Abstract
Systemic vasculitis is a heterogeneous group of multisystem autoimmune disorders characterized by inflammation of blood vessels. Although many progresses in diagnosis and immunotherapies have been achieved over the past decades, there are still many unanswered questions about vasculitis from pathological understanding to more advanced therapeutics. Extracellular vesicles (EVs) are double-layer phospholipid membrane vesicles harboring various cargoes. EVs can be classified into exosomes, microvesicles (MVs), and apoptotic bodies depending on their size and origin of cellular compartment. EVs can be released by almost all cell types and may be involved in physical and pathological processes including, inflammation and autoimmune responses. In systemic vasculitis, EVs may have pathogenic involvement in inflammation, autoimmune responses, thrombosis, endothelium injury, angiogenesis and intimal hyperplasia. EV-associated redox reaction may also be involved in vasculitis pathogenesis by inducing inflammation, endothelial injury and thrombosis. Additionally, EVs may serve as specific biomarkers for diagnosis or monitoring of disease activity and therapeutic efficacy, i.e. AAV-associated renal involvement. In this review, we have discussed the recent advances of EVs, especially their roles in pathogenesis and clinical involvements in vasculitis.
Keywords: extracellular vesicles, systemic vasculitis, inflammation, autoimmunity, redox
1. Introduction
Systemic vasculitis is a heterogeneous group of multisystem autoimmune disorders, including Kawasaki disease (KD), antineutrophil cytoplasmic autoantibody (ANCA)-associated vasculitis (AAV), Takayasu’s arteritis (TA), Behçet’s disease (BD), Polyarteritis nodosa (PAN), Giant cell arteritis (GCA), etc. [1, 2]. Vasculitis is characterized by inflammation in particular-sized blood vessels and commonly accompanied by thrombosis, blood vessel stenosis/occlusion, aneurysm formation or hemorrhage [3]. Although progress has been achieved for earlier diagnosis and the new immunotherapies in the past few decades, patients still experience premature mortality, relapse, comorbidity and poor quality of life [4]. Therefore, better understanding of etiology and pathogenic mechanisms are still needed in order to find better diagnostic biomarkers and establish more advanced therapeutic strategies.
Extracellular vesicles (EVs) are membrane vesicles released by almost all cell types when they undergo cell activation or programmed cell death [5–7]. Many EVs are in relatively low levels under physiological conditions, while may be elevated in various pathological conditions or diseases, such as inflammation [8], thrombosis [9], cardio-metabolic diseases [10], autoimmune disease [11], sepsis [12] as well as cancer [13]. Brogan et al. reported that children with active systemic vasculitis have significant higher levels of plasma platelet EVs and endothelial EVs compared to those in inactive group and healthy controls [14]. The levels of these EVs are closely associated with disease activity [14]. In our early publication, EVs have been shown to mediate intracellular communication, and participate in vasculitis associated pathological processes [11]. In this review, we mainly focus on the most recent progress of EV generation and the potential roles of EVs in the pathogenic involvements and clinical applications in systemic vasculitis.
2. Recent progress in extracellular vesicle research
EVs are cell-derived vesicles that are enclosed by a membrane lipid bilayer. EVs can be released from almost all cell types, such as platelets, erythrocytes, endothelial cells (ECs), monocytes, neutrophils, lymphocytes, mesenchymal stem cells, and malignant cells when they undergo cell activation or several types of programmed cell death [5–7, 11, 15, 16]. By carrying various bioactive molecules, i.e. proteins, lipids, DNA, messenger RNA, long noncoding RNA, microRNA, and circular RNA [7, 17], EVs can exert various biological functions. According to their size and different cellular compartment biogenesis, EVs can be categorized into three subtypes, including exosomes (30–100 nm in diameter), microvesicles (MVs) or microparticles (MPs) (100–1000 nm in diameter), and apoptotic bodies (1000–5000 nm in diameter) [18–20]. EVs can be found in almost all types of body fluid (e.g. blood, urine, milk, semen, synovial fluid, broncho-alveolar lavage fluid, malignant ascites), feces, and solid organ/tissues [19, 21–24]. Here, we briefly summarize the recent progress in EV generation and EV-associated isolation and detection techniques.
2.1. Extracellular vesicles and programmed cell death
It has long been known that EVs can be released during apoptosis [25]. Recent studies indicate that EVs can also be generated during other types of programmed cell death including necroptosis, pyroptosis and NETosis, in addition to apoptosis. Pyroptosis is an inflammatory programmed cell death mediated by caspase-1-dependent canonical inflammasome pathway and non-caspase-1-dependent (caspase-11 in mice, caspase-4/5 in human) pathways [26–29]. Recent study showed that a heterogeneous population of EVs packaged with Gasdermin D (GSDMD), caspase-1and Fas-associated death domain can be released by pyroptotic monocytes [30, 31]. Caspase-1 activation, potassium efflux, and concomitant calcium influx may directly regulate EV secretion and cargo loading [32]. Furthermore, EVs with both cleaved GSDMD and active caspase 1 can induce vascular endothelial injury [30].
Necroptosis is another form of regulated cell death that generally manifests with morphological features of necrosis. This type of cell death is caspase-independent and mediated by the receptor-interacting serine-threonine kinase 1 (RIPK1)- and RIPK3-mixed lineage kinase domain-like (MLKL) signaling axis [33]. The necroptotic cells can release EVs into extracellular space prior their loss of cell viability or membrane integrity [6, 34–36]. Though mechanisms of necroptotic EV biogenesis are still not fully understood, RIPK3-dependent-MLKL phosphorylation and conformational change are indispensable [6]. Compared with apoptotic bodies, these EVs are smaller and have less DNA content. Besides, they contain phosphorylated MLKL, endosomal sorting complex required for transport III members, mitochondria and other proteins [6, 34–37]. Normally, during apoptosis, Ca2+-dependent translocation of phosphatidylserine (PS) to the outer leaflet of the plasma membrane occurs [38], while the necroptotic PS externalization occurs independently of Ca2+ influx, and may be a direct, local, and proportional consequence of MLKL-mediated [33, 39, 40]. However, PS externalization on the necroptotic EVs mediates their recognition and phagocytosis by phagocytes, and involves in the inflammatory response [34, 36].
Neutrophils are the most abundant circulating leukocytes [41]. NETosis, a unique type of neutrophil cell death, is characterized by nuclear envelope rupture, nuclear chromatin externalization and formation of neutrophil extracellular traps (NETs) [42, 43]. Recent publications indicate that NETotic neutrophils can also release membrane EVs during an early stage of NETosis when cytoskeletal rearrangement takes place [44]. Though mechanisms are still not fully understood, a recent work demonstrates that calcium depletion at high glucose concentrations reduce the activity of peptidylarginine deiminase 4 (PAD4) and promote the vesicular NET release [45]. Interestingly, a recent work demonstrates that neutrophils can bind and uptake EVs from the plasma of AAV patients, resulting in NET formation [46]. Therefore, generation of EVs and NETs may be related in certain conditions.
2.2. Extracellular vesicles and redox biology
Oxidation-reduction (redox) reaction is essential in physiological homeostasis and disorders. Redox signaling is also associated with generation and bio-function of EVs. EV formation can be induced by oxidative stress in erythrocytes [47]. In addition, inhibition of NADPH oxidase and nitric oxide synthase-2 attenuates production of EVs from neutrophils [48]. Generally, main steps of EV biogenesis are as follow: formation of clustered microdomains with cargoes, membrane budding, and fission either at the plasma membrane or at the limiting membrane of the multivesicular endosomes [49]. Redox signaling may be involved in different process during EV generation. Calcium influx is a major inducer of EV release, as elevated cytoplasmic calcium enhances fusion of MVBs with plasma membrane and also promotes plasma membrane blebbing and EV formation [50, 51]. Several calcium channels are redox sensitive [52, 53]. Thom and colleagues have shown that reactive oxygen species (ROS) can induce thiol oxidation of inositol-1,3,5-triphosphate receptors, and that triggers calcium release from the endoplasmic reticulum to the cytoplasm, thus consequently resulting in EV release [54]. Additionally, other bio-molecules with redox-sensitive groups (like thiols on cellular surface, actin cytoskeleton, phospholipid flippases, thiol-rich fusion proteins) are potential targets for regulation of EV biogenesis [55].
Redox reactions may also result in the changes of contents in EVs. Alternatively, cells under oxidative stress can remove oxidized proteins through release of EVs, which can be transferred to neighbor or distant cells, thereby triggering an intercellular oxidative stress response between cells [56]. Similarly, oxidative stress increases the abundance of membrane complement regulatory proteins in EVs [57]. In addition, anti-oxidants may affect the composition of EVs. As a reducing reagent, N-acetylcysteine amide has been shown to inhibit the release of EV-associated of TF and PS from cells exposed to oxidative status, thus decreasing the pro-coagulant properties of EVs [55]. Therefore, altered EV concentrations and composition are associated with redox biology, especially under oxidative stress. Additional research is required to identify the specific redox-dependent mechanisms that are involved in regulation of EV generation, and the biofunction, as these may be potential targets for pharmaceutical modulation of EV signaling.
EVs also serve as massage trafficker to modulate redox reactions. A recent proteomic analysis has reported a list of antioxidant activities and related molecules in EVs derived from endothelial cells [58]. Furthermore, these endothelial-derived EVs contain the enzymatic machinery necessary to synthesize NADPH using blood metabolites to feed different biosynthetic pathways [59]. Additionally, nuclear factor erythroid 2-related factor 2 (Nrf2)-targeting miRNAs or other potential components can be incorporated into EVs, thus contributing to redox regulation through the Nrf2/Antioxidant signaling pathway [60]. EVs and ROS have been reported to be closely interacted with each other [61]. On the one hand, EVs can directly modulate intracellular or extracellular contents of ROS, as they carry anti-oxidant/pro-oxidant machinery that may either produce or scavenge ROS [61]. On the other hand, after interacting with their target cells (indirect effect or horizontal transfer), EVs can also increase or reduce ROS metabolism in target cells by transferring enzymatic components, signaling molecules, and/or regulating gene expression that are involved in regulation of redox responses [61].
Redox and inflammatory changes occur during vascular inflammation and endothelial injury [62, 63]. As major free radicals, ROS and reactive nitrogen species (RNS) are involved in vascular diseases [64]. In vasculitis, imbalance of pro-oxidants/anti-oxidants system was observed. For example, enhanced production of ROS through NADPH oxidase in neutrophils is associated with thrombosis in BD patients [65]. In the following section (section 3.), we have discussed pathological involvements of EV-associated redox signaling in vasculitis.
