View Full Text | Abstract |
Article as PDF | Print this Article |
Pubmed | PMC |
PubReader | Export to Citation |
Email Alerts | Open Access |
Exp Neurobiol 2024; 33(2): 47-67
Published online April 30, 2024
https://doi.org/10.5607/en24002
© The Korean Society for Brain and Neural Sciences
Chutithep Teekaput1, Kitti Thiankhaw1, Nipon Chattipakorn2,3 and Siriporn C. Chattipakorn2,3,4*
1Department of Internal Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, 2Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, 3Center of Excellence in Cardiac Electrophysiology, Chiang Mai University, Chiang Mai 50200, 4Department of Oral Biology and Diagnostic Sciences, Faculty of Dentistry, Chiang Mai University, Chiang Mai 50200, Thailand
Correspondence to: *To whom correspondence should be addressed.
TEL: 011-66-53-944-451, FAX: 011-66-53-222-844
e-mail: scchattipakorn@gmail.com
This is an Open Access article distributed under the terms of the Creative Commons Attribution Non-Commercial License (http://creativecommons.org/licenses/by-nc/4.0) which permits unrestricted non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited.
Multiple sclerosis (MS) and neuromyelitis optica spectrum disorder (NMOSD) are two of the most devastating immune-mediated central demyelinating disorders. NMOSD was once considered as a variant of MS until the discovery of an antibody specific to the condition. Despite both MS and NMOSD being considered central demyelinating disorders, their pathogenesis and clinical manifestations are distinct, however the exact mechanisms associated with each disease remain unclear. Extracellular vesicles (EVs) are nano-sized vesicles originating in various cells which serve as intercellular communicators. There is a large body of evidence to show the possible roles of EVs in the pathogenesis of several diseases, including the immune-mediated central demyelinating disorders. Various types of EVs are found across disease stages and could potentially be used as a surrogate marker, as well as acting by carrying a cargo of biochemical molecules. The possibility for EVs to be used as a next-generation targeted treatment for the immune-mediated central demyelinating disorders has been investigated. The aim of this review was to comprehensively identify, compile and discuss key findings from in vitro, in vivo and clinical studies. A summary of all findings shows that: 1) the EV profiles of MS and NMOSD differ from those of healthy individuals, 2) the use of EV markers as liquid biopsy diagnostic tools appears to be promising biomarkers for both MS and NMOSD, and 3) EVs are being studied as a potential targeted therapy for MS and NMOSD. Any controversial findings are also discussed in this review.
Keywords: Demyelination, Multiple sclerosis, Neuromyelitis optica spectrum disorders, Extracellular vesicles, Exosome
Multiple sclerosis (MS) and neuromyelitis optica spectrum disorder (NMOSD), both immune-mediated demyelinating disorders of the central nervous system (CNS), have been acknowledged as being some of the most debilitating neurological conditions. MS is a chronic immune-mediated demyelinating disorder that primarily affects Europeans, and is uncommon in Asians, Africans, and indigenous Americans [1]. It has been suggested that the etiology of MS is centrally demyelination induced as a consequence of inflammatory cells invading the CNS through blood vessels in the blood-brain barrier (BBB) [2]. These inflammatory cells, in addition to activated microglia and astrocytes, have been shown to contribute to oligodendrocyte injury [1, 3, 4]. The relapsing-remitting phenotype of MS, which accounts for 85-90% of all MS cases, is caused by peripheral inflammation as a result of periodic invasion [4, 5]. In the advanced progressive stage of MS, the repetitive destruction of oligodendrocytes causes persistent oxidative stress, mitochondrial dysfunction, brain iron accumulation, hypoxia, defective remyelination, and glutamate hyperexcitability, eventually leading to neurodegeneration [1, 6]. The clinical manifestations of MS vary based on the area affected, frequent locations being the optic nerve, the cerebral cortex, white matter, brainstem, and spinal cord [1]. Disease-modifying therapies (DMTs), which primarily target inflammatory cascades and have pleiotropic effects, have been the standard therapies for MS [3]. The exact pathophysiology of MS, however, remains unknown, and no specific antibody has been identified and investigated facilitating the detection of MS. As a result, the diagnosis of MS still utilizes the 2017 McDonald criteria which indicate space and time dissemination [7].
Until 2004 NMOSD was considered an MS variant but then a specific antibody was discovered [8]. That antibody is immunoglobulin G (IgG) directed against aquaporin 4 (AQP4), in combination with complement system activation [8, 9]. That antibody causes invasion of the CNS by peripheral granulocytes through the CNS, resulting in astrocytic endfeet damage, severe inflammation, secondary oligodendrocytic injury, and demyelination [9, 10]. The six essential clinical characteristics that must be present in addition to the presence of AQP4 IgG for a diagnosis of NMOSD are longitudinally extensive transverse myelitis (LETM), optic neuritis (ON), area postrema syndrome (APS), brainstem encephalitis, diencephalitis, and telencephalitis [9]. Autoantibody elimination and inflammatory cascade suppression have been the main goals of NMOSD therapy [9, 11, 12].
Extracellular vesicles (EVs) are nano-sized, non-nucleated structures that are hypothesized to facilitate intercellular communication and contribute to autoimmune processes. EVs have been connected to the development and deterioration of a range of pathological conditions, including cancer, cerebrovascular disease, Alzheimer’s disease, and central demyelinating diseases (MS and NMOSD) [13-17]. Many studies have described the potential involvement of EVs as possible biomarkers and as a novel diagnostic approach known as a liquid biopsy for a variety of disorders [18-20]. Furthermore, one of the therapeutic uses of EVs which has the greatest potential is as carrier for the drug delivery system [21]. As a result of those previous studies, the aim of this review was to summarize and discuss the evidence available from
The PubMed database was searched using the keywords: “
EVs are double-layered and non-nucleated vesicles that contain a diversity of cytoplasmic biological substances. Different cell types, in both physiological and pathological stages, secrete distinctive EVs in order to maintain cellular homeostasis and communicate with adjacent cells [22]. Considered as a carrier for biochemical compounds, enzymes, signaling molecules, biogenesis factors, chaperones, cytoskeletal factors, and nucleic acids especially microRNA are contained within EVs. Based on their biogenesis, release mechanisms, size, composition, and function, the three primary subtypes of extracellular vesicles (EVs) are exosomes, microvesicles, and apoptotic bodies [13, 23-26]. Exosomes are the smallest EVs, ranging from 30~100 nm in diameter, and are formed by the inward budding of the endosomal compartment, subsequently being released to the periphery by the exocytotic process. Exosomes usually contain lipids and nucleic acids [24]. Microvesicles, however, are approximately 100~1,000 nm in diameter and since they arise directly from the cell membrane, they typically express transmembrane molecules and glycoproteins depending on their parental cells [25]. Apoptotic bodies are approximately 1,000 to 5,000 nm in diameter and mostly develop in the late stage of apoptotic cells. Phosphatidylserine is often transferred to the outer membrane of apoptotic bodies, which then stimulates phagocytes to remove the dead cells [26]. As previously stated, EVs are involved in cell-to-cell communication; hence, dysfunctional EVs lead to pathogenic processes and promote autoimmunity [27].
Extracted EVs from MS and NMOSD patients were investigated using various methods. However, the interpretation and comparison of the findings across studies must be considered carefully. Different isolation and characterization techniques used in the different studies caused different EV findings [28]. Focusing on the isolation technique, ultracentrifugation (UC) is the most widely used isolation method and is considered the gold standard for EV extraction and separation. UC does not need EV labeling and can avoid cross-contamination. However, this technique is time-consuming and expensive [22]. Polymer precipitation is another approach that uses polyethylene glycol as a medium, the technique involves the incubation of EVs under centrifugation conditions by decreasing the EV solubility. Contrarily, this approach has a high false-positive rate, and the polymer has an adhesive feature that makes it difficult to remove for future EV investigation. Size-based isolation by means of size exclusion chromatography is an alternative technology with a high enrichment efficiency and low cost. The main disadvantage of this technique is its impurity [22, 28]. The different characterization techniques also can have impact on different EV characteristics. Characterization techniques are mainly divided into external characterization (primarily morphology and particle size detection) and internal characterization (membrane protein and lipid raft). The electron microscope is used to detect EV morphology, but the technique is complicated and unsuitable for the rapid testing of many EV samples. Nanoparticle tracking analysis (NTA) is a real-time detection method but carries a high risk of impurity [28]. Camera levels and detection thresholds affect the quality of NTA. Western blotting is a conventional technique that can qualitatively and quantitatively analyze marker proteins. However, the detection of marker proteins varies depending on the type of parental cell, and it is not suitable for the detection of EV marker proteins in biological fluids [28].
