [164,165,166,167,168,169].

Optimized environments affect the therapeutic properties of EVs. A 3D graphene scaffold produces exosomes that more effectively reduce Aβ production [160]. Hypoxia preconditioning decreases pro-inflammatory miR-770-3p and promotes M2 microglia polarization. Similarly, pretreatment with TNF-α and IFN-γ decreases microglia activation and promotes neurite outgrowth [262].

AD is a significant model of neurodegeneration caused by the accumulation of toxic proteins. EV therapy acts as a scavenger for misfolded proteins, an immunomodulator, and a promoter of neurogenesis. The targeting properties of EVs are crucial for precise treatment. However, there are still conflicting reports on the main targets of EVs, and the mechanisms by which EVs target specific cell types or areas of inflammation remain unclear. EVs from different sources may target differently; for example, EVs from astrocytes may have more specificity to neurons than those from MSCs. The extent of EV accumulation in other tissues should also be explored for safety, as EV uptake into normal brain parenchyma may potentially overstimulate proliferation, leading to tumorigenesis. Given the lack of effective treatments for AD, incorporating EV therapy into conventional medication regimens may improve outcomes. Further research is warranted to fully unravel the efficacy and safety profiles and mechanisms underlying EV treatment in AD.

Parkinson’s disease (PD)

PD corresponds to the second most rampant neurodegenerative disorder behind AD [268, 269]. PD is marked by degeneration of dopaminergic neurons in the substantia nigra within the midbrain. The pathological feature of PD involves the aberrant accrual of α-synuclein, which forms intracellular inclusions known as Lewy bodies. Neuroinflammation plays a crucial role in the pathogenesis of PD [270], and EVs are closely involved in PD pathogenesis [271]. Similar to AD, EVs are implicated in α-synuclein propagation [82]. Leucine-rich repeat serine/threonine kinase 2 (LRRK2), a mutated protein in monogenic PD, is also released through exosomes [83]. Additionally, the prion protein, a glycosylphosphatidylinositol-anchored membrane protein, is involved in α-synuclein transmission through EVs [272, 273].

EVs show potential as a disease-modifying treatment for PD. MSC-derived exosomes can traverse the BBB and exert neuroprotective effects by promoting cell proliferation and inhibiting apoptosis through autophagy induction [170]. Exosomes derived from the dental pulp of human exfoliated deciduous teeth (SHEDs) reduce apoptosis, whereas MVs from the same cells do not provide therapeutic benefits [274]. Exosomes normalize tyrosine hydroxylase expression [171]. Moreover, miR-200a-3p-enriched EVs isolated from healthy astrocytes reduce the expression of mitogen-activated protein kinase kinase 4 (MKK4), a key kinase in the c-Jun N-terminal kinase cell death pathway [172].

PD shares a similar pathogenesis with AD, characterized by the propagation of toxic proteins leading to neuroinflammation. EV therapy can improve PD by promoting neurogenesis. However, the effects of EVs on the primary toxic proteins in PD, such as α-synuclein and LRRK2, are still not fully explored. Blocking exosome release can reduce the propagation of α-synuclein and LRRK2, but the current techniques lack specificity, raising concerns about potential complications. Exosomes and MVs have different effects on PD, with only exosomes exhibiting therapeutic outcomes, emphasizing the importance of EV characterization and selection. Additionally, there is a limited variety of EV sources and modifications in PD therapy compared to other diseases, despite PD being a common cause of dementia. Further modifications of EV sources that cater to targeting the disease hallmark of dopaminergic depletion and degenerative processes (e.g., α-synuclein accumulation) may improve the efficacy of EV-based therapies for PD.

Amyotrophic lateral sclerosis (ALS)

ALS exhibits a rapid demise of motor neurons in the brain, spinal cord, and peripheral regions, leading to muscle weakness, atrophy, and cognitive impairment [275, 276]. The pathogenesis of ALS involves multiple factors, including genetic mutations in genes like C9orf72, SOD1, TARDBP, and FUS, which result in toxic protein aggregation and impaired RNA processing. Neuroinflammation plays a crucial role, with initial protective responses becoming neurotoxic over time, exacerbated by dysfunctional regulatory T cells (Tregs). Additionally, oxidative stress and mitochondrial dysfunction contribute to cellular damage, while glutamate excitotoxicity further accelerates neuronal death. Despite advancements in understanding these mechanisms, effective therapies remain elusive.

There are only a few studies on EV therapy for ALS [173, 277]. EVs isolated from ASCs can protect SOD1-mutated neurons from oxidative stress and normalize mitochondrial function [175, 176, 278]. MSC-derived EVs were effectively taken up by mouse BECs and restored BBB integrity. MSC-exosomes also promoted neurite growth and upregulated antioxidant and anti-inflammatory genes [173, 174]. In a pilot trial in humans using allogeneic stem cell-derived exosomes, the patient showed signs of stabilization in motor function and respiratory capacity during the infusion period, but deterioration occurred after a pause in treatment [279]. Despite these transient benefits, the patient eventually experienced acute respiratory failure and passed away. Continuous administration may be necessary to maintain benefits.

ALS is a neurodegenerative disorder with a complex pathogenesis. The effects of EV therapy in ALS are primarily focused on reducing oxidative stress and inflammation. However, the sources of EVs are currently limited to MSCs and ASCs. Existing studies are insufficient to fully elucidate the mechanisms of EV therapy in ALS. Pilot studies in humans suggest that intermittent administration of therapeutic EVs may be inadequate for the severe stage. Thus, finding the optimal EV regimen, including dosage and frequency, is needed to enhance therapeutic outcomes of EVs in ALS. Elucidating different EV sources and investigating their specific mechanisms of action may also enhance the efficacy and safety of EV therapy for ALS.

Multiple sclerosis (MS)

MS displays a chronic inflammation coincident with demyelination of the CNS as evidenced by multifocal zones of inflammatory response, leading to neuronal cell death and nerve demyelination [280, 281]. The pathogenesis of MS involves a complex interplay between genetic predisposition and environmental factors, such as exposure to infectious agents, vitamin deficiencies, and smoking. Inflammation leads to oligodendrocyte death and impaired myelin repair. Oxidative stress driven by microglial activation and mitochondrial injury contributes to demyelination and neurodegeneration. Age-related iron accumulation and mitochondrial gene deletions further amplify these effects, particularly in progressive MS. Although current treatments focus on anti-inflammatory and immunomodulatory drugs, they are insufficient to halt neurodegeneration, necessitating the exploration of novel therapeutic strategies.

EVs play a critical role in the neuroinflammation underlying MS. MSCs are widely used as a source of EVs [282]. EVs isolated from MSCs shift microglial polarization towards the M2 phenotype, increasing Tregs and IL-10 [178, 179]. EVs isolated from human periodontal ligament stem cells suppress inflammation and apoptosis via CD90-inducing IL-10 production [180]. EVs derived from microglia co-cultured with immunosuppressive MSCs promote oligodendrocyte progenitor cell recruitment and differentiation with lipid cargo [181]. Interestingly, EVs released from pro-inflammatory microglia interfere with remyelination only when co-cultured with astrocytes, implying that astrocytes may mediate oligodendrocyte toxicity.

EVs are used as immunomodulation therapy in MS models, primarily sourced from MSCs and microglia. To implement EV therapy more effectively, the molecular mechanisms involved in autoimmune-induced inflammation need further elucidation. EVs from immune cells, such as anti-inflammatory Tregs or M2 microglia/macrophages, may more potently target the pathogenesis of MS. Additionally, EV therapy should focus on other aspects of pathogenesis, such as neuronal degeneration and demyelination. Since MS is a systemic disease, the effects of EVs on other organs should also be explored to ensure comprehensive treatment and safety. By understanding these mechanisms and tailoring the EVs to sequester the complex pathogenesis of MS may reveal the optimal treatment regimen of EVs for this autoimmune disease that compromises nerve cells in both brain and spinal cord.

Huntington’s disease (HD)

HD is marked by a progressive degeneration of basal ganglia neurons manifesting with behavioral and psychiatric abnormalities [283]. HD is inherited in an autosomal dominant pattern, caused by a mutation in the huntingtin gene (HTT) [284]. An expanded CAG trinucleotide repeats in the HTT gene results in an abnormal huntingtin protein, known as mutant huntingtin (mHTT). The number of CAG repeats directly correlates with the disease’s severity [285]. Accumulation of mHTT in neurons leads to cellular dysfunction, mitochondrial dysfunction, apoptosis, excitotoxicity, and altered gene expression, especially in the striatum and cortex [286]. Similar to other toxic protein propagations, there is evidence that mHTT is transferred by EVs [84, 287,288,289].

EVs from ASCs and NPSCs effectively restore mitochondrial function, decrease N-terminal cleaved mHTT, and suppress apoptosis [182,183,184]. EVs isolated from human dermal fibroblasts also recover GABAergic synapses and transmission [185, 186]. Moreover, heterogeneous parasymbiosis in a mouse model showed that blood serum containing therapeutic substances, possibly EVs, can decrease mHTT and neuron degeneration [188]. EVs isolated from human cord blood found that they reduced gliosis, increased antioxidant activity, partially prevented neuronal loss, and effectively improved neuromuscular function [290].

HD is a genetic disorder that currently lacks effective treatments. EV therapy has the potential to reduce mHTT, the main culprit behind neuroinflammation in HD. While there are multiple clinical trials involving cell transplantation for HD, no clinical trials for EV therapy in HD have been conducted yet. Given that mHTT contributes to various aspects of HD pathogenesis, experiments should address multiple domains, including mitochondrial dysfunction, RNA instability, excitotoxicity, and proteolysis impairment. Similar to AD and PD, HD pathogenesis is driven by toxic protein accumulation—in this case, mHTT. Therapeutic EVs have been shown to reduce N-terminal cleavage of mHTT, but whether they can act as scavengers for mHTT remains to be explored. Further research is needed to investigate the direct effects of EVs on HD pathogenesis specifically on reducing mHTT and alleviating its downstream degenerative symptoms.

Modification and engineering of EVs

Beyond conventional cell-derived EVs, advanced engineering techniques are extensively explored across various stem cell types. In the realm of EV content selection, the biological components of cell-derived EVs can be modulated through culture preconditioning—such as hypoxia or cytokine supplementation—to enhance their therapeutic efficacy, although the resulting outcomes often lack homogeneity [291]. To incorporate small molecules, RNA, and genetic editing tools into EVs, two primary methods of cargo loading are employed: endogenous and exogenous [292,293,294,295]. Endogenous loading involves the genetic manipulation of parent cells to ensure that the secreted EVs inherently carry the desired molecules. Conversely, exogenous loading introduces cargo directly onto or into the EV membrane through techniques such as electroporation, ultrasound, extrusion, freeze–thaw cycles, chemical treatments, and mechanical stirring. This method, however, faces challenges related to content volume control, membrane disruption, altered surface electrical charge, and dysfunctional surface ligands, which impair uptake. Targeting EVs represents another critical area of investigation, typically achieved through membrane fusion, chemical modification, and the genetic engineering of membrane peptides [294, 296]. Such modifications enhance fusion efficiency, colloidal stability, and the half-life of EVs in the bloodstream while reducing immunogenicity [297, 298]. Targeting strategies also improve the precision of EV delivery; for instance, Lamp2b-expressing EVs significantly increase uptake in neurons, microglia, and oligodendrocytes within the brain [299]. The engineering of EVs thus emerges as a promising approach for large-scale production and versatile drug delivery platforms. Nonetheless, challenges remain in ensuring reproducibility, safety, and regulatory protocols. With ongoing technological advancements, synthetic EVs hold the potential to become a pivotal component of personalized medicine.

Conclusion

EVs are crucial mediators of cell-to-cell communication, playing roles in nearly all physiological and pathological processes. EV therapy addresses multiple aspects of neurological diseases, including neuroinflammation, mitochondrial dysfunction, apoptosis, and BBB leakage. Compared to stem cell therapy, EVs are safer and easier to handle, making them a promising alternative for therapeutic interventions.