2.3. Isolation and detection of extracellular vesicles
Growing interests in EV research have led to a focus on the development of more advanced techniques in EV isolation, to obtain EVs with high quality for further analysis and applications. EVs are heterogeneous, there are no “one-size-fits-all” standardized optimal methods available until now [7, 66]. Based on the isolation principle, namely size-, charge-, and affinity-based techniques, modern isolation and separation techniques in the field of EV research can be subdivided into three groups [66]. Thereinto, separation methods like ultracentrifugation or density gradients are still commonly used, while the method of size exclusion chromatography has also been gradually used [67]. General criteria for efficient isolation and purification techniques include being capable of isolating EVs with satisfactory quality and quantity, as well as being fast, automatic, simple, and suitable for further applications.
In the context of EV detection, flow cytometry is the most commonly used technique according to their physical characteristics and surface markers based on fluorescence and scattering signals [7]. Conventional flow cytometry can only detect EVs larger than 200 nm [23]. However, according to differential fluorescence intensity, high-resolution flow cytometer can detect EVs with a diameter of 100nm [68, 69]. Other techniques including electron microscopy, fluorescence-based antibody array system, nanoparticle tracking analysis, confocal fluorescent microscopy, enzyme-linked immunosorbent assay (ELISA), immunoblotting as well as electrophoresis, have been used by our [7, 16, 70, 71] and other groups [72–74]. More importantly, in vivo identification of apoptotic and extracellular vesicle-bound live cells using image-based deep learning has been reported [75]. The authors also identified cell subsets binding with naturally occurring EVs and EV-associated decoration in vivo [75]. This breakthrough favors clarifying the function of EVs binding with different cell types in various tissues under normal and pathological conditions. Furthermore, new technologies such as single extracellular vesicle detection [76], label-free surface-enhanced raman spectroscopy [77], highly efficient magnetic labelling and magnetic resonance imaging (MRI) tracking [78], have also been introduced to the field. These new techniques alone with classical methods together will contribute to better understanding of EVs and their involvement in physiology and pathophysiology of various human diseases, including vasculitis.
In addition to the above discussed basic research progress in EV generation, and technical progress in EV detection, recent studies have also demonstrated new understandings and new roles of EVs in human pathophysiology. Very interestingly, recent publications have reported that EVs can transfer intergenerational information of metabolic disease risk [27, 79] or even transmit intergenerational stress of the paternal environmental experience [80] from parents to their offspring in an epigenetic fashion. These emerging functions of EVs challenge the classical concepts of genetic transmission across generations [27, 79, 80]. Importantly, the new understanding in EVs expands over the classical mechanisms of cellular interaction [81], and these knowledge may change our way of thinking about human pathophysiology in the subcellular scale [81]. In the following sections of this review article, we will focus on the progress and the most recent information about pathophysiological involvement of EVs in vasculitis.
3. Pathological involvement of extracellular vesicles in vasculitis
EVs are known to participate in the pathological processes of vasculitis through different mechanisms [11]. Based on the current understanding of the vasculitis pathogenesis, we have summarized the potential pathologic involvement of EVs in vasculitis (figure 1.).
Fig. 1. Pathological involvements of EVs in vasculitis.
A. EVs and inflammation in vasculitis: EVs are shown to have proinflammatory effect by interacting with recipient cells and inducing the release of proinflammatory mediators, and directly transporting proinflammatory mediators as well. B.EVs and autoimmunity in vasculitis: EVs participate in autoimmune procedures of vasculitis, including autoantigen loading and presenting, ICs formation and deposition and impaired EV-ICs clearance, NETosis and DAMPs associated innate immunity. C.EVs and Thrombosis in vasculitis: EVs exert prothrombotic ability in vasculitis mainly due to the surface expression of TF and PS. Besides, EVs could participate in three basic prerequisites of thrombosis of Virchow’s triad. D. EVs and vascular endothelial injury in vasculitis: Endothelial cell- (EC-), neutrophil- and monocyte-derived EVs could induce vascular endothelial injury. E. EVs and angiogenesis in vasculitis: EVs could contribute to all steps of angiogenesis, including enzymatic degradation of the vessel’s basement membrane, ECs proliferate and migrate to form vascular tube as well as the stabilization and maturation of the newly formed vascular structures by recruiting pericytes. F. EVs and intimal hyperplasia in vasculitis: As vascular smooth muscle cells (VSMCs) are central to intimal hyperplasia, EVs involve in phenotypic changes of VSMCs through transferring of bioactive cargoes, including HMGB1, IL-1β and miRNAs.
Abbreviations: ECs, endothelial cells; C, complement; Casp-1, caspase-1; MPO, myeloperoxidase; MMPs, matrix metalloproteinases; PR3, proteinase 3; MHC, Major Histocompatibility Complex; HMGB1, High Mobility Group protein B1; TCR, T cell receptor; ICs, immune complexes; PS, phosphatidylserine; TF, tissue factor; VSMCs, vascular smooth muscle cells; PDGF, platelet derived growth factor; VEGF, vascular endothelial growth factor; ROS, reactive oxygen species; GSDMD, Gasdermin D; MVs, microvesicles.
3.1. Extracellular vesicles and inflammation in vasculitis
Systemic vasculitis is an autoimmune rheumatic disease that are characterized by vessel inflammation [82]. EVs are shown to have proinflammatory effects by interacting with immune and nonimmune cells, and inducing the release of proinflammatory mediators/cytokines, and by directly transporting proinflammatory mediators [83–86]. For instance, our recent studies demonstrated that EVs can induce the release of TNF-α from cultured peripheral blood mononuclear cells [87, 88], while TNF-α is a pivotal cytokine in the pathogenesis of vasculitis. EVs released by neutrophils can contribute to inflammatory response in AAV by promoting the release of IL-6 and IL-8 and the expression of intercellular adhesion molecule-1 (ICAM-1) by ECs [89]. EVs released by anti-PR3-IgG-activated neutrophils can promote the release of pro-inflammatory cytokines in ECs in vitro [90]. Interestingly, the levels of these pro-inflammatory cytokines released by ECs are also elevated in blood of patients with GPA [90]. Therefore, one may speculate that EVs from neutrophils in GPA might induce the release of pro-inflammatory cytokines from ECs of vasculature, contributing to pathogenesis in patients with GPA. Moreover, B1 receptor-positive endothelial EVs in patients with vasculitis may contribute to the chronic inflammation by inducing neutrophil chemotaxis [91].
On the other hand, EVs can exert proinflammatory effects through transporting cellular receptors, proteins and miRNA, etc. Leukocyte-derived EVs can transfer kinin B1-receptors to glomerular ECs and promote kinin-associated inflammation in vasculitis [92]. Additionally, our previous study indicates that high mobility group box-1 (HMGB1) can be loaded into EVs and released from activated monocytes [71]. As a representative damage-associated molecular patterns (DAMPs), HMGB1-positive EVs may participate in the pathogenesis of inflammatory and autoimmune diseases [71]. Interestingly, EV-associated oxidized proteins can be recognized as DAMP and induce inflammation [93]. Xie and colleagues identified differentially expressed protein profile in serum exosomes of patients with coronary artery aneurysm (CAA) caused by KD compared with healthy controls [94]. Further analysis have shown that many of these differentially expressed proteins (like complement system, immunoglobulin, leucine-rich alpha-2-glycoprotein) are associated with immune responses and inflammation [94]. Recent advances also show that EVs contain differential miRNAs that are related to inflammation. Wang et. al. detected a differential exosomal miRNA profile in KD patients with CAA as compared to healthy controls [95]. Differential exosomal miRNA target genes are mainly involved in IL-6 signaling pathways, indicating that exosomal microRNAs are associated with the pathogenesis and development of CAA through inflammatory mechanisms in KD [95].
More interestingly, as mentioned above, EVs can be released by pyroptotic monocytes [96]. These EVs with GSDMD and caspases-1 could induce vascular endothelial injury [30]. Recent study showed that pyroptosis plays an important role in KD. In KD patients, serum levels of pyroptosis-related proteins are significantly increased [97]. Moreover, THP-1 cells treated with KD sera could induce NLRP3 (NACHT, LRR, and PYD domains-containing protein 3)-dependent endothelial pyroptosis through HMGB1/ RAGE(receptor for advanced glycation end-products)/cathespin B signaling pathways [97]. Thus, EVs, that are derived from pyroptotic cells, might be generated during inflammation and are involved in the pathologic process in vasculitis.
In contrast, EVs may also have anti-inflammatory effects depending on the environmental conditions. Kolonics et al. showed that EVs released from resting neutrophils, but not activated or apoptotic neutrophils, exert anti-inflammatory action by inhibiting production of reactive oxygen species (ROS) and releasing anti-inflammatory cytokines from neutrophils [98]. Additionally, inflammatory stimulation of mesenchymal stromal cells (MSCs) results in the release of EVs with altered protein and nucleic acid composition, along with enhanced anti-inflammatory properties [99]. These observations represent a new paradigm for generation of anti-vasculitis EVs for potential treatment of vasculitis.
Taken together, EVs may contribute to a autoimmune inflammation and proinflammatory milieu in vasculitis either indirectly by triggering recipient cells to release proinflammatory mediators, or directly by EV-associated proinflammatory cellular receptors, proteins, miRNA, etc. Depending on environment, certain EVs may exert anti-inflammatory effect as well. One specific kind of cell-derived EVs may exert opposite effects towards inflammation under different metabolic status. Further studies, particularly in vivo analyses, are needed to verify these delicate interactions.
3.2. Extracellular vesicles and autoimmunity in vasculitis
Elevated levels of circulating EVs have been described in various autoimmune diseases, including autoimmune vasculitis [7, 100, 101]. Our previous publication has systematically reviewed the role of EVs in autoimmune diseases [7, 102]. During EV generation, certain cargoes from nucleus, cytoplasm and cell membrane can be clustered as autoantigens in EVs [102]. Stimulated EVs from GPA patients can produce ROS and carry dsDNA [46]. Moreover, EVs are known to be able to express peptide-MHC complexes, presenting autoantigens and activating immune cell responses [100]. Significant infiltration of neutrophils in/around blood vessels is a representative histopathological finding of vasculitis. In addition, neutrophil-derived EVs also contribute to vasculitis as bioactive autoantigen carriers. As autoantigens, neutrophil proteinase-3 (PR3) or myeloperoxidase (MPO) can induce the production of corresponding autoantibodies and promote the pathogenesis of AAV. Both PR3-positive EVs and MPO-positive EVs have been reported [103, 104], suggesting their potential role to serve as auto-antigens and their contributions to autoimmune responses in AAV. On the other hand, these neutrophil-derived MPO-positive EVs could induce MPO-mediated damage of vascular endothelium [105]. Similarly, the level of MPO-positive EVs expressing both C3a and C5a is significantly higher in AAV patients with renal involvement [106].