Since the quantity of EVs in blood plasma or serum is believed to be between 107 and 1012 EV/ml, serum EVs have garnered a great deal of attention [29]. However, many investigators have demonstrated the EVs in various samples beyond serum. Different sample types can result in different EV profiles. In contrast with serum, cerebrospinal fluid (CSF) is a type of biological fluid secreted from choroid plexus ependymal cells. CSF surrounds the brain and spinal cord in order to protect the CNS, support homeostasis, and remove waste products. Approximately 80% of CSF EVs originate from the blood, whereas the remaining are produced by nerve cells [30]. However, due to the high integrity of the BBB, the protein level of CSF becomes 100 to 300 times lower than protein found in serum [30, 31]. Therefore, a high amount of CSF is required in order to obtain sufficient EVs for analysis. Albumin and immunoglobulin, the two most abundant proteins in the CSF, may hinder the identification of low-abundance proteins and result in a substantial loss of EV information. As a result, direct comparisons between methods may be difficult according to the advantages and disadvantages of individual techniques. Size, function, sample type, and parental source of EV make isolating and characterization of EV problematic [28, 32].
The central paradigm of central demyelinating diseases, especially in the case of MS, is an increase in the inflammatory process and a downregulation of immune tolerance. EVs derived from various origins carry different characteristics and functions.
Endothelial cell derived extracellular vesicles
The pathophysiology of MS is thought to involve endothelial dysfunction as a crucial factor. The expression of certain markers on the endothelial surface, such as CD31 (PECAM-1), CD51 (integrin), CD54 (ICAM-1), CD62E (ELAM-1 or E-selectin), CD63E, and CD105 (endoglin), increases the adhesion of inflammatory leukocytes to endothelial cells and promotes their transendothelial migration into the CNS [33-37].
It is believed that radiological exacerbation, which refers to an increase in the severity of lesions seen on imaging without the presentation of any clinical symptoms, occurs prior to or simultaneously with clinical exacerbation. Notably, CD31 and CD54, which are localized at endothelial cell intercellular junctions, show high levels of expression during radiological relapse even in the absence of clinical symptoms [34, 36]. Once the exacerbation reaches the clinical exacerbation threshold, endothelial cell damage occurs, leading to the disruption of the BBB. CD31 and CD62E are among the earliest markers to be released, indicating endothelial injury. CD51, along with CD54, which acts as a transmembrane receptor, facilitates leukocyte recruitment and adhesion to endothelial cells via Th1 cell activation [34, 36, 38]. CD51 also plays a role in signaling between the extracellular matrix and cells and has been found to indirectly contribute to nerve regeneration. Following an exacerbation, demyelinated neurons attempt to repair and promote remyelination. The expression of CD51 increases after an attack but remains elevated even after treatment, suggesting that remyelination is a long-term process [39]. However, not all brain regions exhibit the same levels of these endothelial EV markers. Based on the process of neurovascular injury, the subcortical gray matter periventricular region, which is highly vascularized, is the most affected area of the brain [35]. The association between different brain regions may provide additional insights from imaging data for predicting the clinical course and early diagnosis of relapse.
Platelet-derived extracellular vesicles
Platelets have been classified as inflammatory cells due to their multifaceted functions beyond hemostasis. Within platelet-alpha granules reside a wide range of EVs responsible for inflammatory process [40]. Following BBB disruption from impaired endothelial cells and astrocyte foot processes, peripheral inflammatory cells are recruited into the CNS. Platelet-derived EVs have been proposed to play a significant role in leukocyte recruitment and to facilitate transendothelial migration into the CNS, leading to inflammation and demyelination. In patients with MS, CD31+ marker of platelet EV is observed only in the inflammation-predominant stage, RRMS [33, 35]. This phenomenon is not evident in the very early stage, CIS, nor in the advanced stage, SPMS. The postulated explanation is that there is limited degree of inflammation demonstrated in CIS, whereas in SPMS, neurodegeneration rather than neuroinflammation is the primary pathophysiology [41]. However, some level of inflammation persists across all stages of MS evidenced by the heightened expression of CD61 (integrin beta 3), CD62 (P-selectin), and surface IgM on platelets [33, 42, 43]. The alteration of platelet-derived EVs could potentially serve as a marker for identifying MS stages and predicting MS prognosis [33, 35].
Leukocyte-derived extracellular vesicles
Inflammation arises from an imbalance between the increase in proinflammatory leukocytes and the decrease of regulatory T cells (Treg). A recent study has found that specific markers of T cells in CSF, such as CCR3 and CCR5, exhibit high expression during radiological relapse, but not during clinical relapse [44]. It is suggested that activated CD4+ and CD8+ T lymphocytes play a significant role in triggering inflammation. An in vitro study has demonstrated that co-culturing human endothelial cells with T cell EVs leads to a decrease in transendothelial electrical resistance, indicating BBB disruption and astrocyte damage [45, 46]. Furthermore, both in vitro and clinical studies have shown that B cells impact the reduced function of oligodendrocytes [47, 48]. In conjunction with Treg dysfunction, the apoptotic rate of inflammatory leukocytes decreases, resulting in the proliferation of the inflammatory leukocyte population. The excessive accumulation of leukocytes subsequently leads to inflammatory damage within the CNS. However, both T cells and B cells predominantly contribute to the inflammatory stage seen in RRMS. In SPMS, there is a significant decrease in CD14 and CD45 expression, suggesting that neurodegeneration can surpass the extent of neuroinflammation [49].
Microglia/macrophage-derived extracellular vesicles
Microglia, which are considered to be resident macrophages in the CNS, play a crucial role in maintaining self-tolerance and preventing autoimmunity. It has been hypothesized that once leukocytes enter the CNS compartment, microglia may contribute to the promotion of inflammation. In patients with MS, microglial EVs have been found to be significantly upregulated, even in the CIS [50]. During exacerbations, markers associated with microglia/macrophages, such as IB4, showed a significant increase, indicating inflammation mediated by microglia [51]. In an in vitro model, EVs derived from activated microglia/macrophages were shown to impair synaptic function, as evidenced by reduced expression of markers like Syt1 and N1g1 [52]. Additionally, microglia/macrophage EVs might hinder remyelination by inhibiting the accumulation of oligodendrocyte progenitor cells and reducing levels of myelin basic protein (MBP) [53].
Oligodendrocyte-derived extracellular vesicles
Following demyelination, oligodendrocytes play a role in promoting remyelination. Myelin oligodendrocyte glycoprotein (MOG) is secreted by oligodendrocytes in response to demyelination. An elevated level of MOG expression is indicative of increased inflammation. The available evidence suggests that MOG expression in the remitting stage of RRMS is similar to that in healthy individuals but becomes highly expressed during relapsing and progressive stages [54]. Another protein, MBP, also serves as a marker of demyelination. A significant increase in MBP levels in the serum is observed across all stages, whereas healthy individuals do not exhibit such elevation [55]. These findings suggest a potential association between EVs released by oligodendrocytes and the demyelinating response.
Astrocyte-derived extracellular vesicles
Although astrocytes are the primary target of AQP4-IgG in NMOSD, it has also been observed that EVs derived from astrocytes are present in the experimental autoimmune encephalomyelitis (EAE) model of MS. The EVs obtained from EAE mice showed a significantly higher level of glial fibrillary acidic protein (GFAP). Since GFAP is essential for maintaining the cytoskeleton of astrocytes, the increased expression of GFAP in CSF suggests the presence of astrocyte damage and inflammation [56]. However, it is important to note that astrocyte dysfunction may not be the primary mechanism underlying the pathogenesis of MS, even though it is widely accepted as a central process in NMOSD. The subsequent section will provide a more detailed of NMOSD.