Despite the rapid growth in the EV field, much remains to be studied. There are numerous EV classes and subclasses yet to be fully characterized. The heterogeneity within EV classes leads to variability in their effects. Studies isolating EVs from the same cell line report different cargo and mechanisms of action. The effects of EV subpopulations are also not fully understood, with research predominantly focused on exosomes. The roles of MVs are still controversial, as they can be either therapeutic or pathogenic depending on their source. MVs and other large EVs may be worth exploring further since they can deliver more cargo. Characterization and purification of EVs are crucial for clinical application. To control the effects of EVs, production methods need to be strictly replicable to avoid heterogeneity, with specific culture and modulation techniques. EVs are involved in multiple pathological processes, such as inflammation, tumorigenesis, and toxic protein spreading. Blocking these pathological EVs is challenging due to a lack of specificity, necessitating more precise techniques for targeting them. Multiple sources of EVs have been studied, but not thoroughly across all domains of treatment. Some cell sources may be better suited for certain roles; for example, stem cells for neuroregeneration, glia for immunomodulation, and endothelial cells for angiogenesis. The targeting ability of EVs is another area that has not been extensively explored. Optimal EV preconditioning, administration regimens, and safety profiles also require further investigation.

A better understanding of EV subtypes and their specific roles will mark a significant milestone in medicine, leading to safer, more effective and disease-tailored EV therapy for a variety of neurological disorders.

Availability of data and materials

Not applicable.

Abbreviations

AD:

Alzheimer’s disease

AGO2:

Argonaute 2

ALS:

Amyotrophic lateral sclerosis

ApoD:

Apo-lipoprotein D

AQP-4:

Aquaporin-4

ARMMs:

Arrestin domain-containing protein 1-mediated microvesicles

ARRDC1:

Arrestin domain-containing protein 1

ASCs:

Adipose-derived stem cells

Aβ:

Amyloid β

BDNF:

Brain-derived neurotrophic factor

BBB:

Blood–brain barrier

BECs:

Brain endothelial cells

Cav-1:

Caveolin-1

CK18:

Cytokeratin 18

CNS:

Central nervous system

COX-2:

Cyclooxygenase-2

CSF:

Cerebrospinal fluid

ESCRT:

Endosomal sorting complexes required for transport

EVs:

Extracellular vesicles

eef2k:

Eukaryotic elongation factor 2 kinase

FGF-1:

Fibroblast growth factor 1

GDNF:

Glial cell line-derived neurotrophic factor

GSLs:

Glycosphingolipids

HBMVECs:

Human brain microvascular endothelial cells

HD:

Huntington’s disease

HIF-1:

Hypoxia-inducible factor-1

HTT:

Huntingtin gene

IFN:

Interferon

IGF-1:

Insulin-like growth factor 1

IL:

Interleukin

ILVs:

Intraluminal vesicles

INs:

Interneurons

iPSCs:

Induced pluripotent stem cells

I-BAR:

Bin–Amphiphysin–Rvs

LCN2:

Lipocalin 2

LRRK2:

Leucine-rich repeat serine/threonine kinase 2

MGE:

Medial ganglionic eminence

MALAT1:

Metastasis-associated lung adenocarcinoma transcript 1

miRNA:

MicroRNA

MKK4:

Mitogen-activated protein kinase kinase 4

MVBs:

Multivesicular bodies

MVs:

Microvesicles

mHTT:

Mutant huntingtin

MSCs:

Mesenchymal stem cells

MS:

Multiple sclerosis

NET:

Neutrophil extracellular trap

NGF:

Nerve growth factor

NMDA:

N-Methyl-d-aspartate

NPSCs:

Neural stem cell/progenitor cell

NSCs:

Neural stem cells

OECs:

Olfactory ensheathing cells

OPC:

Oligodendrocyte progenitor cell

PD:

Parkinson’s disease

P2RX7:

P2X purinoceptor 7

PDCD6IP (also ALIX):

Programmed cell death 6-interacting protein

PNI:

Peripheral nerve injury

PNS:

Peripheral nervous system

ROS:

Reactive oxygen species

SCs:

Schwann cells

SCI:

Spinal cord injury

SE:

Status epilepticus

SHEDs:

Dental pulp of human exfoliated deciduous teeth

SRSF2:

Splice factor serine-arginine-rich splice factor 2

STAU1:

Staufen homolog 1

TBI:

Traumatic brain injury

TLR4:

Toll-like receptor-4

TNSC6A (also GW182):

Trinucleotide repeat-containing gene 6A protein

TNF-α:

Tumor necrosis factor alpha

Tregs:

Regulatory T cells

TSG101:

Tumor susceptibility gene 101 protein

TSPAN4:

Tetraspanin-4

TRPM7:

Transient receptor potential melastatin 7

VEGF:

Vascular endothelial growth factor

References

  1. El Andaloussi S, Mäger I, Breakefield XO, Wood MJA. Extracellular vesicles: biology and emerging therapeutic opportunities. Nat Rev Drug Discov. 2013;12(5):347–57.

    Article  PubMed  Google Scholar 

  2. Simonides Immanuel van de W, Fleur Michelle M, Joost Petrus Gerardus S, Pieter V. Extracellular vesicle heterogeneity and its impact for regenerative medicine applications. Pharmacolog Rev. 2023;75(5):1043.

    Article  Google Scholar 

  3. Lopez-Verrilli MA, Caviedes A, Cabrera A, Sandoval S, Wyneken U, Khoury M. Mesenchymal stem cell-derived exosomes from different sources selectively promote neuritic outgrowth. Neuroscience. 2016;320:129–39.

    Article  CAS  PubMed  Google Scholar 

  4. Raposo G, Nijman HW, Stoorvogel W, Liejendekker R, Harding CV, Melief CJ, et al. B lymphocytes secrete antigen-presenting vesicles. J Exp Med. 1996;183(3):1161–72.

    Article  CAS  PubMed  Google Scholar 

  5. Ratajczak J, Miekus K, Kucia M, Zhang J, Reca R, Dvorak P, et al. Embryonic stem cell-derived microvesicles reprogram hematopoietic progenitors: evidence for horizontal transfer of mRNA and protein delivery. Leukemia. 2006;20(5):847–56.

    Article  CAS  PubMed  Google Scholar 

  6. Gatti S, Bruno S, Deregibus MC, Sordi A, Cantaluppi V, Tetta C, et al. Microvesicles derived from human adult mesenchymal stem cells protect against ischaemia–reperfusion-induced acute and chronic kidney injury. Nephrol Dial Transplant. 2011;26(5):1474–83.

    Article  CAS  PubMed  Google Scholar 

  7. Tan F, Li X, Wang Z, Li J, Shahzad K, Zheng J. Clinical applications of stem cell-derived exosomes. Signal Transduct Target Thera. 2024;9(1):17.

    Article  Google Scholar 

  8. Zhuang X, Xiang X, Grizzle W, Sun D, Zhang S, Axtell RC, et al. Treatment of brain inflammatory diseases by delivering exosome encapsulated anti-inflammatory drugs from the nasal region to the brain. Mol Ther. 2011;19(10):1769–79.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Wahlgren J, De LKT, Brisslert M, Vaziri Sani F, Telemo E, Sunnerhagen P, et al. Plasma exosomes can deliver exogenous short interfering RNA to monocytes and lymphocytes. Nucleic Acids Res. 2012;40(17):e130.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Wang Q, Yu J, Kadungure T, Beyene J, Zhang H, Lu Q. ARMMs as a versatile platform for intracellular delivery of macromolecules. Nat Commun. 2018;9(1):960.

    Article  PubMed  PubMed Central  Google Scholar 

  11. Théry 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(1):1535750.

    Article  PubMed  PubMed Central  Google Scholar 

  12. Ostrowski M, Carmo NB, Krumeich S, Fanget I, Raposo G, Savina A, et al. Rab27a and Rab27b control different steps of the exosome secretion pathway. Nat Cell Biol. 2010;12(1):19–30.

    Article  CAS  PubMed  Google Scholar 

  13. Baietti MF, Zhang Z, Mortier E, Melchior A, Degeest G, Geeraerts A, et al. Syndecan-syntenin-ALIX regulates the biogenesis of exosomes. Nat Cell Biol. 2012;14(7):677–85.

    Article  CAS  PubMed  Google Scholar 

  14. Jeppesen DK, Fenix AM, Franklin JL, Higginbotham JN, Zhang Q, Zimmerman LJ, et al. Reassessment of exosome composition. Cell. 2019;177(2):428-45.e18.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Mathieu M, Névo N, Jouve M, Valenzuela JI, Maurin M, Verweij FJ, et al. Specificities of exosome versus small ectosome secretion revealed by live intracellular tracking of CD63 and CD9. Nat Commun. 2021;12(1):4389.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. D’Angelo G, Raposo G, Nishimura T, Suetsugu S. Protrusion-derived vesicles: new subtype of EVs? Nat Rev Mol Cell Biol. 2023;24(2):81–2.

    Article  PubMed  Google Scholar 

  17. Nabhan JF, Hu R, Oh RS, Cohen SN, Lu Q. Formation and release of arrestin domain-containing protein 1-mediated microvesicles (ARMMs) at plasma membrane by recruitment of TSG101 protein. Proc Natl Acad Sci USA. 2012;109(11):4146–51.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Rauch S, Martin-Serrano J. Multiple interactions between the ESCRT machinery and arrestin-related proteins: Implications for PPXY-dependent budding. J Virol. 2011;85(7):3546–56.

    Article  CAS  PubMed  Google Scholar 

  19. Sun M, Xue X, Li L, Xu D, Li S, Li SC, et al. Ectosome biogenesis and release processes observed by using live-cell dynamic imaging in mammalian glial cells. Quant Imaging Med Surg. 2021;11(11):4604–16.

    Article  PubMed  PubMed Central  Google Scholar 

  20. Matsui T, Osaki F, Hiragi S, Sakamaki Y, Fukuda M. ALIX and ceramide differentially control polarized small extracellular vesicle release from epithelial cells. EMBO Rep. 2021;22(5):e51475.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Kowal J, Arras G, Colombo M, Jouve M, Morath JP, Primdal-Bengtson B, et al. Proteomic comparison defines novel markers to characterize heterogeneous populations of extracellular vesicle subtypes. Proc Nat Acad Sci. 2016;113(8):968.

    Article  Google Scholar 

  22. Minciacchi VR, You S, Spinelli C, Morley S, Zandian M, Aspuria P-J, et al. Large oncosomes contain distinct protein cargo and represent a separate functional class of tumor-derived extracellular vesicles. Oncotarget. 2015;6(13):11327.

    Article  PubMed  PubMed Central  Google Scholar 

  23. Ma L, Li Y, Peng J, Wu D, Zhao X, Cui Y, et al. Discovery of the migrasome, an organelle mediating release of cytoplasmic contents during cell migration. Cell Res. 2015;25(1):24–38.

    Article  CAS  PubMed  Google Scholar 

  24. Zhao X, Lei Y, Zheng J, Peng J, Li Y, Yu L, et al. Identification of markers for migrasome detection. Cell Discov. 2019;5:27.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Huang Y, Zucker B, Zhang S, Elias S, Zhu Y, Chen H, et al. Migrasome formation is mediated by assembly of micron-scale tetraspanin macrodomains. Nat Cell Biol. 2019;21(8):991–1002.

    Article  CAS  PubMed  Google Scholar 

  26. Rai A, Greening DW, Xu R, Chen M, Suwakulsiri W, Simpson RJ. Secreted midbody remnants are a class of extracellular vesicles molecularly distinct from exosomes and microparticles. Commun Biol. 2021;4(1):400.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Melentijevic I, Toth ML, Arnold ML, Guasp RJ, Harinath G, Nguyen KC, et al. C. elegans neurons jettison protein aggregates and mitochondria under neurotoxic stress. Nature. 2017;542(7641):367–71.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Jeppesen DK, Zhang Q, Franklin JL, Coffey RJ. Extracellular vesicles and nanoparticles: emerging complexities. Trends Cell Biol. 2023;33(8):667–81.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Muralidharan-Chari V, Clancy J, Plou C, Romao M, Chavrier P, Raposo G, et al. ARF6-regulated shedding of tumor cell-derived plasma membrane microvesicles. Curr Biol. 2009;19(22):1875–85.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Gladnikoff M, Shimoni E, Gov NS, Rousso I. Retroviral assembly and budding occur through an actin-driven mechanism. Biophys J. 2009;97(9):2419–28.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Piper RC, Katzmann DJ. Biogenesis and function of multivesicular bodies. Annu Rev Cell Dev Biol. 2007;23:519–47.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Hugel B, Martínez MC, Kunzelmann C, Freyssinet J-M. Membrane microparticles: two sides of the coin. Physiology. 2005;20(1):22–7.