Furthermore, neutrophil NETs can also associate with a wide range of granular enzymes, such as MPO [107, 108]. NETs can serve as autoantigens, activating complement system, or acting as DAMPs and inflammasome activators [107]. Recent works have shown significant increase of NETs formation in vasculitis [109–112]. As we discussed, neutrophils undergoing NETosis can also release EVs [44]. Thus, both NETotic EVs and NETs in extracellular milieu may contribute to pathogenesis of vasculitis. Besides, NETs can also serve as a source of extracellular HMGB1 [113]. Our published work indicated that HMGB1 could be associated with EVs [71]. Other study also showed HMGB1 plays an important role in AAV [114–116], Henoch-Schonlein Purpura [117, 118], KD [119, 120] and BD [121]. Thus, both EV- or NET-associated HMGB1 in the extracellular milieu may participate in pathological processes by activating HMGB1 associated innate immunity in vasculitis.
In autoimmune disease, especially systemic lupus erythematosus (SLE), circulating or tissue resident EVs have been reported to form immune complexes (ICs) and lead to autoimmune responses [122–124]. ICs also present in patients with vasculitis and can be found in the serum of children with KD [125], in the kidneys of AAV patients [126], as well as in vessel walls of leukocytoclastic vasculitis [127]. On the one hand, EVs with ICs could activate monocytes, T and B cells in vitro, and stimulate the expression of pro-inflammatory mediators, therefore amplifying the autoimmune responses and tissue damage [123, 128, 129]. Furthermore, EV-associated ICs may hinder phagocytes to recognize and clear these EVs. By carrying CD47, EVs might also inhibit phagocytosis through CD47-signal regulatory protein α (SIRPα) signaling [130]. Thus, proinflammatory EVs and cell debris may accumulate when phagocytosis is dampened, and leading to excessive activation of autoimmune responses. Since phagocytes from AAV patients have impaired function [131], EVs-mediated interruption of phagocyte capacity may contribute to the pathogenesis of vasculitis.
In summary, during EV generation, autoantigens could be clustered in EVs, especially the cargoes of PR3 and MPO during EV generation and/or NET release from neutrophils. Following autoantigen presentation, the corresponding production of autoantibodies, formation and deposition ICs in vasculature induce subsequent autoimmune responses in vasculitis. Besides, attenuated phagocytosis leads to the accumulation of these detrimental EVs, contributing propagation of autoimmune responses in this autoimmune vascular disease.
3.3. Extracellular vesicles and thrombosis in vasculitis
Vascular thrombosis is a common phenomenon in vasculitis and prompts tissue injury. It has long been known that exposure of procoagulant PS and tissue factor (TF) on EVs make them highly potent in their pro-thrombotic functions. Shifting of PS from the inner leaflet to the cell membrane can favor membrane budding and formation of EVs [49]. PS externalization is also associated with the prothrombotic activity of EVs by electrostatic interaction between negatively charged PS and positively charged carboxyglutamic acid in the clotting proteins [132]. To date, PS positive EVs have been reported to be released from cell surface of almost all cell types including platelet, monocytes, lymphocytes, or ECs [133, 134].
TF is an initiator of the extrinsic coagulation cascade. Previous results of our and others have demonstrated that TF can be carried on EVs derived from monocytes [9, 135–137]. TF activity is enhanced by PS and phosphatidylethanolamine (PE) on the membrane surface of their parental cells [138]. Importantly, procoagulant activity of EVs can also be enhanced when PS and TF are co-existed [132] as well as after oxidation of the thiol group of the TF molecules [139]. Besides, monocyte-derived EVs can also increase TF expression on ECs, and decrease the levels of anticoagulant tissue factor pathway inhibitor and thrombomodulin [140]. On the other hand, TF-bearing EVs can also be released from ECs after stimulation by TNF-α [141]. EVs released by TNF-α-activated human ECs can promote vascular inflammation and monocyte migration [142]. Positive feedback mechanisms are present between monocytes, monocyte-derived TF+ EVs, ECs, and EC-derived TF+ EVs, thus propagating prothrombotic conditions in vasculitis. Further investigations are needed to explore their interactions and mechanisms in pathogenesis in vasculitis.
Prothrombotic ability is the first documented pathological feature of membrane EVs that are released from platelets [143], and EVs are associated with thrombogenesis in systemic vasculitis [144]. BD patients have significantly higher levels of CD62P+ platelet EVs as compared to healthy controls [145]. Another recent study showed the increased levels of procoagulant EVs in BD patients [146]. Furthermore, BD patients with a history of thrombosis have the increased levels of total and TF positive EVs, but have a lower proportion of EVs with tissue factor pathway inhibitor, as compared to those without a history of thrombosis [147]. Moreover, TF can be expressed on EVs and NETs released from neutrophils in AAV. ANCA can induce the release of TF-expressing EVs or NETs in C5a-primed neutrophils [148]. TF plays a pivotal role in EV-dependent thrombin generation as implicated by antibody neutralization studies [149]. In clinical settings, Mendoza et al. demonstrated that elevated EV-TF activity in AAV remission is a strong indicator of venous thromboembolism (VTE) independent of renal function, suggesting that EV-TF activity might be a useful biomarker for identifying patients that are prone to develop VTE [150]. Therefore, EVs exert prothrombotic ability in vasculitis mainly due to their surface expression of TF and PS, and may serve as a potential biomarker of VTE in vasculitis patients.
According to Virchow’s triad, vascular endothelial injury, blood flow disturbance, and blood hypercoagulability are three basic prerequisites for thrombosis [151]. Firstly, as discussed above, monocyte- and neutrophil-derived EVs with TF, cleaved GSDMD, active caspase 1 and MPO could mediate vascular endothelial damage in vasculitis. Endothelial injury could also be caused by pro-inflammatory properties of EVs. Secondly, blood vessel stenosis/occlusion and aneurysm formation are common manifestations of vasculitis. Elevated pro-thrombotic EVs with TF and PS can initiate coagulation system, leading to activation of coagulant factors and platelets. These contributors can affect blood flow disturbance. Thirdly, elevated coagulant factors and inflammation-associated hypercoagulability are also involved in blood hypercoagulability. Collectively, EVs could be involved in thrombosis through different mechanisms.
3.4. Extracellular vesicles and vascular endothelial injury in vasculitis
Endothelial activation and injury, characterized by increased adhesion molecule expression in ECs and prothrombotic endothelial phenotype, are central to the pathogenesis of vasculitis [152]. EC-derived EVs are biomarkers of endothelial activation/ dysfunction and for monitoring disease activity in systemic vasculitis [153–157]. Circulating EC-derived EVs can impair endothelium-dependent vasorelaxation and reduce nitric oxide (NO) production and bioavailability [158]. In contrast, endothelial EVs can transfer miR-10a to monocytic cells and inhibit inflammatory signaling [159]. This suggests that altered endothelial EV release may lead to vascular injury under the complex and delicate regulation network.
In addition, EVs, derived from other cell types, also contribute to vascular injury. EVs, derived from PR3-ANCA stimulated neutrophils, can bind and activate endothelium via induction of ROS and thrombin generation [89]. Neutrophil EVs expressing active MPO can induce endothelial injury in vasculitis through activation of MPO-hydrogen peroxide-chloride pathway [105]. By co-presence of acute phase reactant pentraxin-3 (PTX3) and HMGB1, MPO-positive EVs may participate in the pathogenesis of AAV by promoting endothelial activation and vascular inflammation [104]. EVs from apoptotic T cells can increase the level of inducible NO-synthase and cyclooxygenase-2 in blood vessel and promote vascular dysfunction [160].
Furthermore, EVs can also induce endothelium injury through kallikrein-kinin and complement systems. Excessive complement deposition on the endothelium is reported to promote endothelial injury, and the complement-bearing endothelial EVs can be found in vasculitis patients [161]. Endothelial complement fixation rapidly induces neutrophil-endothelial adhesion, which is an early event in neutrophil recruitment into acute inflammatory lesions [162]. In AAV patients, ANCA-stimulated neutrophils exhibit greater ability to activate the alternative complement pathway as compared to the primed neutrophils from healthy controls [163]. AAV patients also have significantly elevated expression level of C3a and C5a on MPO-positive EVs [106]. Therefore, interaction between neutrophils and complement system may propagate vasculitis. Since blocking the kallikrein-kinin system can reduce endothelial complement activation [161], the effect may be due to different mechanisms.
Since EVs can be released during pyroptosis and NETosis as mentioned above, pyroptosis- and NETosis-mediated EVs may also induce endothelium injury. However, few studies have been conducted in these contexts. Recent report revealed that EVs-contained both cleaved GSDMD and active caspase 1, two markers of pyroptosis, could induce vascular endothelial injury [30]. In addition, ANCA-induced NETs can provoke endothelial injury [164]. NET-related endothelial dysfunction or damages are dependent on their associated nuclear histones from their parental cells [165, 166]. These studies indicate contributions of pyroptosis and NETosis, as well as their released EVs and/or NETs to endothelial injury in vasculitis. More investigations are needed to explore the role of EVs in vasculitis in the context of pyroptosis and NETosis.
All in all, EVs (especially derived from ECs, neutrophil and monocyte) can contribute to vascular endothelial injury in vasculitis, through complement activation, ROS production, or extracellular nuclear histone-associated injury. Besides, endothelial hypoxia caused by EV-associated thrombosis and vessel stenosis/occlusion also contribute to vascular endothelial injury. Furthermore, neuropeptides might also participate in EV-induced endothelial injury. In human dermal microvascular endothelial cells, neuropeptide, especially substance P, can increase expression of VCAM-1 which is associated with inflammation and endothelium activation [167]. Since EV-associated neuropeptides have not been reported, further studies on neuropeptides and EVs, as well as their pathological roles in vasculitis are still needed.
3.5. Extracellular vesicles and angiogenesis in vasculitis
Angiogenesis is the formation of new capillaries from pre-existing vasculature, which is an important pathological change in vasculitis [168]. In vasculitis, angiogenesis may represent a compensatory response to the hypoxic environment which leads to stenosis or occlusion of blood vessel [169]. Moreover, newly-formed vessels can further promote inflammation by expressing adhesion molecules, colony-stimulating factors and chemokines [169]. Angiogenesis is a process regulated by the balance between stimulatory and inhibitory signals. Altered levels of angiogenic and anti-angiogenic factors have been described in vasculitis patients [168, 170–172]. Interestingly, EVs derived from differential cell types can deliver either pro- or anti-angiogenic signals to ECs in vasculitis [173].