Extracellular vesicles with fibrinogen-laden membranes
Fibrinogen-laden EVs are recognized for their significant involvement in inflammation, extending beyond their traditional role in coagulation processes [57]. Within the CN, various neuroglial cells, including oligodendrocytes, astrocytes, and microglia, express receptors for fibrinogen, facilitating their interaction with fibrinogen-laden EVs and subsequently leading to the activation of encephalitogenic CD8+ T cell responses. In the EAE model, mice injected with fibrinogen-enriched EVs demonstrated the development of encephalitogenic T cell responses, while this phenomenon was not observed in the fibrinogen-deficient group. Laboratory confirmation of inflammatory markers such as CXCL10 and CCL2 provided evidence supporting the proposed involvement of the fibrinogen alpha chain in stimulating CD8+ T cells [58]. Furthermore, fibrinogen-laden EVs were observed to directly transfer antigens and pathogenic peptides to antigen-presenting cells (APCs) via major histocompatibility complex (MHC) class I complexes on their surface membrane [59]. In summary, it is hypothesized that fibrinogen-laden EVs may contribute to human leukocyte antigen (HLA) class I disequilibrium and susceptibility to autoimmunity, ultimately playing a role in the development of MS.
Extracellular vesicles with glycolipid membranes
Sulfatide belongs to the sulfoglycolipid group and is associated with myelin formation. Beginning in the endoplasmic reticulum and finishing up in the Golgi apparatus, ceramide is transformed to galactocerebroside which then is sulfated to produce sulfatide. In addition to being a membrane constituent, sulfatide is also associated with protein transportation, intercellular communication, and glial-axon interactions [60]. Recently, a study found that sulfatide mediated immunomodulatory mechanisms in certain conditions such as autoimmunity, infections, and cancers [61]. In MS, a specific sulfatide fraction known as C16:0 exhibited distinct differences compared to those without MS and showed inverse correlation with the expanded disability status scale (EDSS) [62]. These pathways of immune regulation are primarily triggered by the activation of type II natural killer T (NKT) cells which identify sulfatides via CD1d molecules [63]. C16:0 sulfatide exerts an anti-inflammatory role by inhibiting cytokine production. Additionally, the over-functioning of the acid sphingomyelinase enzyme (ASMase), which is usually seen in MS patients, enhances the hydrolysis of other healthy sphingomyelin into ceramide, resulting in a reduction of sphingomyelin and subsequent inflammation [64]. Changes in the quantities, distribution, and metabolism of glycolipids ultimately exacerbate MS.
Protein, peptides, and enzymes
At least 50 CSF proteins showed significant increases compared to healthy individuals [65]. ASMase, responsible for sphingolipid metabolism, was highly expressed in CSF. ASMase is also associated with ASMase-dependent ceramide signaling, which coordinates intracellular signal cascades and governs the activation and proliferation of CD4+ T-cells. ASMase can also induce neuronal mitochondrial dysfunction and axonal damage [64]. Another protein, Apolipoprotein E4 (ApoE4), was found to be enriched in the CSF of MS individuals [65]. ApoE4 is associated with neurodegeneration due to decreased clearance of pathogenic amyloid-β, as commonly seen in many neurological diseases, especially Alzheimer’s disease [66]. This finding may explain the subsequent neurodegeneration observed in SPMS. Other proteins such as KLKB1, DKK3, C6, and S100A9 were also analyzed and found to be responsible for scar healing, homeostasis, and clot formation, as commonly observed as sequelae of demyelination [65]. However, limited data were gathered due to the very small amount of EV extracted from the CSF. Additionally, CSF is not routinely taken in clinical practice.
MicroRNAs
MicroRNA consists of an 18 to 22-nucleotide single-stranded non-coding molecule. MicroRNA genes are generally transcribed by RNA polymerase II within the nucleus and are subsequently processed in the cytoplasm. The mature microRNA is finally transported into the extracellular space in order to control cell-to-cell communication. Circulating microRNAs have been found to have a correlation with various conditions and diseases, even in the case of MS. Many studies have demonstrated the role of microRNA as an inflammatory trigger in serum, and it also confers a degree of immune protection. A study found suppressed function of anti-inflammatory T cells called Foxhead box P3 (FOXP3+) Treg is observed in MS patients. Let-7i is markedly increased in the serum of MS patients and suppresses the function of insulin-like growth factor 1 receptor (IGF1R) and transforming growth factor beta receptor 1 (TGFβR1). IGF1 and TGFβR1 are important for Treg differentiation and clonal expansion, and impairment of both functions subsequently promotes inflammation. Increments of miR-15b-5p, miR-30b-5p, miR-342-3p, and miR-451a were observed in RRMS patients [67]. MiR-15b and miR-23a were found to be targeted at fibroblast growth factor 2 (FGF-2), which is linked to inflammation, demyelination, and remyelination. MiR-23a was also found to be involved with the level of oligodendrocyte differentiation. A higher level of miR-23a meant there was substantial oligodendrocyte damage. MiR-30b and miR-342 had previously been recognized as potential biomarkers for Alzheimer’s disease and Parkinson’s disease [68-71]. The additional association with MS pathology of both miRNAs apparently supported their contribution to neuro-axonal damage in CNS [72]. In contrast, miR-122-5p, miR-196b-5p, miR-301a-3p, and miR-532-5p, which are considered immune protection EVs, are downregulated in MS patients [72]. MiR-122-5p inhibits inflammatory cell proliferation and migration. MiR-196b-5p is secreted from hematopoietic cells in both normal conditions and malignancy. MiR-301a-3p is a critical endogenous regulator of Th17 cell proliferation and maturation. MiR-532-5p can suppress the inflammatory pathways and has previously been reported as a tumor suppressor in ovarian cancer [72].
Not only serum EVs, but also CSF-derived EVs have been investigated in central demyelinating diseases. Upregulation of miR-18a-5p, miR-21, miR-30a-5p, miR-145-5p, miR-150, miR-328, miR-342-3p, miR-374a-5p, miR-645, and Let-7g-5p, together with the down-regulation of miR-106a, miR-132-5p, miR-146a, miR-191, miR-199a-3p, miR-320a-5p, and miR-365 were associated with oligoclonal band production in CSF and inflammation [73, 74]. Increased miR-181c and miR-633 with a reduction in miR-922, were all associated with inflammatory suppression and may be associated with the reactive response to inflammation [75]. Even though there are several studies on MS and miRNAs in the CSF, they do not directly claim the miRNAs found in these studies were EV-associated. Different extraction techniques, study design, and validation procedures contributed to the variability of results in EV studies [76-79].
Several studies have tried to demonstrate the differences of EVs in serum and CSF to explain the exact pathogenesis of MS [54, 80, 81]. Major myelin proteins, including MPB, proteolipid protein, and MOG, were substantially elevated in the serum and CSF in MS patients. However, peripheral blood mononuclear cell (PBMC)-derived EVs, considered as a potential peripheral EV source, failed to cause expression of MOG or any other myelin proteins. In addition, evidence showed that myelin-associated EVs could transmigrate from the CNS into the periphery through the BBB. This result suggested that MS pathology may originate in the CNS and then spread to the periphery [56]. Another study focusing on miRNAs in MS patients found that serum and CSF miRNAs differed from healthy controls and varied across different disease stages. At least twenty-one miRNAs were significantly expressed, mainly upregulated during an MS relapse, in comparison with the clinical remission stage. However, the level of some miRNAs, such as miR-21-5p, miR-142-3p, miR-223-3p, miR-342-3p, miR-423-5p, and let-7f-5p, were also highly expressed in other CNS inflammatory and infectious diseases [82]. The expression of miRNA was associated with pleocytosis and the existence of oligoclonal bands, which were not only unique to MS but also associated with intrathecally immune-mediated processes such as CNS infection and inflammation. However, due to a lack of specificity, using a single miRNA alteration as a surrogate marker for MS may not be applicable, and a combination of miRNA panels may be suitable for this purpose. All data from clinical studies are shown in Table 1 and 2. Table 3 and Table 4 show the data from in vivo and in vitro studies, respectively.