    Article  CAS  PubMed  Google Scholar 

  33. Théry C, Zitvogel L, Amigorena S. Exosomes: composition, biogenesis and function. Nat Rev Immunol. 2002;2(8):569–79.

    Article  PubMed  Google Scholar 

  34. Akers JC, Gonda D, Kim R, Carter BS, Chen CC. Biogenesis of extracellular vesicles (EV): exosomes, microvesicles, retrovirus-like vesicles, and apoptotic bodies. J Neurooncol. 2013;113(1):1–11.

    Article  PubMed  PubMed Central  Google Scholar 

  35. Ciarra A, Kreß S, Weber V, Egger D, Kasper C. Heterogeneity of mesenchymal stem cell-derived extracellular vesicles is highly impacted by the tissue/cell source and culture conditions. Cell Biosci. 2022;12:1–15.

    Google Scholar 

  36. Patel DB, Gray KM, Santharam Y, Lamichhane TN, Stroka KM, Jay SM. Impact of cell culture parameters on production and vascularization bioactivity of mesenchymal stem cell-derived extracellular vesicles. Bioeng Transl Med. 2017;2(2):170–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Chen Q, Takada R, Noda C, Kobayashi S, Takada S. Different populations of Wnt-containing vesicles are individually released from polarized epithelial cells. Sci Rep. 2016;6(1):35562.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Kim JY, Rhim W-K, Seo HJ, Lee JY, Park CG, Han DK. Comparative analysis of MSC-derived exosomes depending on cell culture media for regenerative bioactivity. Tissue Eng Regenerat Med. 2021;18(3):355–67.

    Article  CAS  Google Scholar 

  39. Kucharzewska P, Belting M. Emerging roles of extracellular vesicles in the adaptive response of tumour cells to microenvironmental stress. J Extracell Vesicles. 2013;2(1):20304.

    Article  Google Scholar 

  40. Kilpinen L, Impola U, Sankkila L, Ritamo I, Aatonen M, Kilpinen S, et al. Extracellular membrane vesicles from umbilical cord blood-derived MSC protect against ischemic acute kidney injury, a feature that is lost after inflammatory conditioning. J Extracell Vesicles. 2013;2(1):21927.

    Article  Google Scholar 

  41. Wang J, Pendurthi UR, Rao LVM. Sphingomyelin encrypts tissue factor: ATP-induced activation of A-SMase leads to tissue factor decryption and microvesicle shedding. Blood Adv. 2017;1(13):849–62.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Jaiswal R, Sedger LM. Intercellular vesicular transfer by exosomes, microparticles and oncosomes—implications for cancer biology and treatments. Front Oncol. 2019;9:125.

    Article  PubMed  PubMed Central  Google Scholar 

  43. Kim HY, Kwon S, Um W, Shin S, Kim CH, Park JH, et al. Functional extracellular vesicles for regenerative medicine. Small. 2022;18(36):2106569.

    Article  CAS  Google Scholar 

  44. Klumperman J, Raposo G. The complex ultrastructure of the endolysosomal system. Cold Spring Harb Perspect Biol. 2014;6(10):a016857.

    Article  PubMed  PubMed Central  Google Scholar 

  45. Buschow SI, Nolte-‘t Hoen ENM, Van Niel G, Pols MS, Ten Broeke T, Lauwen M, et al. MHC II in dendritic cells is targeted to lysosomes or T cell-induced exosomes via distinct multivesicular body pathways. Traffic. 2009;10(10):1528–42.

    Article  CAS  PubMed  Google Scholar 

  46. van Niel G, Angelo G, Raposo G. Shedding light on the cell biology of extracellular vesicles. Nat Rev Mol Cell Biol. 2018;19:213.

    Article  PubMed  Google Scholar 

  47. Hromada C, Mühleder S, Grillari J, Redl H, Holnthoner W. Endothelial extracellular vesicles—promises and challenges. Front Physiol. 2017;8:275.

    Article  PubMed  PubMed Central  Google Scholar 

  48. Rezaie J, Ajezi S, Avci ÇB, Karimipour M, Geranmayeh MH, Nourazarian A, et al. Exosomes and their application in biomedical field: difficulties and advantages. Mol Neurobiol. 2018;55(4):3372–93.

    Article  CAS  PubMed  Google Scholar 

  49. Hanson PI, Cashikar A. Multivesicular body morphogenesis. Annu Rev Cell Dev Biol. 2012;28:337–62.

    Article  CAS  PubMed  Google Scholar 

  50. Friand V, David G, Zimmermann P. Syntenin and syndecan in the biogenesis of exosomes. Biol Cell. 2015;107(10):331–41.

    Article  CAS  PubMed  Google Scholar 

  51. Perez-Hernandez D, Gutiérrez-Vázquez C, Jorge I, López-Martín S, Ursa A, Sánchez-Madrid F, et al. The intracellular interactome of tetraspanin-enriched microdomains reveals their function as sorting machineries toward exosomes. J Biol Chem. 2013;288(17):11649–61.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. van Niel G, Charrin S, Simoes S, Romao M, Rochin L, Saftig P, et al. The tetraspanin CD63 regulates ESCRT-independent and -dependent endosomal sorting during melanogenesis. Dev Cell. 2011;21(4):708–21.

    Article  PubMed  PubMed Central  Google Scholar 

  53. Keerthikumar S, Gangoda L, Liem M, Fonseka P, Atukorala I, Ozcitti C, et al. Proteogenomic analysis reveals exosomes are more oncogenic than ectosomes. Oncotarget. 2015;6(17):15375.

    Article  PubMed  PubMed Central  Google Scholar 

  54. de Poret A, Dibsy R, Merida P, Trausch A, Inamdar K, Muriaux D. Extracellular vesicles containing the I-BAR protein IRSp53 are released from the cell plasma membrane in an Arp2/3 dependent manner. Biol Cell. 2022;114(10):259–75.

    Article  PubMed  Google Scholar 

  55. Nishimura T, Oyama T, Hu HT, Fujioka T, Hanawa-Suetsugu K, Ikeda K, et al. Filopodium-derived vesicles produced by MIM enhance the migration of recipient cells. Dev Cell. 2021;56(6):842-59.e8.

    Article  CAS  PubMed  Google Scholar 

  56. Hurbain I, Macé A-S, Romao M, Prince E, Sengmanivong L, Ruel L, et al. Microvilli-derived extracellular vesicles carry Hedgehog morphogenic signals for Drosophila wing imaginal disc development. Curr Biol. 2022;32(2):361-73.e6.

    Article  CAS  PubMed  Google Scholar 

  57. Thamm K, Šimaitė D, Karbanová J, Bermúdez V, Reichert D, Morgenstern A, et al. Prominin-1 (CD133) modulates the architecture and dynamics of microvilli. Traffic. 2019;20(1):39–60.

    Article  CAS  PubMed  Google Scholar 

  58. Inamdar K, Feng-Ching T, Dibsy R, de Poret A, Manzi J, Merida P, et al. Full assembly of HIV-1 particles requires assistance of the membrane curvature factor IRSp53. Elife. 2021;10:e67321.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Willms E, Johansson HJ, Mäger I, Lee Y, Blomberg KEM, Sadik M, et al. Cells release subpopulations of exosomes with distinct molecular and biological properties. Sci Rep. 2016;6(1):22519.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Piccin A, Murphy WG, Smith OP. Circulating microparticles: pathophysiology and clinical implications. Blood Rev. 2007;21(3):157–71.

    Article  CAS  PubMed  Google Scholar 

  61. Moskovich O, Fishelson Z. Live cell imaging of outward and inward vesiculation induced by the complement c5b-9 complex. J Biol Chem. 2007;282(41):29977–86.

    Article  CAS  PubMed  Google Scholar 

  62. Pizzirani C, Ferrari D, Chiozzi P, Adinolfi E, Sandonà D, Savaglio E, et al. Stimulation of P2 receptors causes release of IL-1beta-loaded microvesicles from human dendritic cells. Blood. 2007;109(9):3856–64.

    Article  CAS  PubMed  Google Scholar 

  63. Kahner BN, Dorsam RT, Kunapuli SP. Role of P2Y receptor subtypes in platelet-derived microparticle generation. Front Biosci. 2008;13:433–9.

    Article  CAS  PubMed  Google Scholar 

  64. Sedgwick AE, D’Souza-Schorey C. The biology of extracellular microvesicles. Traffic. 2018;19(5):319–27.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Lakkaraju A, Rodriguez-Boulan E. Itinerant exosomes: emerging roles in cell and tissue polarity. Trends Cell Biol. 2008;18(5):199–209.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Cocucci E, Racchetti G, Meldolesi J. Shedding microvesicles: artefacts no more. Trends Cell Biol. 2009;19(2):43–51.

    Article  CAS  PubMed  Google Scholar 

  67. Kerr JF, Wyllie AH, Currie AR. Apoptosis: a basic biological phenomenon with wide-ranging implications in tissue kinetics. Br J Cancer. 1972;26(4):239–57.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Ihara T, Yamamoto T, Sugamata M, Okumura H, Ueno Y. The process of ultrastructural changes from nuclei to apoptotic body. Virchows Arch. 1998;433(5):443–7.

    Article  CAS  PubMed  Google Scholar 

  69. Taylor RC, Cullen SP, Martin SJ. Apoptosis: controlled demolition at the cellular level. Nat Rev Mol Cell Biol. 2008;9(3):231–41.

    Article  CAS  PubMed  Google Scholar 

  70. Coleman ML, Sahai EA, Yeo M, Bosch M, Dewar A, Olson MF. Membrane blebbing during apoptosis results from caspase-mediated activation of ROCK I. Nat Cell Biol. 2001;3:339.

    Article  CAS  PubMed  Google Scholar 

  71. Atkin-Smith GK, Tixeira R, Paone S, Mathivanan S, Collins C, Liem M, et al. A novel mechanism of generating extracellular vesicles during apoptosis via a beads-on-a-string membrane structure. Nat Commun. 2015;6(1):7439.

    Article  PubMed  Google Scholar 

  72. Sebbagh M, Renvoizé C, Hamelin J, Riché N, Bertoglio J, Bréard J. Caspase-3-mediated cleavage of ROCK I induces MLC phosphorylation and apoptotic membrane blebbing. Nat Cell Biol. 2001;3(4):346–52.

    Article  CAS  PubMed  Google Scholar 

  73. Vandivier RW, Ogden CA, Fadok VA, Hoffmann PR, Brown KK, Botto M, et al. Role of surfactant proteins A, D, and C1q in the clearance of apoptotic cells in vivo and in vitro: Calreticulin and CD91 as a common collectin receptor complex. J Immunol. 2002;169(7):3978–86.

    Article  CAS  PubMed  Google Scholar 

  74. Friedl P, Vischer P, Freyberg MA. The role of thrombospondin-1 in apoptosis. Cell Mol Life Sci. 2002;59(8):1347–57.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Erwig LP, Henson PM. Clearance of apoptotic cells by phagocytes. Cell Death Differ. 2008;15(2):243–50.

    Article  CAS  PubMed  Google Scholar 

  76. Samos J, García-Olmo DC, Picazo MG, Rubio-Vitaller A, García-Olmo D. Circulating nucleic acids in plasma/serum and tumor progression: are apoptotic bodies involved? An experimental study in a rat cancer model. Ann N Y Acad Sci. 2006;1075:165–73.

    Article  CAS  PubMed  Google Scholar 

  77. Bergsmedh A, Szeles A, Henriksson M, Bratt A, Folkman MJ, Spetz AL, et al. Horizontal transfer of oncogenes by uptake of apoptotic bodies. Proc Natl Acad Sci USA. 2001;98(11):6407–11.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Minciacchi VR, Spinelli C, Reis-Sobreiro M, Cavallini L, You S, Zandian M, et al. MYC mediates large oncosome-induced fibroblast reprogramming in prostate cancer. Can Res. 2017;77(9):2306–17.

    Article  CAS  Google Scholar 

  79. Crowell EF, Gaffuri A-L, Gayraud-Morel B, Tajbakhsh S, Echard A. Engulfment of the midbody remnant after cytokinesis in mammalian cells. J Cell Sci. 2014;127(17):3840–51.

    CAS  PubMed  Google Scholar 

  80. Kuo TC, Chen CT, Baron D, Onder TT, Loewer S, Almeida S, et al. Midbody accumulation through evasion of autophagy contributes to cellular reprogramming and tumorigenicity. Nat Cell Biol. 2011;13(10):1214–23.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Nicolás-Ávila JA, Lechuga-Vieco AV, Esteban-Martínez L, Sánchez-Díaz M, Díaz-García E, Santiago DJ, et al. A network of macrophages supports mitochondrial homeostasis in the heart. Cell. 2020;183(1):94-109.e23.