Vasculogenesis is the differentiation of angioblasts into ECs and formation of vascular labyrinth followed by angiogenesis which is the further expansion of the vascular network [174]. Enzymatic degradation of the basement membrane in vasculature is the first step of sprouting angiogenesis [173]. Matrix metalloproteinases (MMPs)-containing EVs can cause extracellular matrix remodeling that paves the way for endothelium invasion and revascularization [175]. Furthermore, EVs, that are taken up by recipient cells, can induce further production of MMPs and their release via EVs [176]. Secondly, ECs proliferate and migrate to form vascular tube [173]. In this step, EVs are also involved. Activated platelet-derived EVs can promote the proliferation and migration of vascular ECs by inducing overexpression of miR-126 and angiogenic factors, including vascular endothelial growth factor (VEGF), basic fibroblast growth factor (bFGF), or transforming growth factor-β1 (TGF-β1) [177]. Thirdly, pericytes are required for the stabilization and maturation of the newly formed blood vessel [173]. Platelet-derived growth factor (PDGF)-B and PDGF receptor-beta play critical roles in recruitment of vascular pericytes [178]. Given that platelet-derived EVs can trigger angiogenesis by EV-associated cytokines (e.g. VEGF, bFGF, and PDGF) [179], platelet-derived EVs may have the potential to recruit vascular pericytes. Thus, EVs could contribute to all steps of angiogenesis through different ways.
In contrast, EVs may also exert anti-angiogenic effects. Lymphocyte-derived EVs can inhibit angiogenesis by increasing oxidative stress and inhibit VEGF-associated signaling [180]. Oxidized low-density lipoprotein (ox-LDL)-stimulated human monocyte-derived macrophages release EVs that can inhibit the proliferation, migration, and angiogenesis of human coronary artery vascular ECs in vitro [181]. Although the role of EVs in vascular formation is well documented, further investigations to study the specific involvement of endogenous EVs in the process may help us to develop EV-related therapeutic strategy.
3.6. Extracellular vesicles and intimal hyperplasia in vasculitis
Intimal hyperplasia is an important pathological process in vasculitis. In response to vascular injury and inflammation, proliferation, migration and decreased expression of smooth muscle cell markers can occur in vascular smooth muscle cells (VSMCs), resulting in intimal hyperplasia [182, 183]. In this pathological process, various cell types, like dendritic cells (DCs), CD4 T cells and macrophages, recruit and produce cytokines, MMPs, as well as growth factors, contributing to intimal hyperplasia [184, 185].
As VSMCs is crucial to intimal hyperplasia, EVs are involved in phenotypic changes of VSMCs through transferring of bioactive cargoes, including HMGB1, IL-1β and miRNAs. HMGB1 have been reported to be critical regulator of intimal hyperplasia after arterial injury [186, 187]. HMGB1 increases IL-1β production in VSMCs via NLRP3 inflammasome, thus prompting VSMC inflammation [188]. In turn, IL-1β enhances VSMC proliferation and migration via P2Y2 receptor-mediated RAGE expression and HMGB1 release [189]. So HMGB1 and IL-1β can regulate VSMC inflammation, proliferation and migration, which are essential to intimal hyperplasia. Our previous publication indicated that nuclear HMGB1 could be externalized and released with EVs from macrophages [71]. EVs with co-expression of MPO and HMGB1 are also associated with AAV disease activity [104]. IL-1β containing EVs have also been detected [190, 191]. These indicate the potential role of EVs in intimal hyperplasia through transporting HMGB1 and IL-1β to VSMCs. Additionally, transfer of miRNAs by EVs also participate in phenotypic changes of VSMCs. It has been reported that transfer of exosomal miR-21–3p from nicotine-stimulated macrophages to VSMCs can enhance migration and proliferation of VSMCs [192]. Besides, transfer and direct overexpression of miR-221–3p from adipose tissue-secreted EVs dramatically enhance VSMCs proliferation and migration [193]. In contrast, EVs may also inhibit migration and proliferation of VSMCs, indicating a potential therapeutic role in vasculitis. Significant inhibitory effects of adipose MSCs-EVs on VSMC proliferation and migration in vitro have been reported to limit intimal hyperplasia [194]. Furthermore, exosomes derived from human umbilical cord MSCs have also been reported to inhibit intimal hyperplasia in a rat model [195].
Collectively, since VSMCs are central to intimal hyperplasia, EVs are involved in phenotypic changes of VSMCs through transferring of bioactive cargoes, including HMGB1, IL-1β and miRNAs. While EVs can either promote or attenuate intimal hyperplasia. One of the major challenges, that the expanding field of EV research faces, is the highly heterogenous of vesicle subgroups and the complexity of the characteristics, compositions and biological functions of EVs.
4. Clinical involvement of extracellular vesicles in systemic vasculitis
As discussed above, EVs may play pivotal role in the pathogenesis of systemic vasculitis mainly through their effects in inflammation, pro-coagulation, autoimmune processes, endothelial injury, and intimal hyperplasia. In clinical aspect, EVs can also be important to serve as biomarkers for diagnosis, to predict treatment efficacy for disease activity monitoring, and to be a potential therapeutic target in systemic vasculitis. Here, we have summarized the published works regarding the involvement of EVs in vasculitis in Table 1.
Table 1.
Summary of the published works about EVs in systemic vasculitis
EV subtype | Cellular origin | Markers | Associated specific component | Samples | Techniques | Study findings | Ref. | Year |
---|---|---|---|---|---|---|---|---|
MVs | Platelets; ECs | PS, CD42a, P-selectin, E-selectin, CD105, ICAM-1, VCAM-1 | NA | Platelet-poor plasma | Flow cytometry | Increased levels of circulating endothelial and platelet MVs were found in children with active vasculitis. The level of endothelial MVs correlated with disease activity of vasculitis. | [14] | 2004 |
EVs | NA | CD63, CD81 | NA | Platelet-poor plasma | Flow cytometry, nanoparticle tracking analysis | Leukotriene B4 and 5-oxo-eicosatetraenoic acid from EVs of GPA patients can stimulate primed neutrophils. | [46] | 2020 |
MVs | Neutrophils | PS, MPO, PR3, CD18, CD11b, CD66b | NA | Supernatant of cultured neutrophils, platelet-poor plasma | Flow cytometry | ANCAs can stimulate primed neutrophils to release MVs that activate ECs and promote the generation of thrombin. | [89] | 2012 |
EVs | Neutrophils | NA | NA | NA | Flow cytometry, nanoparticle tracking, miRNA screening | EVs released by activated neutrophils can promote the release of pro-inflammatory cytokines in ECs corresponding with the regulatory network of miRNAs/mRNAs comprising both EVs miRNAs and ECs transcripts. | [90] | 2021 |
MVs | ECs | CD105, CD144 | B1-receptors | Platelet-free plasma | Flow cytometry | B1 receptor-positive endothelial MVs in patients with vasculitis may contribute to chronic inflammation by inducing neutrophil chemotaxis. | [91] | 2017 |
MVs | Neutrophils, monocytes | CD45, CD66 | B1-receptors | Plasma, renal biopsies | Flow cytometry, TEM | Patients with vasculitis have higher level of plasma leukocyte derived MVs bearing B1-receptors that can dock on glomerular endothelial cells and induce inflammation. | [92] | 2017 |
Exosomes | NA | CD63, HSP90a, Flotillin | tetranectin , Retinol-binding protein 4, leucine-rich alpha-2-glycoprotein 1, apolipoprotein A-IV | Serum | ExoQuick precipitation, TEM, Western blot | Proteomic profile of serum exosomes from children with CAA caused by KD was established. | [94] | 2019 |
Exosomes | NA | NA | let-7i-3p | Serum | ExoQuick precipitation | Exosomal miRNA (let-7i-3p) is a potential biomarker for CAA patients caused by KD. | [95] | 2019 |
MVs | NA | NA | MPO, PTX3, HMGB1 | Platelet-poor plasma | Flow cytometry | Concentration of MPO-positive MVs is increased in plasma from AAV patients. PTX3 and HMGB1 expressed on MPO-positive MVs were associated with disease activity. | [104] | 2020 |
MVs | NA | NA | MPO, C3a, C5a | Platelet-poor plasma | Flow cytometry | The levels C3a and C5a expressed on MPO-positive MVs was associated with renal involvement and disease activity in AAV patients. | [106] | 2020 |
MVs | Platelets, ECs, neutrophils | PS, CD62E, CD11, CD41 | TF | Platelet-poor plasma | Flow cytometry | Enhanced MV-mediated thrombin generation is associated with thrombotic disease in systemic vasculitis. | [144] | 2011 |
MVs | Platelets | CD42a, CD62P | NA | Plasma | Flow cytometry | BD patients had significantly higher level of CD62P+ platelet MVs compared with healthy controls. | [145] | 2011 |
MVs | NA | PS | NA | Platelet-free plasma | ELISA |
The level of procoagulant MVs increased in BD patients. | [146] | 2016 |
MVs | NA | PS | TF | Platelet-poor plasma | Flow cytometry |
BD patients with a history of thrombosis had increased total and TF-positive MV numbers, but had a lower proportion of issue factor pathway inhibitor positive MVs, compared to those without a history of thrombosis. | [147] | 2016 |
MVs | Neutrophils | PS | TF | Supernatant of cultured neutrophils | Flow cytometry | C5a primed neutrophils activated by ANCAs release TF expressing MVs and NETs, which subsequently activate the coagulation system and generate thrombin. | [148] | 2015 |
MVs | Neutrophils | PS, CD66b | TF | Serum | Flow cytometry | Expression of TF in NETs and neutrophil derived MVs proposes a novel mechanism for the induction of thrombosis and inflammation in active AAV. | [149] | 2014 |
MVs | NA | NA | TF | Platelet free plasma | Centrifugation | Elevated TF activity of MVs and increased anti-Plg in remission are strong indicators of VTE independent of renal function in AAV patients. | [150] | 2019 |
MVs | ECs, leukocytes, platelets | PS, CD62E, CD105, CD42a, CD62P, CD45, CD11b | NA | Platelet-poor plasma | Flow cytometry | Endothelial MVs are elevated in active adult AAV. Endothelial MV level correlate with disease activity and might serve as a marker of endothelial activation and damage. | [153] | 2008 |
MVs | ECs | CD144, CD62E, CD105 | NA | Platelet-poor plasma | Flow cytometry | The increased levels of endothelial MVs have significant correlation with decreased values of flow-mediated dilation, both of which may reflect endothelial dysfunction in child KD. | [155] | 2014 |
MVs | Platelets, leukocytes, ECs | PS, vWF, CD105, CD62e, CD45, CD11b, CD42a, CD62p | NA | Platelet-poor plasma | Flow cytometry | MVs in this CSS patient were markedly elevated at initial presentation and rapidly decreased during follow-up and closely paralleled the course of the Birmingham vasculitis activity score | [156] | 2008 |
MVs | ECs | PS, CD144, CD62E | NA | Platelet-poor plasma | Flow cytometry | Elevated levels of circulating endothelial cells , endothelial MVs, endothelial progenitor cells , VEGF, and angiopoietin 2 occurs during active vasculitis, and both circulating endothelial cells and endothelial MVs can be used to monitor disease activity in children with vasculitis. | [157] | 2010 |