Given the underlying issue of astrocyte dysfunction in NMOSD, individuals with this condition typically show increased expression of astrocyte-associated markers. A recent study has revealed that EVs obtained from CSF, such as GFAP, tetraspanin, haptoglobin-related protein, and C4b binding protein-alpha, were significantly upregulated in NMOSD but not in MS [81]. This discovery suggests that the mechanisms underlying BBB dysfunction differ between MS and NMOSD. Experimental models and human NMOSD lesions have demonstrated a transient disruption of the BBB caused by infiltration of immune cells including macrophages and polymorphonuclear cells (PMNs) with only a few T and B cells, a phenomenon absent in MS [83]. The migration of PMNs into the brain could additionally exacerbate NMOSD lesion formation by allowing additional AQP-4 antibodies and complement to enter the brain. The activation of the complement cascade during astrocyte destruction, in turn, results in the release of the anaphylatoxin C5a, a potent PMN chemoattractant and activator leading to a vicious inflammatory cycle [84].
Alteration of circulating miRNAs may serve as possible biomarkers for NMOSD. A study demonstrated miRNA dysregulation, as shown by an increase in miRNA-122-3p and miR-200a-5p, which occurs in NMOSD, but not in MS [85]. The miR-122-3p targets the MAPK signaling pathway associated with both innate and adaptive autoimmunity [86]. The miR-200a-5p targets the Ras and Wnt signaling pathways, modulating cell proliferation and the inflammatory response. Additionally, the elevation of miR-122-3p is correlated with the increasing EDSS, indicating that miR-122-3p might be a potential biomarker for the prediction of NMOSD relapse. Comparable to other well-defined autoimmune diseases, EV alteration in systemic lupus erythematosus (SLE) is associated with the inflammatory pathway and disease progression. Serum EV miR-21 and miR-155 of SLE patients were upregulated, whereas the expression of miR-146a was down-regulated. Even though the specific mechanism of EVs is unclear, these EVs might activate inflammatory cells via the TLR7 pathway, allowing them to constantly release IFN-α and proinflammatory cytokines, which may contribute to the pathogenesis of SLE. Additionally, one study reported that the alteration of the Ras signaling pathway enhanced inflammatory T cell function and triggered autoimmunity [87]. Hence, the overlap of Ras signaling pathway dysfunction across two diseases suggests that EVs may promote self-intolerance and autoimmunity.
Contrarily, certain microRNAs probably play a protective effect on NMOSD patients. An in vitro study demonstrated that co-culturing miR-129-2-3p with oligodendrocytes and rat optic nerve resulted in a reduction of demyelination [88]. These results affirm the dual role of microRNAs in NMOSD, where they can act both as anti-inflammatory and pro-inflammatory agents.
In the antibody-negative NMOSD group the majority had evidence of myelin oligodendrocyte glycoprotein antibody-associated disorders (MOGAD). However, until now, there has been no published data that demonstrates any association between EVs and MOGAD. Further studies are still required. The roles of EVs in MS and NMOSD were summarized in Fig. 1.
Since the launch of an investigation into the first disease-modifying therapy (DMT) as a therapeutic target for central demyelinating diseases. Recent studies have shown an association between DMTs and the alterations in EV profiles. Focusing on Interferon-β or IFNβ (including IFNβ-1a and IFNβ-1b), IFNβ could lessen the expression of endothelial surface EVs. The increases in platelet and leukocyte EVs confirmed the reduction of transendothelial migration from the periphery into the CNS [42, 89-91]. Fingolimod inhibited S1P receptor activity, causing lymphocyte sequestration in peripheral lymph nodes and the prevention of lymphocyte migration into the CNS. An increase in platelet EVs, leukocyte EVs, and monocyte EVs, was demonstrated in the plasma of fingolimod-treated individuals confirming that fingolimod can hinder immune cells from peripheral transmigration into the CNS [92]. Fingolimod also modified the expression of EVs as shown by a decrease in the expression of endothelium-derived EVs such as CD105, which inhibited the adhesion of inflammatory cells to the BBB [93]. Natalizumab, a humanized monoclonal antibody, modified platelets and lymphocyte EVs, resulting in immune cell suppression and 4-integrin adhesion on activated endothelial surfaces [92]. DMTs which can limit the peripheral inflammatory cells, are preferable treatments since most inflammatory cells originate from outside the CNS rather than within it. Considering that DMTs were responsible for changes in EV levels, the measurement of blood EVs may be useful surrogate-markers for the assessment of therapeutic efficacy in MS. However, more studies are required to add weight to these findings and assess this possibility further. All of these findings are summarized in Table 5.
In contrast, the mainstay treatment of NMOSD is immunosuppressants. In acute exacerbation, intravenous methylprednisolone or plasma exchange is the current major therapy [9]. In the maintenance phase, immunosuppressive drugs including azathioprine, mycophenolate mofetil, methotrexate and rituximab are prescribed to NMOSD patients [9]. However, the data on NMOSD treatment and EVs is scant. Further study in the field is still required.
Currently, Investigators have been focusing on the role of EVs as a drug delivery tool. EVs are naturally produced and seldom induce intolerance, therefore they have been considered as next-generation possible therapeutic strategies. Mesenchymal EVs are one of the possible groups of EVs being explored in therapeutic approaches for MS [94-96]. Several researchers have hypothesized that placental mesenchymal cells could have a protective function in MS, since previous studies showed decreased exacerbating rates of MS in pregnant women, especially in the third trimester, and a rapid emergence in exacerbation rates after delivery [97]. At various points throughout pregnancy, the maternal circulation becomes flushed with a wide range of EVs representing the maternal-fetal interaction. Pregnant women secrete numerous EVs to suppress the immune response caused by the presence of fetal antigens. Approximately 20% of circulating exosomes in the pregnant woman are of placental origin, and their concentration increases during pregnancy, reaching their peak in the third trimester. Placental EVs originated from various kinds of cells. In a normal physiologic state, after the blastocyte adheres to the placenta, fetal trophoblastic cells differentiate into cytotrophoblasts and syncytiotrophoblasts. EVs generated by cytotrophoblasts and syncytiotrophoblasts enhance embryonic and placental development. Martire et al. demonstrated an association between placental EVs and MS patients by analyzing EVs secreted from placental tissue culture. Major EV groups, specifically embryonic, mesenchymal, and hematopoietic stem cells, showed high expressive capacity. EVs from these origins have been reported to inhibit the inflammatory cascade and enhance immune tolerance. Contrarily, the EVs derived from antigen-presenting cells (such as HLA-A/B/C molecules, CD86, and CD1c), T cells (CD2, CD3, CD25), activated immune cells with cytotoxic properties (CD56), and B cells (CD19, CD20) were significantly decreased in pregnant women [98]. Tumor necrosis factor (TNF)-related EVs, specifically Fas ligand (FasL) and TNF-related apoptosis-inducing ligand (TRAIL), suppressed inflammation by triggering apoptosis in peripheral mononuclear cells. Trophoblast-related EVs are also related to Treg proliferation and NF-κB activation inhibition [99]. Steroid hormone concentrations, which include estrogen and progesterone, surge in pregnant women, particularly in the third trimester. Progesterone at high concentrations causes lymphocytes to release progesterone-induced binding factor (PIBF). The findings demonstrated that PIBF continuously rises during pregnancy and declines dramatically after delivery. PIBF is associated with the secretion of anti-inflammatory cytokines from Th2 cells, including IL-4, IL-5, and IL-10 [98, 100]. In addition, EAE mice injected with placental EVs showed a decrease in T cell proliferation, increase Treg cell, and a subsequent decrease in inflammation adding weight to these findings [101, 102]. The same effects were observed in EVs produced from bone marrow, periodontal ligament cells, adipose tissue, and umbilical cord mesenchyme, which raised the levels of protective cytokines, such as IL10, transforming growth factor-beta (TGF-β), and stromal cell-derived factor 1 alpha (SDF-1α), but reduced pro-inflammatory cytokines including tumor necrosis factor-alpha (TNF-α) and NALP-3 [94, 96, 103-109].