    Article  PubMed  Google Scholar 

  82. Grey M, Dunning CJ, Gaspar R, Grey C, Brundin P, Sparr E, et al. Acceleration of α-synuclein aggregation by exosomes. J Biol Chem. 2015;290(5):2969–82.

    Article  CAS  PubMed  Google Scholar 

  83. Fraser KB, Moehle MS, Daher JPL, Webber PJ, Williams JY, Stewart CA, et al. LRRK2 secretion in exosomes is regulated by 14-3-3. Hum Mol Genet. 2013;22(24):4988–5000.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  84. Monsellier E, Bousset L, Melki R. α-Synuclein and huntingtin exon 1 amyloid fibrils bind laterally to the cellular membrane. Sci Rep. 2016;6:19180.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  85. Asai H, Ikezu S, Tsunoda S, Medalla M, Luebke J, Haydar T, et al. Depletion of microglia and inhibition of exosome synthesis halt tau propagation. Nat Neurosci. 2015;18(11):1584–93.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. Berckmans RJ, Nieuwland R, Kraan MC, Schaap MCL, Pots D, Smeets TJM, et al. Synovial microparticles from arthritic patients modulate chemokine and cytokine release by synoviocytes. Arthrit Res Thera. 2005;7(3):R536.

    Article  CAS  Google Scholar 

  87. Gasser O, Schifferli JA. Activated polymorphonuclear neutrophils disseminate anti-inflammatory microparticles by ectocytosis. Blood. 2004;104(8):2543–8.

    Article  CAS  PubMed  Google Scholar 

  88. Lambertsen KL, Biber K, Finsen B. Inflammatory cytokines in experimental and human stroke. J Cereb Blood Flow Metab. 2012;32(9):1677–98.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  89. Peinado H, Alečković M, Lavotshkin S, Matei I, Costa-Silva B, Moreno-Bueno G, et al. Melanoma exosomes educate bone marrow progenitor cells toward a pro-metastatic phenotype through MET. Nat Med. 2012;18(6):883–91.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  90. Davila M, Amirkhosravi A, Coll E, Desai H, Robles L, Colon J, et al. Tissue factor-bearing microparticles derived from tumor cells: impact on coagulation activation. J Thromb Haemost. 2008;6(9):1517–24.

    Article  CAS  PubMed  Google Scholar 

  91. Gnecchi M, He H, Noiseux N, Liang OD, Zhang L, Morello F, et al. Evidence supporting paracrine hypothesis for Akt-modified mesenchymal stem cell-mediated cardiac protection and functional improvement. FASEB J. 2006;20(6):661–9.

    Article  CAS  PubMed  Google Scholar 

  92. Gnecchi M, He H, Liang OD, Melo LG, Morello F, Mu H, et al. Paracrine action accounts for marked protection of ischemic heart by Akt-modified mesenchymal stem cells. Nat Med. 2005;11:367.

    Article  CAS  PubMed  Google Scholar 

  93. Long X, Yao X, Jiang Q, Yang Y, He X, Tian W, et al. Astrocyte-derived exosomes enriched with miR-873a-5p inhibit neuroinflammation via microglia phenotype modulation after traumatic brain injury. J Neuroinflammation. 2020;17(1):89.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  94. Wang X, Ye L, Zhang K, Gao L, Xiao J, Zhang Y. Small extracellular vesicles released from miR-211-5p-overexpressed bone marrow mesenchymal stem cells ameliorate spinal cord injuries in rats. Eneuro. 2024;11(2):0361–232023.

    Article  Google Scholar 

  95. Deng Y, Chen D, Gao F, Lv H, Zhang G, Sun X, et al. Exosomes derived from microRNA-138-5p-overexpressing bone marrow-derived mesenchymal stem cells confer neuroprotection to astrocytes following ischemic stroke via inhibition of LCN2. J Biol Eng. 2019;13(1):71.

    Article  PubMed  PubMed Central  Google Scholar 

  96. Xiao B, Chai Y, Lv S, Ye M, Wu M, Xie L, et al. Endothelial cell-derived exosomes protect SH-SY5Y nerve cells against ischemia/reperfusion injury. Int J Mol Med. 2017;40(4):1201–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  97. El Bassit G, Patel RS, Carter G, Shibu V, Patel AA, Song S, et al. MALAT1 in human adipose stem cells modulates survival and alternative splicing of PKCδII in HT22 cells. Endocrinology. 2017;158(1):183–95.

    PubMed  Google Scholar 

  98. Xiao Y, Geng F, Wang G, Li X, Zhu J, Zhu W. Bone marrow-derived mesenchymal stem cells-derived exosomes prevent oligodendrocyte apoptosis through exosomal miR-134 by targeting caspase-8. J Cell Biochem. 2019;120(2):2109–18.

    Article  CAS  PubMed  Google Scholar 

  99. Ni H, Yang S, Siaw-Debrah F, Hu J, Wu K, He Z, et al. Exosomes derived from bone mesenchymal stem cells ameliorate early inflammatory responses following traumatic brain injury. Front Neurosci. 2019;13:14.

    Article  PubMed  PubMed Central  Google Scholar 

  100. Dave KM, Stolz DB, Manickam DS. Delivery of mitochondria-containing extracellular vesicles to the BBB for ischemic stroke therapy. Expert Opin Drug Deliv. 2023;20(12):1769–88.

    Article  CAS  PubMed  Google Scholar 

  101. Dave KM, Stolz DB, Venna VR, Quaicoe VA, Maniskas ME, Reynolds MJ, et al. Mitochondria-containing extracellular vesicles (EV) reduce mouse brain infarct sizes and EV/HSP27 protect ischemic brain endothelial cultures. J Control Release. 2023;354:368–93.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  102. Yu Y, Zhou H, Xiong Y, Liu J. Exosomal miR-199a-5p derived from endothelial cells attenuates apoptosis and inflammation in neural cells by inhibiting endoplasmic reticulum stress. Brain Res. 2020;1726:146515.

    Article  CAS  PubMed  Google Scholar 

  103. Lopez-Leal R, Court FA. Schwann cell exosomes mediate neuron–glia communication and enhance axonal regeneration. Cell Mol Neurobiol. 2016;36(3):429–36.

    Article  CAS  PubMed  Google Scholar 

  104. Williams AM, Higgins GA, Bhatti UF, Biesterveld BE, Dekker SE, Kathawate RG, et al. Early treatment with exosomes following traumatic brain injury and hemorrhagic shock in a swine model promotes transcriptional changes associated with neuroprotection. J Trauma Acute Care Surg. 2020;89(3):536–43.

    Article  CAS  PubMed  Google Scholar 

  105. Chen KH, Chen CH, Wallace CG, Yuen CM, Kao GS, Chen YL, et al. Intravenous administration of xenogenic adipose-derived mesenchymal stem cells (ADMSC) and ADMSC-derived exosomes markedly reduced brain infarct volume and preserved neurological function in rat after acute ischemic stroke. Oncotarget. 2016;7(46):74537–56.

    Article  PubMed  PubMed Central  Google Scholar 

  106. Yang Y, Cai Y, Zhang Y, Liu J, Xu Z. Exosomes secreted by adipose-derived stem cells contribute to angiogenesis of brain microvascular endothelial cells following oxygen–glucose deprivation in vitro through microRNA-181b/TRPM7 axis. J Mol Neurosci. 2018;65(1):74–83.

    Article  CAS  PubMed  Google Scholar 

  107. Venkat P, Cui C, Chopp M, Zacharek A, Wang F, Landschoot-Ward J, et al. MiR-126 mediates brain endothelial cell exosome treatment-induced neurorestorative effects after stroke in type 2 diabetes mellitus mice. Stroke. 2019;50(10):2865–74.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  108. Geng W, Tang H, Luo S, Lv Y, Liang D, Kang X, et al. Exosomes from miRNA-126-modified ADSCs promotes functional recovery after stroke in rats by improving neurogenesis and suppressing microglia activation. Am J Transl Res. 2019;11(2):780–92.

    CAS  PubMed  PubMed Central  Google Scholar 

  109. Tian Y, Zhu P, Liu S, Jin Z, Li D, Zhao H, et al. IL-4-polarized BV2 microglia cells promote angiogenesis by secreting exosomes. Adv Clin Exp Med. 2019;28(4):421–9.

    Article  PubMed  Google Scholar 

  110. Xu B, Zhang Y, Du X-F, Li J, Zi H-X, Bu J-W, et al. Neurons secrete miR-132-containing exosomes to regulate brain vascular integrity. Cell Res. 2017;27(7):882–97.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  111. Zhang Q, Deng P, Chen S, Xu H, Zhang Y, Chen H, et al. Electroacupuncture and human iPSC-derived small extracellular vesicles regulate the gut microbiota in ischemic stroke via the brain-gut axis. Front Immunol. 2023;14:1107559.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  112. Tang TT, Wang B, Lv LL, Liu BC. Extracellular vesicle-based nanotherapeutics: emerging frontiers in anti-inflammatory therapy. Theranostics. 2020;10(18):8111–29.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  113. Al-Nedawi K, Meehan B, Kerbel RS, Allison AC, Rak J. Endothelial expression of autocrine VEGF upon the uptake of tumor-derived microvesicles containing oncogenic EGFR. Proc Natl Acad Sci USA. 2009;106(10):3794–9.

    Article  PubMed  PubMed Central  Google Scholar 

  114. Cai Z, Yang F, Yu L, Yu Z, Jiang L, Wang Q, et al. Activated T cell exosomes promote tumor invasion via Fas signaling pathway. J Immunol. 2012;188(12):5954–61.

    Article  CAS  PubMed  Google Scholar 

  115. Lai CP, Breakefield XO. Role of exosomes/microvesicles in the nervous system and use in emerging therapies. Front Physiol. 2012;3:228.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  116. Lee J-Y, Cho J, D’Egidio F, Vignon C, Streefkerk H, de Kalbermatten M, et al. Probing multiple transplant delivery routes of CD+34 stem cells for promoting behavioral and histological benefits in experimental ischemic stroke. Stem Cells Transl Med. 2024;13(2):177–90.

    Article  CAS  PubMed  Google Scholar 

  117. Zhang Y, Zhang Y, Chopp M, Pang H, Chen L, Zhang ZG, et al. Therapeutic role of microRNAs of small extracellular vesicles from human mesenchymal stromal/stem cells in treatment of experimental traumatic brain injury. J Neurotrauma. 2022;40(7–8):758–71.

    Google Scholar 

  118. Deng M, Xiao H, Zhang H, Peng H, Yuan H, Xu Y, et al. Mesenchymal stem cell-derived extracellular vesicles ameliorates hippocampal synaptic impairment after transient global ischemia. Front Cell Neurosci. 2017;11:205.

    Article  PubMed  PubMed Central  Google Scholar 

  119. Gao X, Yang H, Xiao W, Su J, Zhang Y, Wang H, Ni W, Gu Y. Modified exosomal SIRPα variants alleviate white matter injury after intracerebral hemorrhage via microglia/macrophages. Biomater Res. 2022;26(1):67.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  120. Zhao H, Li Y, Chen L, Shen C, Xiao Z, Xu R, Wang J, Luo Y. HucMSCs-derived miR-206-knockdown exosomes contribute to neuroprotection in subarachnoid hemorrhage induced early brain injury by targeting BDNF. Neuroscience. 2019;417:11–23.

    Article  CAS  PubMed  Google Scholar 

  121. Xiong L, Sun L, Zhang Y, Peng J, Yan J, Liu X. Exosomes from bone marrow mesenchymal stem cells can alleviate early brain injury after subarachnoid hemorrhage through miRNA129-5p-HMGB1 pathway. Stem Cells Dev. 2020;29(4):212–21.

    Article  CAS  PubMed  Google Scholar 

  122. Han M, Cao Y, Guo X, Chu X, Li T, Xue H, Xin D, Yuan L, Ke H, Li G, Wang Z. Mesenchymal stem cell-derived extracellular vesicles promote microglial M2 polarization after subarachnoid hemorrhage in rats and involve the AMPK/NF-κB signaling pathway. Biomed Pharmacother. 2021;133: 111048.