MVs | ECs | CD144, CD105 | C3, C9; CD46, CD55 | Plasma before/after perfusion over primary glomerular endothelial cells | Flow cytometry | Plasma of vasculitis patients had significantly more C3- and C9-positive endothelial MVs and can induced the release of significantly more complement-positive MVs from glomerular endothelial cells. Blockade of the kallikrein-kinin system can reduces endothelial complement activation and thereby the inflammatory response on the endothelium. | [161] | 2019 |
MVs | Neutrophils | PS, CD66 | NA | Supernatant of cultured neutrophils; | Flow cytometry | Neutrophils from AAV patients have a greater ability to activate the alternative complement pathway in vitro compared with those from healthy controls. AND this increased ability could be caused by increased release of MVs. | [163] | 2019 |
MVs | Platelets, ECs, leukocytes | CD41a, CD31, CD45, CD62P | Bioactive proteins | Platelet-free plasma | Flow cytometry | Increased levels of platelet derived MVs may be actively involved in enhancing an acute inflammatory state in AAV. | [198] | 2019 |
MVs | Neutrophils, platelets | CD41a, CD66b | NA | Plasma, supernatant of cultured neutrophils | Centrifugation | Patients with vasculitis have increased level of peripheral MVs derived from neutrophils and platelets compared from healthy controls. | [199] | 2006 |
MVs | ECs | CD144 | miR-145–5p, miR-320a | Serum | Flow cytometry | MicroRNA-145–5p and microRNA-320a encapsulated in endothelial MVs might be involved in inflammatory cytokine regulation and the pathogenesis of vasculitis in acute KD. | [203] | 2018 |
MVs | ECs | PS, CD105, E-selectin, ICAM-1, VCAM-1, CD144, CD31 | NA | Platelet-poor plasma | Flow cytometry | The level of CD105 positive endothelial MVs in the KD group, particularly in the CAA present group, were higher versus healthy control. | [204] | 2015 |
MVs | Platelets, Erythrocytes, neutrophils, monocytes, T cells, B cells, ECs | CD42, CD235, CD66b, CD14, CD3, CD19, CD144 | NA | Platelet-poor plasma | Flow cytometry | The number of KD MVs, mainly derived from ECs and T cells, is significantly increased. MVs may develop from endothelial damage and cell activation. | [205] | 2011 |
MVs | ECs | CD31, CD146 | NA | Platelet-poor plasma | Flow cytometry | Elevated level of endothelial MVs, a biomarker of endothelial cells damage, concomitant with increased levels of TNF-a and IL-6, is seen in patients with KD. | [206] | 2013 |
Exosomes | NA | CD9, CD81, flotillin | Differential proteins | Platelet-poor plasma | Electron microscopy, Western blot, 2Delectrophoresis/MALDI-TOF mass spectrometry | Differential proteome profile was found in patients with CAD caused by KD compared with healthy controls, and the majority of the proteins are involved in the inflammation and coagulation cascades. | [207] | 2016 |
Exosomes | NA | TSG101, CD9, CD81 | miR-1246, miR-4436b-5p, miR-197–3p, miR-671–5p | Serum | ExoQuick precipitation, Western blot, real-time quantitive PCR, microarray | Serum exosomal miRNAs can be candidate diagnostic biomarkers for Kawasaki disease. | [208] | 2017 |
Exosomes | NA | NA | miR-328, miR-575, miR-134, miR-671–5p | Serum | Microarray | Serum exosomal miR-328, miR-575, miR-134 and miR-671–5p may act as potential biomarkers for the diagnosis of KD and the prediction of outcomes for the IVIG therapy by influencing the expression of inflammatory genes. | [209] | 2018 |
Exosomes | NA | CD9, flotillin | Differential proteins | Serum | Electron microscopy, Western blot, 2Delectrophoresis/MALDI-TOF mass spectrometry | Differential proteomic profile of serum exosomes of patients with Kawasaki disease before and after intravenous immunoglobulin therapy was recognized. | [210] | 2017 |
MVs | Platelets | NA | NA | Platelet-poor plasma | Centrifugation, ELISA | Platelet derived MVs can be used as an index to show the degree of platelet activation and the inflammation in children with KD. | [211] | 2019 |
MVs | Platelets | glycoprotein Ib | NA | Platelet-poor plasma | ELISA | Platelet derived MV level before aspirin treatment was significantly higher in acute-phase KD patients than in control group. | [212] | 2014 |
MVs | NA | PS | NA | Platelet-poor plasma | ELISA | Platelet-derived MVs might be used as biomarker of antiplatelet therapy in KD. | [213] | 2017 |
MVs | ECs, T cells, B cells, monocytes | PS, CD144, CD3, CD19, CD14 | NA | Platelet-free plasma | Flow cytometry | Concentrations of endothelial MVs are correlated with inflammation in Takayasu arteritis and may be useful markers to assess disease activity. | [214] | 2019 |
MVs | Platelets | CD62 | NA | Whole blood | Flow cytometry | BD patients with previous thrombosis had a higher percentage of circulating CD62-positive platelets and a higher number of circulating microaggregates than those without thrombosis. However, no significant differences in the number of platelet derived MVs per 5,000 platelets was detected between the two groups. | [222] | 2007 |
MVs | ECs, platelets, leukocytes, neutrophils, monocytes | PS, CD105, CD144, CD41, CD18, CD16b, CD14 | NA | Platelet-poor plasma | Flow cytometry | MV counts and platelet reactivity correlated well with disease activity in GPA. Furthermore, MVs were found to activate vascular endothelial cells and platelets in vitro. | [223] | 2015 |
MVs | ECs | CD144, CD146 | NA | Platelet-free plasma | Flow cytometry | Circulating endothelial MVs could be used as a surrogate marker for subclinical inflammation in Henoch-Schönlein purpura. | [224] | 2011 |
4.1. Extracellular vesicles and renal vasculitis
Systemic vasculitis with renal involvement is common and may become kidney failure in severe cases. Many studies showed EVs play important roles in the pathogenesis of vasculitis-associated renal failure. We have recently reviewed the roles of EVs in pathophysiology and clinical implications in lupus nephritis [102]. AAV mainly affects systemic small vessels and is characterized by the presence of ANCAs in the circulation [196]. Renal involvement is present in the majority of AAV patients (almost 100% in MPA, 70–80% in GPA, 25% in EGPA), and their renal function is closely correlated with survival and the risk of end-stage renal disease in patients [197]. Since there are very few studies about EVs in renal vasculitis that affects large and medium vessels, here, we mainly focused on the studies in AAV (with illustration in Fig. 2).
Fig. 2. Pathological involvements of EVs in renal AAV.
Neutrophil -derived EVs and NETotic EVs could express the ANCA autoantigens (MPO, PR3), TF, kinin B1-receptors or extracellular histones. These EVs are involved in the pathogenesis of renal AAV through promoting autoimmunity, inflammation, thrombosis, endothelial injury and tissue damage. Kinin-receptors antagonists might be used to alleviate vasculitis.
Abbreviations: ANCA, Antineutrophil cytoplasmic autoantibody; AAV, ANCA-associated vasculitis; C, Complement; ECs, Endothelial Cells; EVs, Extracellular Vesicles; MPO, Myeloperoxidase; PAD4, Peptidylarginine deiminase 4; PR3, Proteinase 3; TF, Tissue Factor.
EVs can be detected in the circulation and may be associated with disease severity of renal injury in AAV patients [198]. Leukocyte- and endothelial-derived EVs are important, although many EVs are of platelet origin in patients with ANCA disease [14, 89, 199]. Pauci-immune necrotizing and crescentic glomerulonephritis (GN) is the typical glomerular pathological feature in AAV patient with renal involvement [200]. The most prominent infiltrating cell types in pauci-immune necrotizing GN are neutrophilic granulocytes [201]. ANCA can induce generation of EVs from neutrophils [89, 148]. These EVs expressed a variety of bioactive molecules, including PR3, MPO, and TF, thus promoting autoimmune inflammation of the vessel wall and hypercoagulability in these patients [89, 148]. Additionally, NETs are formed during neutrophil NETosis and are closely associated with EVs [44, 46], as we discussed above. NETs are involved in ANCA-mediated vascular injury and the production of ANCAs, therefore, involved in the pathogenesis of AAV [196]. In renal biopsy of early focal phase ANCA-associated polyangiitis patients, NET-associated components, citrullinated histone, MPO and PAD4 concurrently deposited around fibrinoid necrosis in necrotizing GN, and along an interlobular arterial wall [202]. However, it is not known if EVs from NETotic neutrophils also contain histones, MPO, and PAD4 which can mediate renal AAV. Besides, neutrophil-derived EVs expressing kinin B1-receptors can dock on and transfer kinin B1-receptors to glomerular ECs in kidney biopsies from patients with vasculitis [92]. This may result in a sustained inflammatory response. Furthermore, kinin-receptor antagonists could inhibit the release of complement-positive endothelial EVs and reduce glomerular C3 deposition [161], indicating the role of interrupting transfer of kinin-receptors from neutrophil-EVs to endothelial-EVs in amelioration of vasculitis.
Therefore, by expressing ANCA autoantigens, TF, or kinin B1-receptors, neutrophil-derived EVs are involved in pathogenesis of renal AAV through promoting autoimmune inflammation and hypercoagulation. Moreover, kinin-receptors antagonists might be used for amelioration of vasculitis. In addition, EVs from NETotic neutrophil may also be involved in renal AAV, but further investigation is need.
4.2. Extracellular vesicles in vasculitis diagnosis and disease severity evaluation
Over the past decades, a number of studies have demonstrated the roles of EVs in diagnosis and disease evaluation of vasculitis. In KD, the levels of EC-derived EVs were increased [203–206], particularly in patients with CAA [204]. Differential proteome profile has been found in KD patients with coronary artery dilatation (CAD) as compared with healthy controls, and majority of the EV-associated proteins are involved in inflammation and coagulation cascades [207]. Recently, Wang and colleagues reported that serum exosomal microRNA let-7i-3p could distinguish KD complicated CAA patients from healthy children, as well as virus-infected patients [95]. Furthermore, exosomal microRNAs (miR-1246, miR-4436b-5p, miR-197–3p and miR-671–5p) are differentially expressed between KD patients, other febrile patients and healthy controls, indicating a role as candidate diagnostic biomarkers for KD [208]. Zhang et al also found the differential expression of exosomal miRNA between the healthy children and KD patients, or between KD patients and KD patients following IVIG therapy [209], indicating the role of serum exosomal miRNA as potential biomarkers for diagnosis of KD, and the predictive value for outcomes of the IVIG therapy. Similarly, differential exosomal proteomic profiles have been found in patients with KD before and after intravenous immunoglobulin therapy [210]. Besides, significant higher levels of platelet-derived EVs have been reported in KD children [211], while intravenous immunoglobulin (IVIG) significantly decreased the levels of platelet-derived EVs [205, 212, 213]. Moreover, levels of platelet-derived EVs were significantly higher in IVIG-resistant patients than those in IVIG-effective patients before the IVIG treatment [211].