The effectiveness of mesenchymal EVs has not only been observed in human-derived mesenchymal EVs, but also in EAE mice treated with Rhesus monkey mesenchymal stromal cell EVs. This treatment showed evidence of stimulation of oligodendrocytes and prevention of the polarization of M2 to M1 microglia, resulting in remyelination [95]. EVs from doxycycline-inducible let-7g transgenic (Let-7Tg) mice also resulted in measurable benefits with regard to inflammatory suppression. Let-7 inhibits CD4+ T cell proliferation, which may contribute to the lower number of CNS-infiltrated Let-7Tg CD4+ T cells. Th17 differentiation and IL1 receptor 1 (IL1R1) activity were suppressed by the Let-7g transgene, resulting in a pro-regenerative environment [110].
Since the goal of improving MS demyelination is to promote remyelination, many EVs were examined to investigate their efficacy with regard to possible effects [111-113]. Intranasally administered miR-219a-5p extracts boosted OPC proliferation in EAE mice resulting in remyelination [114]. IFN-stimulated dendritic cell EV aided remyelination, as indicated by an increase in MBP. One study found that the exposure of aging animals to EV-containing serum from youth animals also increases oligodendrocyte proliferation and promotes remyelination [115]. The experimental model discovered that environmentally enriched EVs primarily containing miR-219 can promote remyelination, but miR-219 deficient EVs cause remyelination failure and inhibit oligodendrocyte progenitor cell proliferation [116, 117]. An
EVs have been shown to be linked to the pathogenesis of immune-mediated central demyelinating disorders, however the actions and presence of individual EVs have been found to vary between different diseases. Platelet and endothelial EVs, as well as activated microglia-derived exosomes, were found to be associated with synaptic and BBB dysfunctions in MS.
This work was supported by the Distinguished Research Professor grant from the National Research Council of Thailand (N42A660301 to SCC); Research Chair Grant from the National Research Council of Thailand (NC) and a Chiang Mai University Center of Excellence Award (NC).
The authors have no competing interests to declare that are relevant to the content of this article.
Extracellular vesicle marker in central demyelinating diseases related to inflammatory pathways from plasma or serum: reports from
Methods of EV detection (specimen) | Major findings of each disorder | Interpretation | Ref | |||||||
---|---|---|---|---|---|---|---|---|---|---|
Multiple sclerosis | Subtypes of multiple sclerosis | NMOSD | Relapsing disease activity | |||||||
RRMS | SPMS | PPMS | CIS | Clinical | Radiologic | |||||
EVs classified by origin | ||||||||||
Endothelial cells | ||||||||||
Flow cytometry (platelet-poor plasma) | ↑ PMP ↑ CD31 ↑ CD62E | ↑ PMP ↑ CD31 ↔ CD62E | ↑ PMP ↑ CD31 ↔ CD62E | ↑ PMP ↑ CD31 ↔ CD62E | ↔ PMP ↑ CD31 ↑ CD62E | NA | NA | NA | An increase in EMPs-CD31 involved in all courses of MS. However, PMP was associated with chronic MS but not with early stage, increased EMP-CD62E was found in the early stage of disease. | [33] |
Flow cytometry and 3T MRI (plasma) | NA | ↑ CD31+/CD51+/CD61+/CD54+ (EMPs) | ↓CD31+/CD51-/CD61-/CD54- (PMPs) | NA | NA | NA | NA | NA | An increase in EMPs was more common in RRMS than in SPMS. PMP levels rose in SPMS but not in SPMS. | [35] |
Flow cytometry, scanning EM (platelet-poor plasma) | ↑ CD105 | NA | NA | NA | NA | NA | NA | NA | Untreated MS had high endothelial microparticles detected from CD105. | [93] |
Flow cytometry (platelet-poor plasma) | NA | ↑ CD51+ | NA | NA | NA | NA | ↑ CD31+ ↑ CD51+ | ↑ CD31+ | CD31+ associated with clinical and radiologic exacerbation of MS, while CD51+ was associated with natural course of MS. | [34] |
Flow cytometry with fluorescent dye (plasma) | NA | NA | NA | NA | NA | NA | ↑ CD54+-monocyte conjugate ↑ CD63E+ | ↑ CD54+-monocyte conjugate | EMP triggered monocyte-CD54+ binding and activation. | [36] |
Flow cytometry (plasma) | NA | NA | NA | NA | NA | NA | ↑ CD31 ↑ CD54 ↑ CD62E | NA | EMP associated with exacerbation of MS. | [90] |
Tandem spectrometry (plasma) | ↑ Fibrinogen alpha-chain | ↑ Fibrinogen alpha-chain | NA | NA | NA | NA | NA | NA | Fibrinogen EV associated with CD8+ activation and caused RRMS. | [58] |
Platelet | ||||||||||
Flow cytometry (platelet-rich plasma) | ↑ CD62p ↑ PMP ↔ Platelet surface IgG ↑ Platelet surface IgM | NA | NA | NA | NA | NA | NA | NA | MS patients had a high level of long-platelet EV activation. | [43] |
Flow cytometry (plasma) | ↑ CD61 ↔ CD45 ↔ CD14 | ↑ CD61 ↑ CD45 ↑ CD14 | ↔ CD61 ↔ CD45 ↔ CD14 | NA | NA | NA | NA | NA | Platelet EV, but not leukocyte EV, played a role in early MS while SPMS reduced all PMP and LMP due to stage changes in neurodegeneration. | [42] |
Leukocytes and microglia | ||||||||||
Flow cytometry (plasma) | ↑ CD61 ↔ CD45 ↔ CD14 | ↑ CD61 ↑ CD45 ↑ CD14 | ↔ CD61 ↔ CD45 ↔ CD14 | NA | NA | NA | NA | NA | Platelet EV, but not leukocyte EV, played a role in early MS while SPMS reduced all PMP and LMP due to stage changes in neurodegeneration. | [42] |
Immunoprecipitation, NTA | ↑ AMPB (T-cell EVs) ↑ FIBB (T-cell EVs) ↑ GELS (B-cell EVs) | NA | NA | NA | NA | NA | ↑ GELS (B-cell EVs) | NA | Both T and B cell-dervied EVs correlated with MS. Only B cell associated with active disease. | [48] |
Oligodendrocytes | ||||||||||
ExoQuick,NTA, Western blot, ELISA (serum) | NA | ↔ MOG | ↑ MOG | NA | NA | NA | ↑ MOG | ↑ MOG | Progressive disease activity correlated with MOG level. | [54] |
Exo-check, immuno-gold TEM | NA | ↑ Myelin basic protein | NA | ↑ Myelin basic protein | ↑ Myelin basic protein | NA | NA | NA | Increased oligodencrocyte-dervied EVs associated with MS in all stages. | [55] |
EVs categorized by EV surface membrane expression | ||||||||||
Fibrinogen | ||||||||||
Tandem spectrometry (plasma) | ↑ Fibrinogen alpha-chain | ↑ Fibrinogen alpha-chain | NA | NA | NA | NA | NA | NA | Fibrinogen EV associated with CD8+ activation and caused RRMS. | [58] |
Glycolipids | ||||||||||
UC, EM, NTA, Negative ion electrospray mass spectrometry (plasma) | ↑ C16:0 sulfatide | ↑ C16:0 sulfatide | ↑ C16:0 sulfatide | NA | NA | NA | NA | NA | Sulfatide C16:0 triggered CD1d and activated T cell. | [62] |
EVs classified by containing substances | ||||||||||
microRNAs | ||||||||||