    Article  CAS  PubMed  Google Scholar 

  123. Qian Y, Li Q, Chen L, Sun J, Cao K, Mei Z, Lu X. Mesenchymal stem cell-derived extracellular vesicles alleviate M1 microglial activation in brain injury of mice with subarachnoid hemorrhage via microRNA-140-5p delivery. Int J Neuropsychopharmacol. 2022;25(4):328–38.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  124. Yi X, Tang X. Exosomes from miR-19b-3p-modified ADSCs inhibit ferroptosis in intracerebral hemorrhage mice. Front Cell Dev Biol. 2021;9:661317.

    Article  PubMed  PubMed Central  Google Scholar 

  125. Lee HJ, Kim MK, Kim HJ, Kim SU. Human neural stem cells genetically modified to overexpress Akt1 provide neuroprotection and functional improvement in mouse stroke model. PLoS ONE. 2009;4(5):e5586.

    Article  PubMed  PubMed Central  Google Scholar 

  126. Lee HJ, Lim IJ, Lee MC, Kim SU. Human neural stem cells genetically modified to overexpress brain-derived neurotrophic factor promote functional recovery and neuroprotection in a mouse stroke model. J Neurosci Res. 2010;88(15):3282–94.

    Article  CAS  PubMed  Google Scholar 

  127. Lee H, Park I, Kim H, et al. Human neural stem cells overexpressing glial cell line-derived neurotrophic factor in experimental cerebral hemorrhage. Gene Ther. 2009;16(8):1066–76.

    Article  CAS  PubMed  Google Scholar 

  128. Qian Y, Chen B, Sun E, Lu X, Li Z, Wang R, et al. Mesenchymal stem cell-derived extracellular vesicles alleviate brain damage following subarachnoid hemorrhage via the interaction of miR-140-5p and HDAC7. Mol Neurobiol. 2024.

  129. Kodali M, Madhu LN, Reger RL, Milutinovic B, Upadhya R, Gonzalez JJ, et al. Intranasally administered human MSC-derived extracellular vesicles inhibit NLRP3-p38/MAPK signaling after TBI and prevent chronic brain dysfunction. Brain Behav Immun. 2023;108:118–34.

    Article  CAS  PubMed  Google Scholar 

  130. Kim DK, Nishida H, An SY, Shetty AK, Bartosh TJ, Prockop DJ. Chromatographically isolated CD63+CD81+ extracellular vesicles from mesenchymal stromal cells rescue cognitive impairments after TBI. Proc Natl Acad Sci USA. 2016;113(1):170–5.

    Article  CAS  PubMed  Google Scholar 

  131. Gao W, Li F, Liu L, Xu X, Zhang B, Wu Y, et al. Endothelial colony-forming cell-derived exosomes restore blood–brain barrier continuity in mice subjected to traumatic brain injury. Exp Neurol. 2018;307:99–108.

    Article  CAS  PubMed  Google Scholar 

  132. Li D, Huang S, Yin Z, Zhu J, Ge X, Han Z, et al. Increases in miR-124-3p in microglial exosomes confer neuroprotective effects by targeting FIP200-mediated neuronal autophagy following traumatic brain injury. Neurochem Res. 2019;44(8):1903–23.

    Article  CAS  PubMed  Google Scholar 

  133. Yang Y, Ye Y, Kong C, Su X, Zhang X, Bai W, et al. MiR-124 enriched exosomes promoted the M2 polarization of microglia and enhanced hippocampus neurogenesis after traumatic brain injury by inhibiting TLR4 pathway. Neurochem Res. 2019;44(4):811–28.

    Article  CAS  PubMed  Google Scholar 

  134. Ge X, Guo M, Hu T, Li W, Huang S, Yin Z, et al. Increased microglial exosomal miR-124-3p alleviates neurodegeneration and improves cognitive outcome after rmTBI. Mol Ther. 2020;28(2):503–22.

    Article  CAS  PubMed  Google Scholar 

  135. Huang S, Ge X, Yu J, Han Z, Yin Z, Li Y, et al. Increased miR-124-3p in microglial exosomes following traumatic brain injury inhibits neuronal inflammation and contributes to neurite outgrowth via their transfer into neurons. FASEB J. 2018;32(1):512–28.

    Article  CAS  PubMed  Google Scholar 

  136. Yang Z, Liang Z, Rao J, Xie H, Zhou M, Xu X, et al. Hypoxic-preconditioned mesenchymal stem cell-derived small extracellular vesicles promote the recovery of spinal cord injury by affecting the phenotype of astrocytes through the miR-21/JAK2/STAT3 pathway. CNS Neurosci Therapeut. 2024;30(3):e14428.

    Article  CAS  Google Scholar 

  137. Li C, Jiao G, Wu W, Wang H, Ren S, Zhang L, et al. Exosomes from bone marrow mesenchymal stem cells inhibit neuronal apoptosis and promote motor function recovery via the Wnt/β-catenin signaling pathway. Cell Transplant. 2019;28(11):1373–83.

    Article  PubMed  PubMed Central  Google Scholar 

  138. 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;25(4):2406.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  139. Xu J, Ren Z, Niu T, Li S. Epigenetic mechanism of miR-26b-5p-enriched MSCs-EVs attenuates spinal cord injury. Regenerat Thera. 2024;25:35–48.

    Article  CAS  Google Scholar 

  140. Rong Y, Liu W, Lv C, Wang J, Luo Y, Jiang D, et al. Neural stem cell small extracellular vesicle-based delivery of 14-3-3t reduces apoptosis and neuroinflammation following traumatic spinal cord injury by enhancing autophagy by targeting Beclin-1. Aging (Albany NY). 2019;11(18):7723–45.

    Article  CAS  PubMed  Google Scholar 

  141. Mohammed I, Ijaz S, Mokhtari T, Gholaminejhad M, Mahdavipour M, Jameie B, et al. Subventricular zone-derived extracellular vesicles promote functional recovery in rat model of spinal cord injury by inhibition of NLRP3 inflammasome complex formation. Metab Brain Dis. 2020;35(5):809–18.

    Article  CAS  PubMed  Google Scholar 

  142. Zhong D, Cao Y, Li C-J, Li M, Rong Z-J, Jiang L, et al. Neural stem cell-derived exosomes facilitate spinal cord functional recovery after injury by promoting angiogenesis. Exp Biol Med. 2020;245(1):54–65.

    Article  CAS  Google Scholar 

  143. Jiang D, Gong F, Ge X, Lv C, Huang C, Feng S, et al. Neuron-derived exosomes-transmitted miR-124-3p protect traumatically injured spinal cord by suppressing the activation of neurotoxic microglia and astrocytes. J Nanobiotechnol. 2020;18(1):105.

    Article  CAS  Google Scholar 

  144. Bucan V, Vaslaitis D, Peck C-T, Strauß S, Vogt PM, Radtke C. Effect of exosomes from rat adipose-derived mesenchymal stem cells on neurite outgrowth and sciatic nerve regeneration after crush injury. Mol Neurobiol. 2019;56(3):1812–24.

    Article  CAS  PubMed  Google Scholar 

  145. Ma Y, Ge S, Zhang J, Zhou D, Li L, Wang X, et al. Mesenchymal stem cell-derived extracellular vesicles promote nerve regeneration after sciatic nerve crush injury in rats. Int J Clin Exp Pathol. 2017;10(9):10032–9.

    PubMed  PubMed Central  Google Scholar 

  146. Ma Y, Dong L, Zhou D, Li L, Zhang W, Zhen Y, et al. Extracellular vesicles from human umbilical cord mesenchymal stem cells improve nerve regeneration after sciatic nerve transection in rats. J Cell Mol Med. 2019;23(4):2822–35.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  147. López-Leal R, Díaz-Viraqué F, Catalán RJ, Saquel C, Enright A, Iraola G, et al. Schwann cell reprogramming into repair cells increases miRNA-21 expression in exosomes promoting axonal growth. J Cell Sci. 2020;133(12):jcs239004.

    Article  PubMed  Google Scholar 

  148. Lopez-Verrilli MA, Picou F, Court FA. Schwann cell-derived exosomes enhance axonal regeneration in the peripheral nervous system. Glia. 2013;61(11):1795–806.

    Article  PubMed  Google Scholar 

  149. Zhan C, Ma C-b, Yuan H-m, Cao B-y, Zhu J-j. Macrophage-derived microvesicles promote proliferation and migration of Schwann cell on peripheral nerve repair. Biochem Biophys Res Commun. 2015;468(1):343–8.

    Article  CAS  PubMed  Google Scholar 

  150. Xia B, Gao J, Li S, Huang L, Ma T, Zhao L, et al. Extracellular vesicles derived from olfactory ensheathing cells promote peripheral nerve regeneration in rats. Front Cell Neurosci. 2019;13:548.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  151. Yin GN, Shin TY, Ock J, Choi M-J, Limanjaya A, Kwon M-H, et al. Pericyte-derived extracellular vesicles-mimetic nanovesicles improve peripheral nerve regeneration in mouse models of sciatic nerve transection. Int J Mol Med. 2022;49(2):18.

    Article  CAS  PubMed  Google Scholar 

  152. Chai Y, Liu Y, Liu Z, Wei W, Dong Y, Yang C, et al. Study on the role and mechanism of exosomes derived from dental pulp stem cells in promoting regeneration of myelin sheath in rats with sciatic nerve injury. Mol Neurobiol. 2024.

  153. Long Q, Upadhya D, Hattiangady B, Kim D-K, An SY, Shuai B, et al. Intranasal MSC-derived A1-exosomes ease inflammation, and prevent abnormal neurogenesis and memory dysfunction after status epilepticus. Proc Nat Acad Sci. 2017;114(17):e3536.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  154. Bodart-Santos V, de Carvalho LRP, de Godoy MA, Batista AF, Saraiva LM, Lima LG, et al. Extracellular vesicles derived from human Wharton’s jelly mesenchymal stem cells protect hippocampal neurons from oxidative stress and synapse damage induced by amyloid-β oligomers. Stem Cell Res Ther. 2019;10(1):332.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  155. Wang H, Liu Y, Li J, Wang T, Hei Y, Li H, et al. Tail-vein injection of MSC-derived small extracellular vesicles facilitates the restoration of hippocampal neuronal morphology and function in APP/PS1 mice. Cell Death Discov. 2021;7(1):230.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  156. Wei H, Xu Y, Chen Q, Chen H, Zhu X, Li Y. Mesenchymal stem cell-derived exosomal miR-223 regulates neuronal cell apoptosis. Cell Death Dis. 2020;11(4):290.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  157. Kaniowska D, Wenk K, Rademacher P, Weiss R, Fabian C, Schulz I, et al. Extracellular vesicles of mesenchymal stromal cells can be taken up by microglial cells and partially prevent the stimulation induced by β-amyloid. Stem Cell Rev Rep. 2022;18(3):1113–26.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  158. Chen Y-A, Lu C-H, Ke C-C, Chiu S-J, Jeng F-S, Chang C-W, et al. Mesenchymal stem cell-derived exosomes ameliorate Alzheimer’s disease pathology and improve cognitive deficits. Biomedicines. 2021;9(6):594.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  159. Nakano M, Kubota K, Kobayashi E, Chikenji TS, Saito Y, Konari N, et al. Bone marrow-derived mesenchymal stem cells improve cognitive impairment in an Alzheimer’s disease model by increasing the expression of microRNA-146a in hippocampus. Sci Rep. 2020;10(1):10772.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  160. Yang L, Zhai Y, Hao Y, Zhu Z, Cheng G. The regulatory functionality of exosomes derived from hUMSCs in 3D culture for Alzheimer’s disease therapy. Small. 2020;16(3):e1906273.

    Article  PubMed  Google Scholar 

  161. Hao P, Liang Z, Piao H, Ji X, Wang Y, Liu Y, et al. Conditioned medium of human adipose-derived mesenchymal stem cells mediates protection in neurons following glutamate excitotoxicity by regulating energy metabolism and GAP-43 expression. Metab Brain Dis. 2014;29(1):193–205.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  162. Lee M, Ban J-J, Yang S, Im W, Kim M. The exosome of adipose-derived stem cells reduces β-amyloid pathology and apoptosis of neuronal cells derived from the transgenic mouse model of Alzheimer’s disease. Brain Res. 2018;1691:87–93.

    Article  CAS  PubMed  Google Scholar 

  163. Ma X, Huang M, Zheng M, Dai C, Song Q, Zhang Q, et al. ADSCs-derived extracellular vesicles alleviate neuronal damage, promote neurogenesis and rescue memory loss in mice with Alzheimer’s disease. J Control Release. 2020;327:688–702.