As for TA patients, Cheng and colleagues showed that the concentrations of EC-derived EVs, but not EVs from T cells, B cells or monocytes, increased significantly in TA patients and are associated positively with disease activity [214]. Moreover, the concentrations of EC-derived EVs increased during disease relapse in four TA patients [214]. However, the authors pointed that since the concentrations of EC-derived EVs also increased in patients with other vasculitis, the use of EC-derived EVs as a diagnostic marker in TA might be limited [214].
Similarly, the levels of EC-derived EVs are also increased in adult patients with active AAV and correlated with disease activity [153]. MPO-positive EVs expressing PTX3 and HMGB1 are significantly higher in AAV patients, and are correlated with disease activity in these patients [104], indicating that PTX3 and HMGB1 expressing MPO-positive EVs might be used as promising biomarkers to monitor disease activity in AAV patients [104]. In the study from Miao et al, the levels of platelet-derived EVs are significantly correlated with disease activity, inflammation, and renal damage in AAV patients [198]. Additionally, compared to patients in remission, differentially expressed proteins in platelet-derived EVs from patients with active disease are associated with chemotaxis, cell adhesion, growth and apoptosis [198]. Overall, EVs from different cellular origins as well as their cargoes could be used as candidate biomarkers in diagnosis and disease activity evaluation for vasculitis including KD, TA and AAV.
4.3. Extracellular vesicles and clinical treatment of vasculitis
Aside from being promising biomarkers, EVs can also be used as novel vehicles containing endogenously and exogenously packaged molecules that exert therapeutic role. EVs harbor many features and molecules from their parental cells. Therefore, they generally have low immunogenicity and toxicity with increased circulating time, and can protect RNA or DNA from hydrolysis by nucleases in blood [8, 215]. Therefore, using drug-loaded EVs as therapeutic vehicles has been used as promising strategy for drug delivery.
One approach is to load exogenous cargoes inside EVs to target endothelium. During vascular inflammation, ICAM-1, which binds to integrin β2 on neutrophil membrane, is upregulated in endothelium through NF-κB signaling pathway [216]. Inspired by this interaction, nanovesicles expressing integrin β2 have been generated [217]. Administration of neutrophil nanovesicles loaded with TPCA-1 (a NF-κB inhibitor) markedly mitigated mouse inflamed vasculature [217]. Moreover, rabies virus glycoprotein-decorated EVs with HMGB1-siRNA have also been reported [218]. Therefore, one may expect to administrate neutrophil-derived EVs loaded with HMGB1-siRNA or specific drugs for inhibition of HMGB1 activation and its associated innate immunity as potential therapeutics.
Another approach is to equip endogenous EVs with specific cargoes. MicroRNA-100 transferred by MSC-derived EVs inhibits in vitro angiogenesis through down-regulation of VEGF [219]. The miRNA-126–3p from human umbilical cord MSC enhances endothelial function in an EV-associated manner in vitro and attenuates intimal hyperplasia in vivo [220]. Interestingly, engineered decoy EVs are generated from genetically modified donor cells which express cytokine receptors without the signaling domain [221]. Expression of cytokine receptors on surface of EVs can decoy the pro-inflammatory cytokines (like TNF-α and IL-6) specifically and significantly attenuate inflammation in vitro and in vivo [221]. Therefore, engineered EVs have been shown great potential to be the next generation biotherapeutics.
5. Conclusions
In summary, we have summarized the latest progress in EV generation and EV-associated isolation and detection techniques in this review. In addition to apoptosis, EVs could also be generated from necroptotic, pyroptotic and NETotic cells. We have also summarized the association between EVs and redox biology. The article also summarized the current literature and provide our interpretation based on our experiences in EV research for over a decade. In the article, we summarized EVs secreted by various cell types possess important biological properties in the pathogenesis of systemic vasculitis, including inflammation, autoimmune responses, thrombosis, endothelium injury, angiogenesis and intimal hyperplasia. Thereinto, redox metabolism involves in many aspects. As for clinical implications, EVs may be used as biomarkers for diagnosis and disease activity monitoring. Importantly, EVs play an important role in AAV associated renal involvement. EVs may also reflect the therapeutic responses of immune therapy in patients with vasculitis, thus might predict the prognosis. Moreover, EVs could also be used as novel vehicles for therapeutic purpose. Further studies including animal experiments will allow us to investigate the specific involvement of endogenous EVs in vasculitis and are necessary to determine when and how EVs can be protective or pathogenic.
Acknowledgements
The authors would like to acknowledge the colleagues in the departments for their help in the preparation of this work.
Funding
The study was supported by the grants from the National Natural Science Foundation of China (Grant No. 81370422), Lupus Research Alliance (416805), and National Institute of Health of the USA (R21AI144838).
Abbreviations:
- AAV
ANCA-associated vasculitis
- ANCA
antineutrophil cytoplasmic autoantibody
- BD
Behçet’s disease
- bFGF
basic fibroblast growth factor
- CAA
coronary artery aneurysm
- CSS
Churg-Strauss Syndrome
- DAMPs
damage-associated molecular patterns
- DCs
dendritic cells
- ECs
endothelial cells
- EGPA
eosinophilic granulomatosis with polyangiitis
- ELISA
enzyme-linked immunosorbent assay
- EVs
extracellular vesicles
- GCA
giant cell arteritis
- GN
glomerulonephritis
- GPA
granulomatosis with polyangiitis
- GSDMD
Gasdermin D
- HMGB1
high mobility group box-1
- ICs
immune complexes
- ICAM-1
intercellular adhesion molecule 1
- IL
interleukin
- IVIG
intravenous immunoglobulin
- KD
Kawasaki disease
- MHC
major histocompatibility complex
- MLKL
mixed lineage kinase domain-like
- MMPs
matrix metalloproteinases
- MPs
microparticles
- MPO
myeloperoxidase
- MPA
microscopic polyangiitis
- MRI
magnetic resonance imaging
- MSCs
mesenchymal stromal cells
- MVs
microvesicles
- NA
not available
- NETs
neutrophil extracellular traps
- NF-κB
nuclear factor-kappaB
- NLRP3
NACHT, LRR, and PYD domains-containing protein 3
- NO
nitric oxide
- Nrf2
Nuclear factor erythroid 2-related factor 2
- ox-LDL
oxidized low-density lipoprotein
- PAD4
peptidylarginine deiminase 4
- PAN
polyarteritis nodosa
- PDGF
platelet derived growth factor
- PE
phosphatidylethanolamine
- PR3
proteinase-3
- PS
phosphatidylserine
- RIPK
receptor-interacting serine-threonine kinase
- ROS
reactive oxygen species
- SIRPα
signal regulatory protein α
- SLE
systemic lupus erythematosus
- TA
Takayasu’s arteritis
- TEM
transmission electron microscopy
- TF
tissue factor
- TGF-β1
transforming growth factor-β1
- TNF-α
tumor necrosis factor-α
- VCAM-1
vascular cell adhesion molecule 1
- VEGF
vascular endothelial growth factor
- VSMCs
vascular smooth muscle cells
- VTE
venous thromboembolism
- vWF
von Willebrand factor
- WG
Wegener’s granulomatosis
Footnotes
Declaration of competing interest
The authors declare that they have no competing interests.
References
- [1].Weyand CM, Goronzy JJ. Medium- and large-vessel vasculitis. The New England journal of medicine, 2003;349:160–9. [DOI] [PubMed] [Google Scholar]
- [2].Seo P, Stone JH. Small-vessel and medium-vessel vasculitis. Arthritis and rheumatism, 2007;57:1552–9. [DOI] [PubMed] [Google Scholar]
- [3].Langford CA. Vasculitis. The Journal of allergy and clinical immunology, 2010;125:S216–25. [DOI] [PubMed] [Google Scholar]
- [4].Basu N, Karabayas M, Pusey C. Prognosis and future developments in vasculitis. Best practice & research Clinical rheumatology, 2018;32:148–65. [DOI] [PubMed] [Google Scholar]
- [5].Bergsbaken T, Fink SL, Cookson BT. Pyroptosis: host cell death and inflammation. Nature reviews Microbiology, 2009;7:99–109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [6].Yoon S, Kovalenko A, Bogdanov K, Wallach D. MLKL, the Protein that Mediates Necroptosis, Also Regulates Endosomal Trafficking and Extracellular Vesicle Generation. Immunity, 2017;47:51–65.e7. [DOI] [PubMed] [Google Scholar]
- [7].Liu ML, Williams KJ, Werth VP. Microvesicles in Autoimmune Diseases. Advances in clinical chemistry, 2016;77:125–75. [DOI] [PubMed] [Google Scholar]
- [8].Buzas EI, György B, Nagy G, Falus A, Gay S. Emerging role of extracellular vesicles in inflammatory diseases. Nature reviews Rheumatology, 2014;10:356–64. [DOI] [PubMed] [Google Scholar]
- [9].Li M, Yu D, Williams KJ, Liu ML. Tobacco smoke induces the generation of procoagulant microvesicles from human monocytes/macrophages. Arteriosclerosis, thrombosis, and vascular biology, 2010;30:1818–24. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [10].Charla E, Mercer J, Maffia P, Nicklin SA. Extracellular vesicle signalling in atherosclerosis. Cellular signalling, 2020;75:109751. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [11].Wu X, Liu Y, Wei W, Liu ML. Extracellular vesicles in autoimmune vasculitis - Little dirts light the fire in blood vessels. Autoimmunity reviews, 2019;18:593–606. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [12].Raeven P, Zipperle J, Drechsler S. Extracellular Vesicles as Markers and Mediators in Sepsis. Theranostics, 2018;8:3348–65. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [13].Bebelman MP, Smit MJ, Pegtel DM, Baglio SR. Biogenesis and function of extracellular vesicles in cancer. Pharmacol Ther, 2018;188:1–11. [DOI] [PubMed] [Google Scholar]
- [14].Brogan PA, Shah V, Brachet C, Harnden A, Mant D, Klein N et al. Endothelial and platelet microparticles in vasculitis of the young. Arthritis and rheumatism, 2004;50:927–36. [DOI] [PubMed] [Google Scholar]
- [15].Harrell CR, Jovicic N, Djonov V, Arsenijevic N, Volarevic V. Mesenchymal Stem Cell-Derived Exosomes and Other Extracellular Vesicles as New Remedies in the Therapy of Inflammatory Diseases. Cells, 2019;8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [16].Folkesson M, Li C, Frebelius S, Swedenborg J, Wågsäter D, Williams KJ et al. Proteolytically active ADAM10 and ADAM17 carried on membrane microvesicles in human abdominal aortic aneurysms. Thrombosis and haemostasis, 2015;114:1165–74. [DOI] [PubMed] [Google Scholar]