High throughput NGS (serum) | NA | ↑ miR-15b-5p ↑ miR-30b-5p ↑ miR-342-3p ↑ miR-451a | ↑ miR-127-3p ↑ miR-370-3p ↑ miR-409-3p ↑ miR-432-5p ↑ miR-15b-5p ↑ miR-223-3p ↑ miR-23-3p | ↑ miR-127-3p ↑ miR-370-3p ↑ miR-409-3p ↑ miR-432-5p ↑ miR-15b-5p ↑ miR-223-3p ↑ miR-23-3p | NA | NA | ↓ miR-30b-5p ↓ miR-342-3p ↓ miR-374a-5p ↑ miR-432-5p ↑ miR-433-3p ↑ miR-485-3p | NA | Dysregulated miRNA associated with MS subtypes. | [67] |
NGS (serum) | NA | ↓ miR-122-5p ↓ miR-196b-5p ↓ miR-301a-3p ↓ miR-532-5p | NA | NA | NA | NA | ↓ miR-122-5p ↓ miR-196b-5p ↓ miR-301a-3p ↓ miR-532-5p | ↓ miR-122-5p ↓ miR-196b-5p ↓ miR-301a-3p ↓ miR-532-5p | Decreased miRNA was associated with the exacerbation of disease | [72] |
High throughput NGS (serum) | NA | NA | NA | NA | ↑ miR-126-5p ↑ let-7f-5p ↑ let-7a-5p ↑ miR-23a-3p ↑ miR-223-3p ↓ let-7b-5p ↓ miR-24-3p ↓ let-79-5p ↓ miR-25-3p | NA | NA | NA | CIS patients had different patterns of serum exosomes when compared with HC. | [77] |
miScript miRNA techniques RT-PCR (serum) | NA | NA | ↑ miR-376c-3p ↑ miR-191-5p ↑ miR-26a-5p | ↑ miR-376c-3p ↑ miR-191-5p ↑ miR-26a-5p ↑ miR-128-3p ↑ miR-24-3p | NA | NA | NA | NA | miR-128-3p and miR-24-3p were associated with PPMS. | [78] |
NGS with RT-qPCR. EV detection by CD63, CD81 (serum) | ↔ miR-380-3p ↔ miR-216a-5p ↔ miR-548p ↔ miR-153-3p ↔ miR-448 | ↔ miR-380-3p ↔ miR-216a-5p ↔ miR-548p ↔ miR-153-3p ↔ miR-448 | NA | NA | NA | ↑ miR-122-3p ↓ miR-4424 ↓ miR-6764-3p ↓ miR-412-3p ↓ miR-380-3p ↓ miR-216a-5p ↓ miR-548p ↓ miR-153-3p ↓ miR-448 | ↑ miR-200a-5p | NA | Mir-122-3p was unique in NMOSD and miR-200a-5p correlated with clinical NMOSD relapse. | [85] |
Microarray, Flow cytometry (peripheral blood) | ↑ Let-7i | NA | NA | NA | NA | NA | NA | NA | Let-7i inhibited Treg differentiation by inhibiting IGF1R and TGFBR1. | [79] |
CIS, clinically isolated syndrome; EMP, endothelial microparticles; MS, multiple sclerosis; EM, electron microscope; EV, extracellular vesicles; HC, healthy controls; ICAM1, Intercellular Adhesion Molecule 1; IGF1R, insulin-like growth factor 1; LMP, leukocyte microparticles; MHC, major histocompatibility complex; miRNA, microribonucleic acid; MOG, myelin oligodendrocyte glycoprotein; NGS, next-generation sequencing; NMOSD, neuromyelitis optica spectrum disorder; NTA, nanoparticle tracking analysis; PECAM, Platelet endothelial cell adhesion molecule; PMP, platelet microparticles; PPMS; primary progressive multiple sclerosis; RRMS, relapsing-remitting multiple sclerosis; RT-PCR, reverse transcription polymerase chain reaction; RT-qPCR, quantitative reverse transcription polymerase chain reaction; SPMS, secondary progressive multiple sclerosis; TEM, transmission electron microscope; TGFBR1, Transforming Growth Factor Beta Receptor 1; UC, ultracentrifugation, WM, white matter.
Extracellular vesicle markers in central demyelinating diseases related to inflammatory pathways from csf: reports from
Methods of EV detection | Major findings of each disorder | Interpretation | Ref | |||||||
---|---|---|---|---|---|---|---|---|---|---|
Multiple sclerosis | Subtypes of multiple sclerosis | NMOSD | Relapsing disease activity | |||||||
RRMS | SPMS | PPMS | CIS | Clinical | Radiologic | |||||
EVs classified by origin | ||||||||||
Astrocyte | ||||||||||
Proteosome analysis, western blot | ↔ GFAP ↔ Tetraspanin ↔ Haptoglobin related protein ↔ C4b binding protein alpha ↑ fibronectin | NA | NA | NA | NA | ↑ GFAP ↑ Tetraspanin ↑ Haptoglobin related protein ↑ C4b binding protein alpha ↔ Fibronectin | NA | NA | Exosomes in NMOSD differed from MS. Exosomes played role in BBB dysfunction, inflammatory induction, and astrocyte damage. Exosomes in MS only showed impaired myelination markers but not astrocyte damage. | [81] |
Lymphocytes and microglia | ||||||||||
Flow cytometry with Nanoparticle tracking Analysis | NA | ↔ IB4 | ↔ IB4 | ↔ IB4 | ↔ IB4 | NA | ↓ IB4 ↓ CD19+/CD200+ | ↑ CCR3 ↑ CCR5 ↑ CD4/CCR3 ↑ CD4/CCR5 ↑ CCR3/CCR5 | Clinical relapse was associated with a decrease in naive B cell and microglia EVs. In radiologic relapse, the markers CD4+ and CD8+ T cells were significantly elevated. | [44] |
Slow off-rate modified DNA aptamer assay with cluster grouping analysis | NA | ↑ MMP7 ↑ SERPINA3 ↑ GZMA ↑ CLIC1 ↑ DSG2 ↑ TNFRSF25 ↑ CXCL13 | ↑ MMP7 ↑ SERPINA3 ↑ GZMA ↑ CLIC1 ↓ SPARC ↑ DSG2 ↑ TNFRSF25 | ↑ MMP7 ↑ SERPINA3 ↑ GZMA ↑ CLIC1 ↑ DSG2 ↑ TNFRSF25 ↑ CXCL13 | NA | NA | NA | NA | Microglia and myeloid cell EVs were seen in all phases of MS. The expression of toxic astrocyte-specific markers rises with the duration of MS. | [50] |
Flow cytometry, RT-PCR | NA | ↔ IB4 | NA | ↔ IB4 | ↑ IB4 | NA | ↑ IB4 | NA | Resident microglia EVs were abundant in CIS, also association with in clinical recurrence. | [51] |
Oligodendrocytes | ||||||||||
ExoQuick,NTA, Western blot, ELISA | NA | ↑ MOG | ↑ MOG | NA | NA | NA | ↑ MOG | NA | MOG correlated with MS. | [54] |
EVs classified by containing substances | ||||||||||
Proteins and enzymes | ||||||||||
Flow cytometry, Nanoparticle tracking, immunoblotting | NA | ↑ 50 types of CSF protein | NA | NA | NA | NA | NA | NA | At least 50 proteins increased in CSF of RRMS patients. | [65] |
Liquid chromatography, western blot, flow cytometry | ↑ ASMase | NA | NA | NA | NA | NA | NA | NA | CSF exosomes of MS contained higher ASMase levels. | [64] |
microRNA | ||||||||||
TEM, western blotting, RT-PCR | NA | ↑ Let-7 g-5p ↑ miR-18a-5p ↑ miR-145-5p ↑ miR-374a-5p ↑ miR-150-5p ↑ miR-342-3p ↓ miR-132-5p ↓ miR-320a-5p | NA | NA | NA | NA | NA | NA | Higher anti-inflammatory EV and lower pro-inflammatory associated with MS in order to prevent further demyelination. | [74] |
ASMase, acid sphingomyelinase; CCR3, C-C Motif Chemokine Receptor 3; CCR5, C-C Motif Chemokine Receptor 5; CIS, clinically isolated syndrome. CLIC1, Chloride Intracellular Channel 1; CSF, cerebrospinal fluid; cxcl13, C-X-C Motif Chemokine Ligand 13; DSG2, Desmoglein 2; ELISA, enzyme-linked immunosorbent assay; EV, extracellular vesicles; FITC, Fluorescein isothiocyanate; GFAP, Glial fibrillary acidic protein; GZMA, Granzyme A; HC, healthy controls; IB4, isolectin-B4; MMP7, matrix metalloproteinase 7; MOG, myelin oligodendrocyte glycoprotein; MP, microparticles; MS, multiple sclerosis; NMOSD, neuromyelitis optica spectrum disorder; NTA, nanoparticle tracking analysis; PPMS, primary progressive multiple sclerosis; RRMS, relapsing-remitting multiple sclerosis; RT-PCR, reverse transcription polymerase chain reaction; SERPINA3, Serpin Family A Member 3; SPMS, secondary progressive multiple sclerosis; TEM, transmission electron microscope; TNFRSF25, TNF Receptor Superfamily Member 25.