    Article  CAS  PubMed  Google Scholar 

  164. Li B, Liu J, Gu G, Han X, Zhang Q, Zhang W. Impact of neural stem cell-derived extracellular vesicles on mitochondrial dysfunction, sirtuin 1 level, and synaptic deficits in Alzheimer’s disease. J Neurochem. 2020;154(5):502.

    Article  CAS  PubMed  Google Scholar 

  165. Webb RL, Kaiser EE, Scoville SL, Thompson TA, Fatima S, Pandya C, et al. Human neural stem cell extracellular vesicles improve tissue and functional recovery in the murine thromboembolic stroke model. Transl Stroke Res. 2018;9(5):530–9.

    Article  CAS  PubMed  Google Scholar 

  166. Attaluri S, Jaimes Gonzalez J, Kirmani M, Vogel AD, Upadhya R, Kodali M, et al. Intranasally administered extracellular vesicles from human induced pluripotent stem cell-derived neural stem cells quickly incorporate into neurons and microglia in 5xFAD mice. Front Aging Neurosci. 2023;15:1200445.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  167. An K, Klyubin I, Kim Y, Jung JH, Mably AJ, O’Dowd ST, et al. Exosomes neutralize synaptic-plasticity-disrupting activity of Aβ assemblies in vivo. Mol Brain. 2013;6:47.

    Article  PubMed  PubMed Central  Google Scholar 

  168. Yuyama K, Sun H, Sakai S, Mitsutake S, Okada M, Tahara H, et al. Decreased amyloid-β pathologies by intracerebral loading of glycosphingolipid-enriched exosomes in Alzheimer model mice. J Biol Chem. 2014;289(35):24488–98.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  169. Pan J, He R, Huo Q, Shi Y, Zhao L. Brain microvascular endothelial cell derived exosomes potently ameliorate cognitive dysfunction by enhancing the clearance of Aβ through up-regulation of P-gp in mouse model of AD. Neurochem Res. 2020;45(9):2161–72.

    Article  CAS  PubMed  Google Scholar 

  170. Chen HX, Liang FC, Gu P, Xu BL, Xu HJ, Wang WT, et al. Exosomes derived from mesenchymal stem cells repair a Parkinson’s disease model by inducing autophagy. Cell Death Disease. 2020;11(4):288.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  171. Narbute K, Piļipenko V, Pupure J, Dzirkale Z, Jonavičė U, Tunaitis V, et al. Intranasal administration of extracellular vesicles derived from human teeth stem cells improves motor symptoms and normalizes tyrosine hydroxylase expression in the substantia nigra and striatum of the 6-hydroxydopamine-treated rats. Stem Cells Transl Med. 2019;8(5):490–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  172. Norshalena S, Ogura M, Yamaki J, Homma Y. Astrocyte-derived exosomal microRNA miR-200a-3p prevents MPP+-induced apoptotic cell death through down-regulation of MKK4. Neurochem Res. 2020;45(5):1020–33.

    Article  Google Scholar 

  173. Sadanandan N, Lee J-Y, Garbuzova-Davis S. Extracellular vesicle-based therapy for amyotrophic lateral sclerosis. Brain Circul. 2021;7(1):23.

    Article  Google Scholar 

  174. Gschwendtberger T, Thau-Habermann N, von der Ohe J, Luo T, Hass R, Petri S. Protective effects of EVs/exosomes derived from permanently growing human MSC on primary murine ALS motor neurons. Neurosci Lett. 2023;816: 137493.

    Article  CAS  PubMed  Google Scholar 

  175. Bonafede R, Scambi I, Peroni D, Potrich V, Boschi F, Benati D, et al. Exosome derived from murine adipose-derived stromal cells: neuroprotective effect on in vitro model of amyotrophic lateral sclerosis. Exp Cell Res. 2016;340(1):150–8.

    Article  CAS  PubMed  Google Scholar 

  176. Lee M, Ban J-J, Kim KY, Jeon GS, Im W, Sung J-J, et al. Adipose-derived stem cell exosomes alleviate pathology of amyotrophic lateral sclerosis in vitro. Biochem Biophys Res Commun. 2016;479(3):434–9.

    Article  CAS  PubMed  Google Scholar 

  177. Bonafede R, Turano E, Scambi I, Busato A, Bontempi P, Virla F, et al. ASC-exosomes ameliorate the disease progression in SOD1(G93A) murine model underlining their potential therapeutic use in human ALS. Int J Mol Sci. 2020;21(10):3651.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  178. Li Z, Liu F, He X, Yang X, Shan F, Feng J. Exosomes derived from mesenchymal stem cells attenuate inflammation and demyelination of the central nervous system in EAE rats by regulating the polarization of microglia. Int Immunopharmacol. 2019;67:268–80.

    Article  CAS  PubMed  Google Scholar 

  179. Fathollahi A, Hashemi SM, Haji Molla Hoseini M, Tavakoli S, Farahani E, Yeganeh F. Intranasal administration of small extracellular vesicles derived from mesenchymal stem cells ameliorated the experimental autoimmune encephalomyelitis. Int Immunopharmacol. 2021;90:107207.

    Article  CAS  PubMed  Google Scholar 

  180. Rajan TS, Giacoppo S, Diomede F, Ballerini P, Paolantonio M, Marchisio M, et al. The secretome of periodontal ligament stem cells from MS patients protects against EAE. Sci Rep. 2016;6(1):38743.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  181. Lombardi M. Detrimental and protective action of microglial extracellular vesicles on myelin lesions: astrocyte involvement in remyelination failure [Ph.D.]. England: Open University (United Kingdom); 2020.

    Google Scholar 

  182. Lee M, Liu T, Im W, Kim M. Exosomes from adipose-derived stem cells ameliorate phenotype of Huntington’s disease in vitro model. Eur J Neurosci. 2016;44(4):2114–9.

    Article  PubMed  Google Scholar 

  183. Lee S-T, Chu K, Jung K-H, Im W-S, Park J-E, Lim H-C, et al. Slowed progression in models of Huntington disease by adipose stem cell transplantation. Ann Neurol. 2009;66(5):671–81.

    Article  CAS  PubMed  Google Scholar 

  184. Heon-Chang L, Soon-Tae L, Kon C, Kyung Min J, Lami K, Woo-Seok I, et al. Neuroprotective effect of neural stem cell-conditioned media in in vitro model of Huntington’s disease. Neurosci Lett. 2008;435(3):175–80.

    Article  Google Scholar 

  185. Beatriz M, Rodrigues R, Vilaça R, Egas C, Pinheiro P, Daley GQ, et al. Extracellular vesicles improve GABAergic transmission in Huntington’s disease iPSC-derived neurons. Theranostics. 2022;13:3707.

    Article  Google Scholar 

  186. Beatriz M, Rodrigues RJ, Vilaça R, Egas C, Pinheiro PS, Daley GQ, et al. Extracellular vesicles improve GABAergic transmission in Huntington’s disease iPSC-derived neurons. Theranostics. 2023;13(11):3707–24.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  187. Beatriz M, Vilaça R, Rodrigues R, Schlaeger T, Daley G, Januário C, et al. Fibroblasts-derived extracellular vesicles revert synaptic dysfunction in human Huntington’s disease striatal neurons. Eur J Clin Invest. 2021;51(Suppl 1):106–7.

    Google Scholar 

  188. Lee M, Im W, Kim M. Exosomes as a potential messenger unit during heterochronic parabiosis for amelioration of Huntington’s disease. Neurobiol Dis. 2021;155: 105374.

    Article  CAS  PubMed  Google Scholar 

  189. Saini V, Guada L, Yavagal DR. Global epidemiology of stroke and access to acute ischemic stroke interventions. Neurology. 2021;97(20_Supplement_2):S6.

    Article  PubMed  Google Scholar 

  190. Mayer SA, Rincon F. Treatment of intracerebral haemorrhage. Lancet Neurol. 2005;4(10):662–72.

    Article  PubMed  Google Scholar 

  191. Khoshnam SE, Winlow W, Farzaneh M, Farbood Y, Moghaddam HF. Pathogenic mechanisms following ischemic stroke. Neurol Sci. 2017;38(7):1167–86.

    Article  PubMed  Google Scholar 

  192. Unnithan AKA, Das JM, Mehta P. Hemorrhagic stroke. In: StatPearls [Internet]. Treasure Island (FL): StatPearls Publishing; 2024 Jan. Available from: https://www.ncbi.nlm.nih.gov/books/NBK559173/. Updated 2023 May 8.

  193. Alvarez MM, Salazar FE, Rodriguez T, D’Egidio F, Borlongan CV, Lee JY. Endogenous extracellular vesicles participate in brain remodeling after ischemic stroke. Int J Mol Sci. 2023;24(23):16857.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  194. Jin R, Yang G, Li G. Inflammatory mechanisms in ischemic stroke: role of inflammatory cells. J Leukoc Biol. 2010;87(5):779–89.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  195. Li Y, Liu B, Chen Y, Quan X, Han Y, Zheng Y, et al. Extracellular vesicle application as a novel therapeutic strategy for ischemic stroke. Transl Stroke Res. 2022;13(1):171–87.

    Article  PubMed  Google Scholar 

  196. Gordon J, Borlongan CV. An update on stem cell therapy for stroke patients: where are we now? J Cereb Blood Flow Metab. 2024:0271678241227022.

  197. Russo E, Alberti G, Corrao S, Borlongan CV, Miceli V, Conaldi PG, et al. The truth is out there: biological features and clinical indications of extracellular vesicles from human perinatal stem cells. Cells. 2023;12(19):2347.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  198. Borlongan CV, Lee J-Y, D’Egidio F, Kalbermatten Md, Garitaonandia I, Guzman R. Nose-to-brain delivery of stem cells in stroke: the role of extracellular vesicles. Stem Cell Transl Med. 2024. Forthcoming.

  199. Tian H, Tian F, Ma D, Xiao B, Ding Z, Zhai X, et al. Priming and combined strategies for the application of mesenchymal stem cells in ischemic stroke: a promising approach. Mol Neurobiol. 2024.

  200. Haupt M, Gerner ST, Huttner HB, Doeppner TR. Preconditioning concepts for the therapeutic use of extracellular vesicles against stroke. Stem Cells Transl Med. 2023;12(11):707–13.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  201. Wang C, Börger V, Sardari M, Murke F, Skuljec J, Pul R, et al. Mesenchymal stromal cell-derived small extracellular vesicles induce ischemic neuroprotection by modulating leukocytes and specifically neutrophils. Stroke. 2020;51(6):1825–34.

    Article  CAS  PubMed  Google Scholar 

  202. Song Y, Li Z, He T, Qu M, Jiang L, Li W, et al. M2 microglia-derived exosomes protect the mouse brain from ischemia–reperfusion injury via exosomal miR-124. Theranostics. 2019;9(10):2910–23.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  203. Wang J, Chen S, Ma X, Cheng C, Xiao X, Chen J, et al. Effects of endothelial progenitor cell-derived microvesicles on hypoxia/reoxygenation-induced endothelial dysfunction and apoptosis. Oxid Med Cell Longev. 2013;2013: 572729.

    Article  PubMed  PubMed Central  Google Scholar 

  204. Dave KM, Venna VR, Rao KS, Stolz DB, Quaicoe VA, Maniskas ME, et al. Mitochondria-containing extracellular vesicles from mouse vs. human brain endothelial cells for ischemic stroke therapy. bioRxiv. 2024:2024.01.16.575903.

  205. Wang C, Yan B, Liao P, Chen F, Lei P. Meta-analysis of the therapeutic effects of stem cell-derived extracellular vesicles in rodent models of hemorrhagic stroke. Stem Cells Transl Med. 2024;13(7).

  206. Yan J, Zhang Z, Shi H. HIF-1 is involved in high glucose-induced paracellular permeability of brain endothelial cells. Cell Mol Life Sci. 2012;69(1):115–28.

    Article  CAS  PubMed  Google Scholar 

  207. Zhang ZG, Zhang L, Jiang Q, Zhang R, Davies K, Powers C, et al. VEGF enhances angiogenesis and promotes blood–brain barrier leakage in the ischemic brain. J Clin Investig. 2000;106(7):829–36.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  208. Faul M, Coronado V. Epidemiology of traumatic brain injury. In: Grafman J, Salazar AM, editors. Handbook of clinical neurology, vol. 127. Amsterdam: Elsevier; 2015. p. 3–13.