- [17].Kim KM, Abdelmohsen K, Mustapic M, Kapogiannis D, Gorospe M. RNA in extracellular vesicles. Wiley interdisciplinary reviews RNA, 2017;8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [18].Greening DW, Simpson RJ. Understanding extracellular vesicle diversity - current status. Expert review of proteomics, 2018;15:887–910. [DOI] [PubMed] [Google Scholar]
- [19].Xu R, Greening DW, Zhu HJ, Takahashi N, Simpson RJ. Extracellular vesicle isolation and characterization: toward clinical application. The Journal of clinical investigation, 2016;126:1152–62. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [20].Raposo G, Stoorvogel W. Extracellular vesicles: exosomes, microvesicles, and friends. The Journal of cell biology, 2013;200:373–83. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [21].Carnino JM, Lee H, Jin Y. Isolation and characterization of extracellular vesicles from Bronchoalveolar lavage fluid: a review and comparison of different methods. Respiratory research, 2019;20:240. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [22].Park KS, Lee J, Lee C, Park HT, Kim JW, Kim OY et al. Sepsis-Like Systemic Inflammation Induced by Nano-Sized Extracellular Vesicles From Feces. Frontiers in microbiology, 2018;9:1735. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [23].van der Pol E, Böing AN, Harrison P, Sturk A, Nieuwland R. Classification, functions, and clinical relevance of extracellular vesicles. Pharmacological reviews, 2012;64:676–705. [DOI] [PubMed] [Google Scholar]
- [24].Mercadal M, Herrero C, López-Rodrigo O, Castells M, de la Fuente A, Vigués F et al. Impact of Extracellular Vesicle Isolation Methods on Downstream Mirna Analysis in Semen: A Comparative Study. International journal of molecular sciences, 2020;21. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [25].Xu X, Lai Y, Hua ZC. Apoptosis and apoptotic body: disease message and therapeutic target potentials. Bioscience reports, 2019;39. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [26].Kesavardhana S, Malireddi RKS, Kanneganti TD. Caspases in Cell Death, Inflammation, and Pyroptosis. Annual review of immunology, 2020;38:567–95. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [27].Li CJ, Fang QH, Liu ML, Lin JN. Current understanding of the role of Adipose-derived Extracellular Vesicles in Metabolic Homeostasis and Diseases: Communication from the distance between cells/tissues. Theranostics, 2020;10:7422–35. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [28].Man SM, Karki R, Kanneganti TD. Molecular mechanisms and functions of pyroptosis, inflammatory caspases and inflammasomes in infectious diseases. Immunological reviews, 2017;277:61–75. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [29].Jorgensen I, Miao EA. Pyroptotic cell death defends against intracellular pathogens. Immunological reviews, 2015;265:130–42. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [30].Mitra S, Exline M, Habyarimana F, Gavrilin MA, Baker PJ, Masters SL et al. Microparticulate Caspase 1 Regulates Gasdermin D and Pulmonary Vascular Endothelial Cell Injury. American journal of respiratory cell and molecular biology, 2018;59:56–64. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [31].Mouasni S, Gonzalez V, Schmitt A, Bennana E, Guillonneau F, Mistou S et al. The classical NLRP3 inflammasome controls FADD unconventional secretion through microvesicle shedding. Cell death & disease, 2019;10:190. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [32].Cypryk W, Nyman TA, Matikainen S. From Inflammasome to Exosome-Does Extracellular Vesicle Secretion Constitute an Inflammasome-Dependent Immune Response? Frontiers in immunology, 2018;9:2188. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [33].Samson AL, Garnish SE, Hildebrand JM, Murphy JM. Location, location, location: A compartmentalized view of TNF-induced necroptotic signaling. Science signaling, 2021;14. [DOI] [PubMed] [Google Scholar]
- [34].Zargarian S, Shlomovitz I, Erlich Z, Hourizadeh A, Ofir-Birin Y, Croker BA et al. Phosphatidylserine externalization, “necroptotic bodies” release, and phagocytosis during necroptosis. PLoS biology, 2017;15:e2002711. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [35].Spencer DM, Dye JR, Piantadosi CA, Pisetsky DS. The release of microparticles and mitochondria from RAW 264.7 murine macrophage cells undergoing necroptotic cell death in vitro. Experimental cell research, 2018;363:151–9. [DOI] [PubMed] [Google Scholar]
- [36].Edry-Botzer L, Gerlic M. Exploding the necroptotic bubble. Cell stress, 2017;1:107–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [37].Li Y, Zhu X, Wang G, Tong H, Su L, Li X. Proteomic analysis of extracellular vesicles released from heat-stroked hepatocytes reveals promotion of programmed cell death pathway. Biomed Pharmacother, 2020;129:110489. [DOI] [PubMed] [Google Scholar]
- [38].Nagata S, Suzuki J, Segawa K, Fujii T. Exposure of phosphatidylserine on the cell surface. Cell death and differentiation, 2016;23:952–61. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [39].Gong YN, Guy C, Olauson H, Becker JU, Yang M, Fitzgerald P et al. ESCRT-III Acts Downstream of MLKL to Regulate Necroptotic Cell Death and Its Consequences. Cell, 2017;169:286–300.e16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [40].Samson AL, Zhang Y, Geoghegan ND, Gavin XJ, Davies KA, Mlodzianoski MJ et al. MLKL trafficking and accumulation at the plasma membrane control the kinetics and threshold for necroptosis. Nature communications, 2020;11:3151. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [41].Papayannopoulos V. Neutrophil extracellular traps in immunity and disease. Nature reviews Immunology, 2018;18:134–47. [DOI] [PubMed] [Google Scholar]
- [42].Frangou E, Vassilopoulos D, Boletis J, Boumpas DT. An emerging role of neutrophils and NETosis in chronic inflammation and fibrosis in systemic lupus erythematosus (SLE) and ANCA-associated vasculitides (AAV): Implications for the pathogenesis and treatment. Autoimmunity reviews, 2019;18:751–60. [DOI] [PubMed] [Google Scholar]
- [43].Li Y, Li M, Weigel B, Mall M, Werth VP, Liu ML. Nuclear envelope rupture and NET formation is driven by PKCα-mediated lamin B disassembly. EMBO reports, 2020;21:e48779. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [44].Thiam HR, Wong SL, Qiu R, Kittisopikul M, Vahabikashi A, Goldman AE et al. NETosis proceeds by cytoskeleton and endomembrane disassembly and PAD4-mediated chromatin decondensation and nuclear envelope rupture. Proceedings of the National Academy of Sciences of the United States of America, 2020;117:7326–37. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [45].S. B V, Shaji S, H. V H, S, A. Y P. Calcium depletion at high glucose concentration promotes vesicle-mediated NET release in response to Staphylococcus aureus. Molecular immunology, 2020;124:211–7. [DOI] [PubMed] [Google Scholar]
- [46].Surmiak M, Gielicz A, Stojkov D, Szatanek R, Wawrzycka-Adamczyk K, Yousefi S et al. LTB(4) and 5-oxo-ETE from extracellular vesicles stimulate neutrophils in granulomatosis with polyangiitis. J Lipid Res, 2020;61:1–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [47].Sudnitsyna J, Skverchinskaya E, Dobrylko I, Nikitina E, Gambaryan S, Mindukshev I. Microvesicle Formation Induced by Oxidative Stress in Human Erythrocytes. Antioxidants (Basel, Switzerland), 2020;9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [48].Thom SR, Bhopale VM, Yang M. Neutrophils generate microparticles during exposure to inert gases due to cytoskeletal oxidative stress. The Journal of biological chemistry, 2014;289:18831–45. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [49].van Niel G, D’Angelo G, Raposo G. Shedding light on the cell biology of extracellular vesicles. Nat Rev Mol Cell Biol, 2018;19:213–28. [DOI] [PubMed] [Google Scholar]
- [50].Bucki R, Bachelot-Loza C, Zachowski A, Giraud F, Sulpice JC. Calcium induces phospholipid redistribution and microvesicle release in human erythrocyte membranes by independent pathways. Biochemistry, 1998;37:15383–91. [DOI] [PubMed] [Google Scholar]
- [51].Savina A, Furlán M, Vidal M, Colombo MI. Exosome release is regulated by a calcium-dependent mechanism in K562 cells. The Journal of biological chemistry, 2003;278:20083–90. [DOI] [PubMed] [Google Scholar]
- [52].Zima AV, Blatter LA. Redox regulation of cardiac calcium channels and transporters. Cardiovasc Res, 2006;71:310–21. [DOI] [PubMed] [Google Scholar]
- [53].More JY, Bruna BA, Lobos PE, Galaz JL, Figueroa PL, Namias S et al. Calcium Release Mediated by Redox-Sensitive RyR2 Channels Has a Central Role in Hippocampal Structural Plasticity and Spatial Memory. Antioxid Redox Signal, 2018;29:1125–46. [DOI] [PubMed] [Google Scholar]
- [54].Thom SR, Bhopale VM, Hu J, Yang M. Increased carbon dioxide levels stimulate neutrophils to produce microparticles and activate the nucleotide-binding domain-like receptor 3 inflammasome. Free radical biology & medicine, 2017;106:406–16. [DOI] [PubMed] [Google Scholar]
- [55].Benedikter BJ, Weseler AR, Wouters EFM, Savelkoul PHM, Rohde GGU, Stassen FRM. Redox-dependent thiol modifications: implications for the release of extracellular vesicles. Cell Mol Life Sci, 2018;75:2321–37. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [56].Yarana C, St Clair DK. Chemotherapy-Induced Tissue Injury: An Insight into the Role of Extracellular Vesicles-Mediated Oxidative Stress Responses. Antioxidants (Basel, Switzerland), 2017;6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [57].Carver KA, Yang D. N-Acetylcysteine Amide Protects Against Oxidative Stress-Induced Microparticle Release From Human Retinal Pigment Epithelial Cells. Investigative ophthalmology & visual science, 2016;57:360–71. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [58].Bodega G, Alique M, Bohórquez L, Ciordia S, Mena MC, Ramírez MR. The Antioxidant Machinery of Young and Senescent Human Umbilical Vein Endothelial Cells and Their Microvesicles. Oxid Med Cell Longev, 2017;2017:7094781. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [59].Bodega G, Alique M, Bohórquez L, Morán M, Magro L, Puebla L et al. Young and Especially Senescent Endothelial Microvesicles Produce NADPH: The Fuel for Their Antioxidant Machinery. Oxid Med Cell Longev, 2018;2018:3183794. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [60].Tian C, Gao L, Zucker IH. Regulation of Nrf2 signaling pathway in heart failure: Role of extracellular vesicles and non-coding RNAs. Free radical biology & medicine, 2021;167:218–31. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [61].Bodega G, Alique M, Puebla L, Carracedo J, Ramírez RM. Microvesicles: ROS scavengers and ROS producers. Journal of extracellular vesicles, 2019;8:1626654. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [62].Arcambal A, Taïlé J, Couret D, Planesse C, Veeren B, Diotel N et al. Protective Effects of Antioxidant Polyphenols against Hyperglycemia-Mediated Alterations in Cerebral Endothelial Cells and a Mouse Stroke Model. Molecular nutrition & food research, 2020;64:e1900779. [DOI] [PubMed] [Google Scholar]