Targeting extracellular vesicle markers in central demyelinating diseases related to inflammatory pathways: data from
Mechanisms | Model and Methods | Multiple sclerosis | NMOSD | Interpretation | Ref |
---|---|---|---|---|---|
Fibrinogen activation | EAE mice with MOG33-35 injection. EV analysis by flow cytometry | ↑ Cxcl10 ↑ Ccl2 | NA | Fibrinogen EV triggered CD8+ function and caused MS. | [58] |
Myeloid activation | EAE mice with Th1/LPS-primed microglia EV injection | ↑ GFAP ↑ CD86 ↑ iNOS ↑ IL6 ↑ IL1β ↑ COX ↑ CD45 ↑ Iba1+ ↓ CD206 | NA | Microglia EVs were found to be responsible for generating inflammation and suppressing remyelination. | [51] |
Astrocytes | EAE mice with exosome isolation and detected with flow cytometry | ↑ GFAP | NA | Astrocyte-derived exosome associated with demyelination. | [56] |
ASMase, acid sphingomyelinase; Ccl2, Chemokine (C-C motif) ligands 2; CIS, clinically isolated syndrome; COX, cyclooxygenase; Cxcl10, C-X-C Motif Chemokine Ligand 10; EAE, experimental autoimmune encephalitis; EV, extracellular vesicle; GFAP, Glial fibrillary acidic protein; iNOS, inducible nitric oxide synthase; MOG, myelin oligodendrocyte glycoprotein; MP, microparticles; NA, not available; NMOSD, neuromyelitis optica spectrum disorder; PPMS, primary progressive multiple sclerosis; RRMS, relapsing-remitting multiple sclerosis; SPMS, secondary progressive multiple sclerosis.
Targeting extracellular vesicle markers in central demyelinating diseases related to inflammatory processes: data from
Mechanisms | Model and Methods | Multiple sclerosis | NMOSD | Interpretation | Ref |
---|---|---|---|---|---|
Treg cell | Mononuclear cells cultured with Treg extract exosome | ↑ T cell proliferation ↓ Apoptotic rate | NA | Dysregulated Treg induced T cell proliferation and decreased T cell apoptosis causing inflammation. | [49] |
T cell | Human cerebral microvascular endothelial cell + CCL4/CCL5 with TEER measurement | ↓ TEER change | NA | CCL4 and CCL5 enhanced BBB dysfunction. | [45] |
Astrocytes co-cultured with MS patient-derived T cells | ↑ IFN-γ ↑ IL-17A ↑ CCL2 | NA | EVs from astrocytes can stimulate T cells into pro-inflammatory stage. | [46] | |
B cell | Oligodendrocyte from rat brain cultured in exosome enrich extracts | ↑ Cell surface components ↑ External cell membrane components | NA | B cells are the key instigators of the pathogenesis of MS. Key cargo of B cell exosomes in MS derived from cell surface and external cell membrane components. | [47] |
Endothelial cells | Endothelial microvesicle detection from cultured human brain microvascular endothelial cell | ↑ CD4+ ↑ CD8+ | NA | EMP stimulated CD4+ and C8+ T cell proliferation. | [37] |
Human cerebral microvascular endothelial cell + TNFα and measure transendothelial migration assay | ↑ Monocyte transmigration | NA | Endothelial EV enhanced adhesion of monocyte to endothelium and migration of monocyte | [36] | |
Activated microglia | Astrocyte cultured with microglia | ↓ Syt1 ↓ N1g1 | NA | Activated microglia derived exosomes associated with synaptic dysfunction. | [52] |
Platelet and endothelial cells | TEER measurement | ↑ F-actin ↑ VE-adherin ↑ ZO-1 | NA | Platelet and endothelial derived EV triggered BBB dysfunction in MS. | [33] |
Synthetic EV | Microglia cultured with OPC and IL4 | ↑ Argenase 1 ↑ MBP ↑ OPC accumulation | NA | Pro-regenerative exposed microglia-induced remyelination. | [53] |
Microglia cultured with OPC and Th1 inflammatory cytokines | ↑ IL-1a ↑ C1q ↑ IL-1β ↑ iNOS ↓ MBP | NA | Pro-inflammatory exposed microglia inhibited remyelination. | ||
Astrocyte cultured with OPC and microglia derived Th1 inflammatory cytokines | ↓ MBP ↓ OPC accumulation | NA | Astrocyte exposed microglia-derived EV and inflammatory cytokines inhibited OPC. | ||
Adeno-associated virus (AAV) with miR-129-2-3p cultured with oligodencrocyte and rat optic nerve tissue | NA | ↓ | miR-129-2-3p targeting SMAD3 gene can inhibit demyelinating process in NMOSD | [88] |
BBB, blood brain barrier; CCL4, Chemokine (C-C motif) ligands 4; CCL5, Chemokine (C-C motif) ligands 5; CIS, clinically isolated syndrome. EMP, Endothelial microparticles; EV, extracellular vesicles; IL, interleukin; iNOS, inducible nitric oxide synthase; MBP, myelin basic protein; MS, multiple sclerosis; NA, not available; NMOSD, neuromyelitis optica spectrum disorder; OPC, oligodendrocyte progenitor cells; PBMC, peripheral blood mononuclear cells; PPMS, primary progressive multiple sclerosis; RRMS, relapsing-remitting multiple sclerosis; SPMS, secondary progressive multiple sclerosis; TEER, transendothelial electrical resistance; Th1, T helper 1 cell; TNFα, tumor necrosis factor alpha; Treg, regulatory T cells.
Extracellular vesicles markers in central demyelinating diseases and therapeutic strategies: reports from
Treatment | Design and population | Methods | Specimens | EV origins | Major findings in treated group | Interpretation | Ref |
---|---|---|---|---|---|---|---|
Fingolimod | Treated: 19 Control: Untreated MS 15, Healthy: 15 | Scanning electron microscope, flow cytometry | Platelet-free plasma | Endothelium | ↓ CD105 | Fingolimod reduced endothelial MP levels. | [93] |
Fingolimod | Treated: 11 Untreated: 5 Healthy: 8 | Nanoparticle tracking system, cryo-electron microscopy, flow cytometry | plasma | Platelet Leukocyte Monocyte | ↑ EV concentration at 5 hours ↓ Lymphocyte activation at 5 hours | Fingolimod inhibited EV immediately after initiation and activated compensatory mechanisms, resulting in increased EV at 5 hours. | [92] |
IFN-β | Treated: 7 Untreated: 4 | qPCR | Serum | NA | ↑ miR-22-3p ↑ miR-660-5p ↓ miR-486-5p ↓ miR-451a ↓ let-7b-5p ↓ miR-320b ↓ miR-122-5p ↓ miR-215-5p ↓ miR-320d ↓ miR-19-3p ↓ miR-26a-5p ↓ miR-142-3p ↓ miR-146a-5p ↓ miR-15b-3p ↓ miR-23a-3p ↓ miR-223-3p | Many inflammatory related exosomes altered after IFN-β treatment. | [91] |
IFN-β | Treated: 20 Untreated: 24 | Flow cytometry | blood | Platelet Leukocyte | ↑ CD61 (PMP) ↑ CD45 (LMP) ↑ CD14 (LMP) | IFN-β increased PMP and LMP. | [42] |
IFN-β1a | Treated: 30 Untreated: 79 | Flow cytometry | Platelet-poor plasma | Endothelium | ↓ CD31+ (EMP) | IFN-β1a decreased EMP. | [89] |
IFN-β1a | Treated: 16 | Flow cytometry | plasma | Endothelium | ↓ CD31+ at 3, 6, 12 months ↓ CD146+ at 3 months ↓ CD54+ at 3, 6, 12 months ↓ T2-weighted lesion volume | CD31+ and CD54+ could be used as surrogate markers for IFN-β1a treatment. | [39] |
IFN-β1b | Comparing before and after treatment: 11 relapsing MS, 9 remitting MS Healthy: 10 | Cultured BMVEC with EMP and detection by flow cytometry from | plasma | Endothelium | ↓ CD31 ↓ CD54 ↓ CD62E ↓ Transmigration of monocyte ↓ Transmigration of monocyte:EMP | IFN-β1b inhibited the adhesion of lymphocytes and endothelium and prevented T cells entering BBB. | [90] |
Natalizumab | Treated: 20 Untreated: 24 | Flow cytometry | blood | Platelet Leukocyte | ↑ CD61 (PMP) ↑ CD45 (LMP) ↑ CD14 (LMP) | Natalizumab increased PMP and LMP. | [42] |
ASMase, acid sphingomyelinase; BBB, blood brain barrier; BMVEC; brain microvascular endothelial cells; EV, extracellular vesicles; HC, healthy control; IFN-β, interferon-beta; EMP, endothelial microparticle; LMP, leukocyte microparticle; miR, microribonucleic acid; MP, microparticle; MS, multiple sclerosis; NA, not available; PBMC, peripheral blood mononuclear cells; PMP, platelet microparticle; qPCR, quantitative polymerase chain reactio.