    Google Scholar 

  209. Ghajar J. Traumatic brain injury. The Lancet. 2000;356(9233):923–9.

    Article  CAS  Google Scholar 

  210. Li F, Liu Y, Li L, Peng R, Wang C, Liu C, et al. Brain-derived extracellular vesicles mediate traumatic brain injury associated multi-organ damage. Biochem Biophys Res Commun. 2023;665:141–51.

    Article  CAS  PubMed  Google Scholar 

  211. Li L, Li F, Bai X, Jia H, Wang C, Li P, et al. Circulating extracellular vesicles from patients with traumatic brain injury induce cerebrovascular endothelial dysfunction. Pharmacol Res. 2023;192: 106791.

    Article  CAS  PubMed  Google Scholar 

  212. Li F, Li L, Peng R, Liu C, Liu X, Liu Y, et al. Brain-derived extracellular vesicles mediate systemic coagulopathy and inflammation after traumatic brain injury. Int Immunopharmacol. 2024;130: 111674.

    Article  CAS  PubMed  Google Scholar 

  213. Sharma N, Verma R, Kumawat KL, Basu A, Singh SK. miR-146a suppresses cellular immune response during Japanese encephalitis virus JaOArS982 strain infection in human microglial cells. J Neuroinflammation. 2015;12(1):30.

    Article  PubMed  PubMed Central  Google Scholar 

  214. Zhao Y, Gan Y, Xu G, Yin G, Liu D. MSCs-derived exosomes attenuate acute brain injury and inhibit microglial inflammation by reversing CysLT2R-ERK1/2 mediated microglia M1 polarization. Neurochem Res. 2020;45(5):1180–90.

    Article  CAS  PubMed  Google Scholar 

  215. Kodali M, Madhu LN, Reger RL, Milutinovic B, Upadhya R, Attaluri S, et al. A single intranasal dose of human mesenchymal stem cell-derived extracellular vesicles after traumatic brain injury eases neurogenesis decline, synapse loss, and BDNF-ERK-CREB signaling. Front Mol Neurosci. 2023;16:1185883.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  216. Rassart E, Bedirian A, Do Carmo S, Guinard O, Sirois J, Terrisse L, et al. Apolipoprotein D. Biochim Biophys Acta (BBA) Protein Struct Mol Enzymol. 2000;1482(1):185–98.

    Article  CAS  Google Scholar 

  217. Gao Y, Wang C, Jin F, Han G, Cui C. Therapeutic effect of extracellular vesicles from different cell sources in traumatic brain injury. Tissue Cell. 2022;76: 101772.

    Article  CAS  PubMed  Google Scholar 

  218. Zhong L, Wang J, Wang P, Liu X, Liu P, Cheng X, et al. Neural stem cell-derived exosomes and regeneration: cell-free therapeutic strategies for traumatic brain injury. Stem Cell Res Ther. 2023;14(1):198.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  219. Yuan P, Ding L, Chen H, Wang Y, Li C, Zhao S, et al. Neural stem cell-derived exosomes regulate neural stem cell differentiation through miR-9-Hes1 axis. Front Cell Dev Biol. 2021;9:601600.

    Article  PubMed  PubMed Central  Google Scholar 

  220. Zhao M, Gao Y, Wang F, Cheng X, Zhao T, Zhao Y, et al. Neural progenitor cells-secreted exosomal miR-210 induced by hypoxia influences cell viability. NeuroReport. 2020;31(11):798.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  221. Li W, Shan B, Cheng X, He H, Qin J, Zhao H, et al. crcRNA Acbd6 promotes neural stem cell differentiation into cholinergic neurons via the miR-320–5p-Osbpl2 axis. J Biol Chem. 2022;298(4):101828.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  222. 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(2):110–6.

    Article  CAS  PubMed  Google Scholar 

  223. Ahuja CS, Wilson JR, Nori S, Kotter MRN, Druschel C, Curt A, et al. Traumatic spinal cord injury. Nat Rev Dis Primers. 2017;3(1):17018.

    Article  PubMed  Google Scholar 

  224. Witiw CD, Fehlings MG. Acute spinal cord injury. Clin Spine Surg. 2015;28(6).

  225. Dutta D, Khan N, Wu J, Jay SM. Extracellular vesicles as an emerging frontier in spinal cord injury pathobiology and therapy. Trends Neurosci. 2021;44(6):492–506.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  226. 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(6):5184–206.

    Article  CAS  PubMed  Google Scholar 

  227. Lombardi M, Parolisi R, Scaroni F, Bonfanti E, Gualerzi A, Gabrielli M, et al. Detrimental and protective action of microglial extracellular vesicles on myelin lesions: astrocyte involvement in remyelination failure. Acta Neuropathol. 2019;138(6):987–1012.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  228. Liang Z, Yang Z, Xie H, Rao J, Xu X, Lin Y, et al. Small extracellular vesicles from hypoxia-preconditioned bone marrow mesenchymal stem cells attenuate spinal cord injury via miR-146a-5p-mediated regulation of macrophage polarization. Neural Regen Res. 2024;19(10):2259.

    PubMed  PubMed Central  Google Scholar 

  229. 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(24):12278–94.

    Article  CAS  PubMed  Google Scholar 

  230. Namini MS, Daneshimehr F, Beheshtizadeh N, Mansouri V, Ai J, Jahromi HK, et al. Cell-free therapy based on extracellular vesicles: a promising therapeutic strategy for peripheral nerve injury. Stem Cell Res Ther. 2023;14(1):254.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  231. Lee SK, Wolfe SW. Peripheral nerve injury and repair. JAAOS J Am Acad Orthopaedic Surgeons. 2000;8(4).

  232. Arslantunali D, Dursun T, Yucel D, Hasirci N, Hasirci V. Peripheral nerve conduits: technology update. Medical Devices Evid Res. 2014;7:405–24.

    CAS  Google Scholar 

  233. Yi S, Yuan Y, Chen Q, Wang X, Gong L, Liu J, et al. Regulation of Schwann cell proliferation and migration by miR-1 targeting brain-derived neurotrophic factor after peripheral nerve injury. Sci Rep. 2016;6(1):29121.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  234. Zhang Y, Liu J, Wang X, Zhang J, Xie C. Extracellular vesicle-encapsulated microRNA-23a from dorsal root ganglia neurons binds to A20 and promotes inflammatory macrophage polarization following peripheral nerve injury. Aging (Albany NY). 2021;13(5):6752–64.

    Article  CAS  PubMed  Google Scholar 

  235. Shiue S-J, Rau R-H, Shiue H-S, Hung Y-W, Li Z-X, Yang KD, et al. Mesenchymal stem cell exosomes as a cell-free therapy for nerve injury-induced pain in rats. Pain. 2019;160(1):210.

    Article  CAS  PubMed  Google Scholar 

  236. Yin GN, Park S-H, Ock J, Choi M-J, Limanjaya A, Ghatak K, et al. Pericyte-derived extracellular vesicle-mimetic nanovesicles restore erectile function by enhancing neurovascular regeneration in a mouse model of cavernous nerve injury. J Sex Med. 2020;17(11):2118–28.

    Article  CAS  PubMed  Google Scholar 

  237. Pan J, Zhao M, Yi X, Tao J, Li S, Jiang Z, et al. Acellular nerve grafts supplemented with induced pluripotent stem cell-derived exosomes promote peripheral nerve reconstruction and motor function recovery. Bioactive Mater. 2022;15:272–87.

    Article  CAS  Google Scholar 

  238. Chen J, Zhu Y, Gao H, Chen X, Yi D, Li M, et al. HucMSCs delay muscle atrophy after peripheral nerve injury through exosomes by repressing muscle-specific ubiquitin ligases. Stem Cells. 2024;42(5):460–74.

    Article  PubMed  PubMed Central  Google Scholar 

  239. Wang L, Chopp M, Szalad A, Lu X, Zhang Y, Wang X, et al. Exosomes derived from Schwann cells ameliorate peripheral neuropathy in type 2 diabetic mice. Diabetes. 2020;69(4):749–59.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  240. Zhang Y, Li C, Qin Y, Cepparulo P, Millman M, Chopp M, et al. Small extracellular vesicles ameliorate peripheral neuropathy and enhance chemotherapy of oxaliplatin on ovarian cancer. J Extracell Vesicles. 2021;10(5):12073.

    Article  Google Scholar 

  241. Xia B, Gao J, Li S, Huang L, Zhu L, Ma T, et al. Mechanical stimulation of Schwann cells promotes peripheral nerve regeneration via extracellular vesicle-mediated transfer of microRNA 23b-3p. Theranostics. 2020;10(20):8974–95.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  242. Feng R, Huan N, Zhao M, Gao X, Zheng F. Combination of 1% platelet-rich plasma and bone marrow mesenchymal stem cells improves the recovery of peripheral nerve injury. Chinese J Tissue Eng Res. 2024;28(7):985–92.

    Google Scholar 

  243. Zhang Y, Yi D, Hong Q, Cao J, Geng X, Liu J, et al. Platelet-rich plasma-derived exosomes boost mesenchymal stem cells to promote peripheral nerve regeneration. J Control Release. 2024;367:265–82.

    Article  CAS  PubMed  Google Scholar 

  244. Cong M, Hu J-J, Yu Y, Li X-L, Sun X-T, Wang L-T, et al. miRNA-21-5p is an important contributor to the promotion of injured peripheral nerve regeneration using hypoxia-pretreated bone marrow-derived neural crest cells. Neural Regen Res. 2025;20(1):277.

    Article  PubMed  Google Scholar 

  245. Yu M, Gu G, Cong M, Du M, Wang W, Shen M, et al. Repair of peripheral nerve defects by nerve grafts incorporated with extracellular vesicles from skin-derived precursor Schwann cells. Acta Biomater. 2021;134:190–203.

    Article  CAS  PubMed  Google Scholar 

  246. Liu B, Alimi OA, Wang Y, Kong Y, Kuss M, Krishnan MA, et al. Differentiated mesenchymal stem cells-derived exosomes immobilized in decellularized sciatic nerve hydrogels for peripheral nerve repair. J Control Release. 2024;368:24–41.

    Article  CAS  PubMed  Google Scholar 

  247. Gao Y, Dai C, Zhang M, Zhang J, Yin L, Li W, et al. Biomimetic silk fibroin hydrogel for enhanced peripheral nerve regeneration: synergistic effects of graphene oxide and fibroblast exosome. Adv Func Mater. 2024;34(17):2314610.

    Article  CAS  Google Scholar 

  248. Xia B, Gao X, Qian J, Li S, Yu B, Hao Y, et al. A novel superparamagnetic multifunctional nerve scaffold: a remote actuation strategy to boost in situ extracellular vesicles production for enhanced peripheral nerve repair. Adv Mater. 2024;36(3):2305374.

    Article  CAS  Google Scholar 

  249. Thijs RD, Surges R, O’Brien TJ, Sander JW. Epilepsy in adults. The Lancet. 2019;393(10172):689–701.

    Article  Google Scholar 

  250. Upadhya D, Shetty AK. Promise of extracellular vesicles for diagnosis and treatment of epilepsy. Epilepsy Behav. 2021;121:106499.

    Article  PubMed  Google Scholar 

  251. Alayli A, Lockard G, Gordon J, Connolly J, Monsour M, Schimmel S, et al. Stem cells: recent developments redefining epilepsy therapy. Cell Transplant. 2023;32:09636897231158967.

    Article  PubMed  PubMed Central  Google Scholar 

  252. Abhijna Ballal R, Shivakumar Reddy K, Chandran D, Hegde S, Upadhya R, Se PK, et al. Cell-specific extracellular vesicle-encapsulated exogenous GABA controls seizures in epilepsy. Stem Cell Res Ther. 2024;15(1):108.

    Article  Google Scholar 

  253. Tahami Monfared AA, Byrnes MJ, White LA, Zhang Q. Alzheimer’s disease: epidemiology and clinical progression. Neurol Thera. 2022;11(2):553–69.

    Article  Google Scholar 

  254. Tiwari S, Atluri V, Kaushik A, Yndart A, Nair M. Alzheimer’s disease: pathogenesis, diagnostics, and therapeutics. Int J Nanomed. 2019;14:5541–54.

    Article  CAS  Google Scholar 

  255. Liu X, Chen C, Jiang Y, Wan M, Jiao B, Liao X, et al. Brain-derived extracellular vesicles promote bone-fat imbalance in Alzheimer’s disease. Int J Biol Sci. 2023;19(8):2409–27.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  256. Cyr B, Cabrera Ranaldi EDLRM, Hadad R, Dietrich WD, Keane RW, de Rivero Vaccari JP. Extracellular vesicles mediate inflammasome signaling in the brain and heart of Alzheimer’s disease mice. Front Mol Neurosci. 2024;17:1369781.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  257. Gomes P, Tzouanou F, Skolariki K, Vamvaka-Iakovou A, Noguera-Ortiz C, Tsirtsaki K, et al. Extracellular vesicles and Alzheimer’s disease in the novel era of precision medicine: implications for disease progression, diagnosis and treatment. Exp Neurol. 2022;358: 114183.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  258. Dinkins MB, Enasko J, Hernandez C, Wang G, Kong J, Helwa I, et al. Neutral sphingomyelinase-2 deficiency ameliorates Alzheimer’s disease pathology and improves cognition in the 5XFAD mouse. J Neurosci. 2016;36(33):8653.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  259. Crivelli SM, Quadri Z, Vekaria HJ, Zhu Z, Tripathi P, Elsherbini A, et al. Inhibition of acid sphingomyelinase reduces reactive astrocyte secretion of mitotoxic extracellular vesicles and improves Alzheimer’s disease pathology in the 5xFAD mouse. Acta Neuropathol Commun. 2023;11(1):135.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  260. Sobue A, Ito N, Nagai T, Shan W, Hada K, Nakajima A, et al. Astroglial major histocompatibility complex class I following immune activation leads to behavioral and neuropathological changes. Glia. 2018;66(5):1034–52.

    Article  PubMed  Google Scholar 

  261. Ruan Z, Delpech J-C, Venkatesan Kalavai S, Van Enoo AA, Hu J, Ikezu S, et al. P2RX7 inhibitor suppresses exosome secretion and disease phenotype in P301S tau transgenic mice. Mol Neurodegen. 2020;15(1):47.

    Article  CAS  Google Scholar 

  262. Yin T, Liu Y, Ji W, Zhuang J, Chen X, Gong B, et al. Engineered mesenchymal stem cell-derived extracellular vesicles: a state-of-the-art multifunctional weapon against Alzheimer’s disease. Theranostics. 2023;13(4):1264–85.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  263. Bai Z, Ge K, Fu J, Yu D, Hua Z, Xue S, et al. Engineered urinary-derived extracellular vesicles loaded nanoenzymes as Trojan horses to regulate the inflammatory microenvironment for treatment of Alzheimer’s disease. Chem Eng J. 2023;465: 142955.

    Article  CAS  Google Scholar 

  264. Perets N, Betzer O, Shapira R, Brenstein S, Angel A, Sadan T, et al. Golden exosomes selectively target brain pathologies in neurodegenerative and neurodevelopmental disorders. Nano Lett. 2019;19(6):3422–31.

    Article  CAS  PubMed  Google Scholar 

  265. de Godoy MA, Saraiva LM, de Carvalho LRP, Vasconcelos-dos-Santos A, Beiral HJV, Ramos AB, et al. Mesenchymal stem cells and cell-derived extracellular vesicles protect hippocampal neurons from oxidative stress and synapse damage induced by amyloid-β oligomers. J Biol Chem. 2018;293(6):1957–75.

    Article  PubMed  Google Scholar 

  266. Benhamron S, Nitzan K, Valitsky M, Lax N, Karussis D, Kassis I, et al. Cerebrospinal fluid (CSF) exchange therapy with artificial CSF enriched with mesenchymal stem cell secretions ameliorates cognitive deficits and brain pathology in Alzheimer’s disease mice. J Alzheimers Dis. 2020;76(1):369–85.

    Article  CAS  PubMed  Google Scholar 

  267. Katsuda T, Tsuchiya R, Kosaka N, Yoshioka Y, Takagaki K, Oki K, et al. Human adipose tissue-derived mesenchymal stem cells secrete functional neprilysin-bound exosomes. Sci Rep. 2013;3:1197.

    Article  PubMed  PubMed Central  Google Scholar 

  268. Tysnes O-B, Storstein A. Epidemiology of Parkinson’s disease. J Neural Transm. 2017;124(8):901–5.

    Article  PubMed  Google Scholar 

  269. de Lau LML, Breteler MMB. Epidemiology of Parkinson’s disease. The Lancet Neurol. 2006;5(6):525–35.

    Article  PubMed  Google Scholar 

  270. Leggio L, Paternò G, Vivarelli S, L’Episcopo F, Tirolo C, Raciti G, et al. Extracellular vesicles as nanotherapeutics for Parkinson’s disease. Biomolecules. 2020;10(9):1327.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  271. Thompson AG, Gray E, Heman-Ackah SM, Mäger I, Talbot K, Andaloussi SE, et al. Extracellular vesicles in neurodegenerative disease—pathogenesis to biomarkers. Nat Rev Neurol. 2016;12(6):346–57.

    Article  CAS  PubMed  Google Scholar 

  272. Leng B, Sun H, Zhao J, Liu Y, Shen T, Liu W, et al. Plasma exosomal prion protein levels are correlated with cognitive decline in PD patients. Neurosci Lett. 2020;723: 134866.

    Article  CAS  PubMed  Google Scholar 

  273. Urrea L, Ferrer I, Gavín R, del Río JA. The cellular prion protein (PrPC) as neuronal receptor for α-synuclein. Prion. 2017;11(4):226–36.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  274. Jarmalavičiūtė A, Tunaitis V, Pivoraitė U, Venalis A, Pivoriūnas A. Exosomes from dental pulp stem cells rescue human dopaminergic neurons from 6-hydroxy-dopamine-induced apoptosis. Cytotherapy. 2015;17(7):932–9.

    Article  PubMed  Google Scholar 

  275. Thonhoff JR, Simpson EP, Appel SH. Neuroinflammatory mechanisms in amyotrophic lateral sclerosis pathogenesis. Curr Opin Neurol. 2018;31(5):635.

    Article  CAS  PubMed  Google Scholar 

  276. Morgan S, Orrell RW. Pathogenesis of amyotrophic lateral sclerosis. Br Med Bull. 2016;119(1):87–98.

    Article  CAS  PubMed  Google Scholar 

  277. Bonafede R, Mariotti R. ALS pathogenesis and therapeutic approaches: the role of mesenchymal stem cells and extracellular vesicles. Front Cell Neurosci. 2017;11:80.

    Article  PubMed  PubMed Central  Google Scholar 

  278. Lockard G, Gordon J, Schimmel S, El Sayed B, Monsour M, Garbuzova-Davis S, et al. Attenuation of amyotrophic lateral sclerosis via stem cell and extracellular vesicle therapy: an updated review. Neuroprotection. 2023;1(2):130–8.

    Article  PubMed  PubMed Central  Google Scholar 

  279. Goldschmidt-Clermont PJ, Khan A, Guest JD, Jimsheleishvili G, Graham P, Brooks A, et al. A novel therapy for ALS: allogeneic Schwann cell extracellular vesicles. medRxiv. 2023:2023.01.18.23284378.

  280. Lassmann H, van Horssen J, Mahad D. Progressive multiple sclerosis: pathology and pathogenesis. Nat Rev Neurol. 2012;8(11):647–56.

    Article  CAS  PubMed  Google Scholar 

  281. Ghasemi N, Razavi S, Nikzad E. Multiple sclerosis: pathogenesis, symptoms, diagnoses and cell-based therapy. Cell J. 2017;19(1):1–10.

    PubMed  Google Scholar 

  282. Barabadi M, Paton MCB, Kumar N, Lim R, Payne NL. Stem cell derived extracellular vesicle therapy for multiple sclerosis, a systematic review and meta-analysis of preclinical studies. Stem Cells Transl Med. 2024;13(5):436–47.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  283. Rawlins MD, Wexler NS, Wexler AR, Tabrizi SJ, Douglas I, Evans SJW, et al. The prevalence of Huntington’s disease. Neuroepidemiology. 2016;46(2):144–53.

    Article  PubMed  Google Scholar 

  284. Bates GP, Dorsey R, Gusella JF, Hayden MR, Kay C, Leavitt BR, et al. Huntington disease. Nat Rev Disease Primers. 2015;1(1):15005.

    Article  PubMed  Google Scholar 

  285. Walker FO. Huntington’s disease. The Lancet. 2007;369(9557):218–28.

    Article  CAS  Google Scholar 

  286. Li S-H, Li X-J. Huntingtin–protein interactions and the pathogenesis of Huntington’s disease. Trends Genet. 2004;20(3):146–54.

    Article  PubMed  Google Scholar 

  287. Costanzo M, Abounit S, Marzo L, Danckaert A, Chamoun Z, Roux P, et al. Transfer of polyglutamine aggregates in neuronal cells occurs in tunneling nanotubes. J Cell Sci. 2013;126(Pt 16):3678–85.

    CAS  PubMed  Google Scholar 

  288. Yang W, Dunlap JR, Andrews RB, Wetzel R. Aggregated polyglutamine peptides delivered to nuclei are toxic to mammalian cells. Hum Mol Genet. 2002;11(23):2905–17.

    Article  CAS  PubMed  Google Scholar 

  289. Herrera F, Tenreiro S, Miller-Fleming L, Outeiro TF. Visualization of cell-to-cell transmission of mutant huntingtin oligomers. PLoS Curr. 2011;3.

  290. Bahar R, Darabi S, Norouzian M, Roustaei S, Torkamani-dordshaikh S, Hasanzadeh M, et al. Neuroprotective effect of human cord blood-derived extracellular vesicles by improved neuromuscular function and reduced gliosis in a rat model of Huntington’s disease. J Chem Neuroanat. 2024;138: 102419.

    Article  CAS  PubMed  Google Scholar 

  291. Kwon S, Shin S, Do M, Oh BH, Song Y, Bui VD, Lee ES, Jo DG, Cho YW, Kim DH, Park JH. Engineering approaches for effective therapeutic applications based on extracellular vesicles. J Control Release. 2021;330:15–30.

    Article  CAS  PubMed  Google Scholar 

  292. Liang Y, Iqbal Z, Lu J, Chen X, Duan L, Xia J. Cell-derived nanovesicle-mediated drug delivery to the brain: principles and strategies for vesicle engineering. Mol Ther. 2023;31(5):1207–24.

    Article  CAS  PubMed  Google Scholar 

  293. Choi HK, Chen M, Goldston LL, et al. Extracellular vesicles as nanotheranostic platforms for targeted neurological disorder interventions. Nano Convergence. 2024;11:19.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  294. Wang L, Wang D, Ye Z, Xu J. Engineering extracellular vesicles as delivery systems in therapeutic applications. Adv Sci. 2023;10(18):2300552.

    Article  CAS  Google Scholar 

  295. Armstrong JPK, Holme MN, Stevens MM. Re-engineering extracellular vesicles as smart nanoscale therapeutics. ACS Nano. 2017;11(1):69–83.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  296. Ramasubramanian A, Kumar P, Wang A. Extracellular vesicles as therapeutic delivery vehicles in regenerative medicine. Biomolecules. 2020;10(1):48.

    Article  CAS  Google Scholar 

  297. Sato Y, Umezaki K, Sawada S, et al. Engineering hybrid exosomes by membrane fusion with liposomes. Sci Rep. 2016;6:21933.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  298. Lin Y, Wu J, Gu W, Huang Y, Tong Z, Huang L, Tan J. Exosome–liposome hybrid nanoparticles deliver CRISPR/Cas9 system in MSCs. Adv Sci. 2018;5(4):1700611.

    Article  Google Scholar 

  299. Alvarez-Erviti L, Seow Y, Yin H, et al. Delivery of siRNA to the mouse brain by systemic injection of targeted exosomes. Nat Biotechnol. 2011;29(4):341–5.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

Not applicable.

Funding

This research received no specific grant from any funding agency in the public, commercial, or not-for-profit sectors.

Author information

Authors and Affiliations

Authors

Contributions

NP was the major contributor in reviewing and writing the manuscript with support from JYL. CVB supervised all processes. All authors read and approved the final manuscript.

Corresponding author

Correspondence to Cesario V. Borlongan.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Additional information

Publisher's Note

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

Rights and permissions

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

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Putthanbut, N., Lee, J.Y. & Borlongan, C.V. Extracellular vesicle therapy in neurological disorders. J Biomed Sci 31, 85 (2024). https://doi.org/10.1186/s12929-024-01075-w

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12929-024-01075-w

Keywords