Extracellular vesicles markers in central demyelinating diseases and therapeutic strategy: reports from
Treatment | Design and population | EV origins | Major finding in treated group | Interpretation | Ref |
---|---|---|---|---|---|
In vivo studies | |||||
Placental exosomes | EAE mice with placental EV injection | Human placenta | ↓ T cell proliferation | Placental EV suppressed T cell proliferation and inflammation. | [101] |
EAE mice injected with mice placental EV | Mice placenta | ↑ Treg | Placental EV promote Treg population resulting in anti-inflammatory state | [102] | |
Human MSC | EAE mice injected with supernatant of cultured BMSC | Bone marrow | ↑ IL10 ↑ TGFβ ↓ TNFα | BMSC inhibited inflammatory process | [94] |
EAE mice injected with BMSC containing miR-367-3p | Bone marrow | ↑ SLC7A11 ↑ GPX4 ↓ ferroptosis | BMSC-derived EVs suppress ferroptosis | [104] | |
EAE mice injected with BMSC containing miR-23b-3p | Bone marrow | ↓ IL17 ↓ Microglia inflammation | BMSC inhibited inflammatory process | [105] | |
EAE mice injected with BMSC | Bone marrow | ↑ Anti-inflammation: miR-193, miR-146a ↓ Proinflammation: miR-155, miR-21, and miR-326 | BMSC inhibited inflammatory process | [108] | |
EAE mice injected with Chimiric TAxI-peptide-chimeric Umbilical cord MSC-exos | Chimeric Umbilical cord MSC | ↑ Anti-inflammation: IL-4, IL-10, TGF-β, IDO-1 ↓ proinflammation: IL-2, IL-6, IL-17A, IFN-γ, and TNF-α | Chimeric umbilical cord MSCs can suppress inflammatory process of MS | [106] | |
EAE mice injected with Umblical cord MSC containing miR-23a-3p | Umbilical cord MSC | ↑ OPC | Umbilical cord MSC can promote remyelination in MS model | [107] | |
Periodontal ligament stem cell exosomes | EAE mice injected with hPDLSC | Periodontal ligaments | ↑ IL10 ↑ TGFβ ↑ SDF-1α ↓ NALP3 | hPDLSC inhibited inflammatory process. | [103] |
Rhesus monkey MSC | EAE mice injected with MSC | Mesenchymal cells | ↑ MBP ↓ Amyloid-β precursor protein ↑ M2 microglia ↓ M1 microglia | MSC promoted remyelination by promoted oligodendrocyte and inhibited inflammatory microglia. | [95] |
Mouse MSC | EAE mice injected with mouse MSC loaded with FOX3P gene | Bone marrow | ↑ Treg ↑ IL10 | BSC EV promote Treg population and function resulting in anti-inflammatory. | [111] |
M2 macrophage derived exosomes | EAE mice injected with si-lncRNA PVT1 | M2 macrophages | ↑ Treg activity ↓ Th17 | lncRNA PVT1 could suppress inflammation of RRMS. | [112] |
EAE mice injected with miR-21-5p | M2 macrophages | ↓ Th17 | miR-21-5p caused Th17 differentiation silencing. | ||
miR-219a-5p exosomes | EAE mice intranasally injected with miR-219a-5p | miR-219a-5p | ↑ OPC | miR-219a-5p promoted remyelination. | [114] |
Let-7 | EAE mice with Let-7g transgene | Let-7 | ↓ Th17 differentiation ↓ IL1R1 ↓ IL23R ↓ CCR2 ↓ CCR5 | Let-7 exosomes reduced Th17 differentiation by blocking IL1R1 and IL23R and inhibition of CCR3 and CCR5. | [110] |
IFNγ-stimulated dendritic cells | Intranasal administration of IFNγ-stimulated DC exosome with lysolecithin injection | Dendritic cells | ↑ MBP | IFNγ-stimulated dendritic cell exosomes associated with remyelination. | [116] |
Environmental enrichment exosomes | Serum from enriched rat cultured with rat hippocampus with lysolecithin injection | Blood exosome | ↑ MBP | Serum environmental enriched exosomes associated with remyelination. | [117] |
In vitro studies | |||||
IL4 | In vitro study: Microglia cultured with OPC IL4 | IL4 | ↑ Argenase 1 ↑ MBP ↑ OPC accumulation | Pro-regenerative exposed microglia induced remyelination | [53] |
MSC | Umbilical cord MSC and their exosome co-culture with PBMC from MS patients and HC. EV detected by flow cytometry | Mesenchymal stem cells | ↓ PBMC proliferation | MSCs inhibited mononuclear cell proliferation. | [96] |
Umbilical cord MSC and their exosome co-cultured with T cells | Umbilical cord mesenchymal stem cells | ↑ IL10 | MSCs induced anti-inflammatory IL-10 secreting T cell. | [113] | |
Adipose tissue MSC containing miR-29b co-cultured with T cells | Adipose tissue | ↓ IL-4 ↓ IL-17 ↓ Tbx21 ↓ RORγt | Adipose tissue MSC associated with T cell differentiation into anti-inflammatory T cells. | [109] | |
IFNγ-stimulated DC | IFNγ-stimulated DC exosome cultured with mice hippocampus | Dendritic cells | ↑ miR-219 ↑ MBP ↓ Reduced glutathione ↓ PDGFRα ↓ ELOVL7 | IFNγ-stimulated DC exosomes mainly contained miR-219 and were associated with remyelination. | [116] |
Environmental enrichment exosomes | Environmental enrichment exosomes cultured with rat hippocampus | Serum with Environmental enrichment | ↑ MBP ↑ miR-219 | Environmental enriched exosomes mainly contained miR-219 and associated with remyelination. | [117] |
BMSC, bone marrow stem cells; CCR2, C-C Motif Chemokine Receptor 2; CCR5, C-C Motif Chemokine Receptor 5; DC, dendritic cells; EAE, experimental autoimmune encephalitis; ELOVL7, fatty acid elongase 7; EV, extracellular vesicles; hPDLSC, human periodontal ligament stem cells exosome; let-7, lethal-7; IFNγ, interferon gamma; IL, interleukin; IL1R1, Interleukin 1 receptor Type 1;IL23R, Interleukin 23 receptor; lncRNA, long non-coding ribonucleic acid; iNOS, inducible nitric oxide synthase; MBP, myelin basic protein; MHC II, major histocompatibility complex class 2; miR, microribonucleic acid; MSC, mesenchymal cells; NALP3, NLR family pyrin domain containing 3; OPC, oligodendrocyte precursor cell; PDGFRα, platelet derived growth factor receptor alpha; PLP, proteolipid protein; PVT1, Plasmacytoma Variant Translocation 1; RRMS, relapsing remitting multiple sclerosis; SDF-1α, stromal cell-derived factor-1 alpha; si-lncRNA, silencing long non-coding ribonucleic acid; TGFβ, transforming growth factor beta; Th17, T helper 17 cells; TNFα, tumor necrosis factor alpha; Treg, regulatory T cells.