- [63].Gao ZJ, Jiang Q, Chen Q, Xu KM, Liu ZQ, Chen XB et al. [Clinical manifestation and gene analyses of 15 patients with intellectual disability or developmental delay complicated with congenital nystagmus]. Zhonghua er ke za zhi = Chinese journal of pediatrics, 2017;55:824–9. [DOI] [PubMed] [Google Scholar]
- [64].Gao H, Zou YD, Zhang XW, He J, Zhang J, Sun Y et al. Interstitial lung disease in non-sicca onset primary Sjogren’s syndrome: a large-scale case-control study. International journal of rheumatic diseases, 2018;21:1423–9. [DOI] [PubMed] [Google Scholar]
- [65].Emmi G, Becatti M, Bettiol A, Hatemi G, Prisco D, Fiorillo C. Behçet’s Syndrome as a Model of Thrombo-Inflammation: The Role of Neutrophils. Frontiers in immunology, 2019;10:1085. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [66].Liangsupree T, Multia E, Riekkola ML. Modern isolation and separation techniques for extracellular vesicles. J Chromatogr A, 2021;1636:461773. [DOI] [PubMed] [Google Scholar]
- [67].Royo F, Théry C, Falcón-Pérez JM, Nieuwland R, Witwer KW. Methods for Separation and Characterization of Extracellular Vesicles: Results of a Worldwide Survey Performed by the ISEV Rigor and Standardization Subcommittee. Cells, 2020;9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [68].van der Vlist EJ, Nolte-’t Hoen EN, Stoorvogel W, Arkesteijn GJ, Wauben MH. Fluorescent labeling of nano-sized vesicles released by cells and subsequent quantitative and qualitative analysis by high-resolution flow cytometry. Nature protocols, 2012;7:1311–26. [DOI] [PubMed] [Google Scholar]
- [69].Chandler WL, Yeung W, Tait JF. A new microparticle size calibration standard for use in measuring smaller microparticles using a new flow cytometer. Journal of thrombosis and haemostasis : JTH, 2011;9:1216–24. [DOI] [PubMed] [Google Scholar]
- [70].Li CJ, Liu Y, Chen Y, Yu D, Williams KJ, Liu ML. Novel proteolytic microvesicles released from human macrophages after exposure to tobacco smoke. The American journal of pathology, 2013;182:1552–62. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [71].Chen Y, Li G, Liu Y, Werth VP, Williams KJ, Liu ML. Translocation of Endogenous Danger Signal HMGB1 From Nucleus to Membrane Microvesicles in Macrophages. Journal of cellular physiology, 2016;231:2319–26. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [72].Dragovic RA, Gardiner C, Brooks AS, Tannetta DS, Ferguson DJ, Hole P et al. Sizing and phenotyping of cellular vesicles using Nanoparticle Tracking Analysis. Nanomedicine : nanotechnology, biology, and medicine, 2011;7:780–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [73].van der Pol E, Coumans FA, Grootemaat AE, Gardiner C, Sargent IL, Harrison P et al. Particle size distribution of exosomes and microvesicles determined by transmission electron microscopy, flow cytometry, nanoparticle tracking analysis, and resistive pulse sensing. Journal of thrombosis and haemostasis : JTH, 2014;12:1182–92. [DOI] [PubMed] [Google Scholar]
- [74].Maas SL, de Vrij J, van der Vlist EJ, Geragousian B, van Bloois L, Mastrobattista E et al. Possibilities and limitations of current technologies for quantification of biological extracellular vesicles and synthetic mimics. Journal of controlled release : official journal of the Controlled Release Society, 2015;200:87–96. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [75].Kranich J, Chlis NK, Rausch L, Latha A, Schifferer M, Kurz T et al. In vivo identification of apoptotic and extracellular vesicle-bound live cells using image-based deep learning. Journal of extracellular vesicles, 2020;9:1792683. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [76].Wang S, Khan A, Huang R, Ye S, Di K, Xiong T et al. Recent advances in single extracellular vesicle detection methods. Biosensors & bioelectronics, 2020;154:112056. [DOI] [PubMed] [Google Scholar]
- [77].Shin H, Seo D, Choi Y. Extracellular Vesicle Identification Using Label-Free Surface-Enhanced Raman Spectroscopy: Detection and Signal Analysis Strategies. Molecules (Basel, Switzerland), 2020;25. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [78].Han Z, Liu S, Pei Y, Ding Z, Li Y, Wang X et al. Highly efficient magnetic labelling allows MRI tracking of the homing of stem cell-derived extracellular vesicles following systemic delivery. Journal of extracellular vesicles, 2021;10:e12054. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [79].Siklenka K, Erkek S, Godmann M, Lambrot R, McGraw S, Lafleur C et al. Disruption of histone methylation in developing sperm impairs offspring health transgenerationally. Science (New York, NY), 2015;350:aab2006. [DOI] [PubMed] [Google Scholar]
- [80].Chan JC, Morgan CP, Adrian Leu N, Shetty A, Cisse YM, Nugent BM et al. Reproductive tract extracellular vesicles are sufficient to transmit intergenerational stress and program neurodevelopment. Nature communications, 2020;11:1499. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [81].Zhang PL, Liu ML. Extracellular vesicles mediate cellular interactions in renal diseases-Novel views of intercellular communications in the kidney. Journal of cellular physiology, 2021. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [82].Zanatta E, Colombo C, D’Amico G, d’Humières T, Dal Lin C, Tona F. Inflammation and Coronary Microvascular Dysfunction in Autoimmune Rheumatic Diseases. International journal of molecular sciences, 2019;20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [83].Loyer X, Vion AC, Tedgui A, Boulanger CM. Microvesicles as cell-cell messengers in cardiovascular diseases. Circulation research, 2014;114:345–53. [DOI] [PubMed] [Google Scholar]
- [84].Lawson C, Kovacs D, Finding E, Ulfelder E, Luis-Fuentes V. Extracellular Vesicles: Evolutionarily Conserved Mediators of Intercellular Communication. The Yale journal of biology and medicine, 2017;90:481–91. [PMC free article] [PubMed] [Google Scholar]
- [85].Withrow J, Murphy C, Liu Y, Hunter M, Fulzele S, Hamrick MW. Extracellular vesicles in the pathogenesis of rheumatoid arthritis and osteoarthritis. Arthritis research & therapy, 2016;18:286. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [86].Ardoin SP, Shanahan JC, Pisetsky DS. The role of microparticles in inflammation and thrombosis. Scandinavian journal of immunology, 2007;66:159–65. [DOI] [PubMed] [Google Scholar]
- [87].Liu M, Xu H, Bashir M, Sharma M, Werth V. Proinflammatory microvesicles in patients with cutaneous lupus erythematosus. Journal of Investigative Dermatology, 2014;134. [Google Scholar]
- [88].Liu M, Bashir M, Sharma M, Xu H, Anyanwu C. Novel proinflammatory microvesicles that carry LL-37 in patients with cutaneous lupus. Arthritis & Rheumatology, 2013;65. [Google Scholar]
- [89].Hong Y, Eleftheriou D, Hussain AA, Price-Kuehne FE, Savage CO, Jayne D et al. Anti-neutrophil cytoplasmic antibodies stimulate release of neutrophil microparticles. Journal of the American Society of Nephrology : JASN, 2012;23:49–62. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [90].Surmiak M, Kosalka-Węgiel J, Polański S, Sanak M. Endothelial cells response to neutrophil-derived extracellular vesicles miRNAs in anti-PR3 positive vasculitis. Clinical and experimental immunology, 2021. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [91].Mossberg M, Ståhl AL, Kahn R, Kristoffersson AC, Tati R, Heijl C et al. C1-Inhibitor Decreases the Release of Vasculitis-Like Chemotactic Endothelial Microvesicles. Journal of the American Society of Nephrology : JASN, 2017;28:2472–81. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [92].Kahn R, Mossberg M, Ståhl AL, Johansson K, Lopatko Lindman I, Heijl C et al. Microvesicle transfer of kinin B1-receptors is a novel inflammatory mechanism in vasculitis. Kidney international, 2017;91:96–105. [DOI] [PubMed] [Google Scholar]
- [93].Binder CJ, Papac-Milicevic N, Witztum JL. Innate sensing of oxidation-specific epitopes in health and disease. Nature reviews Immunology, 2016;16:485–97. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [94].Xie XF, Chu HJ, Xu YF, Hua L, Wang ZP, Huang P et al. Proteomics study of serum exosomes in Kawasaki disease patients with coronary artery aneurysms. Cardiology journal, 2019;26:584–93. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [95].Wang YF, Lian XL, Zhong JY, Su SX, Xu YF, Xie XF et al. Serum exosomal microRNA let-7i-3p as candidate diagnostic biomarker for Kawasaki disease patients with coronary artery aneurysm. IUBMB life, 2019;71:891–900. [DOI] [PubMed] [Google Scholar]
- [96].Baxter AA, Phan TK, Hanssen E, Liem M, Hulett MD, Mathivanan S et al. Analysis of extracellular vesicles generated from monocytes under conditions of lytic cell death. Scientific reports, 2019;9:7538. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [97].Jia C, Zhang J, Chen H, Zhuge Y, Chen H, Qian F et al. Endothelial cell pyroptosis plays an important role in Kawasaki disease via HMGB1/RAGE/cathespin B signaling pathway and NLRP3 inflammasome activation. Cell death & disease, 2019;10:778. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [98].Kolonics F, Kajdácsi E, Farkas VJ, Veres DS, Khamari D, Kittel Á et al. Neutrophils produce proinflammatory or anti-inflammatory extracellular vesicles depending on the environmental conditions. Journal of leukocyte biology, 2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [99].Harting MT, Srivastava AK, Zhaorigetu S, Bair H, Prabhakara KS, Toledano Furman NE et al. Inflammation-Stimulated Mesenchymal Stromal Cell-Derived Extracellular Vesicles Attenuate Inflammation. Stem Cells, 2018;36:79–90. [DOI] [PubMed] [Google Scholar]
- [100].Turpin D, Truchetet ME, Faustin B, Augusto JF, Contin-Bordes C, Brisson A et al. Role of extracellular vesicles in autoimmune diseases. Autoimmunity reviews, 2016;15:174–83. [DOI] [PubMed] [Google Scholar]
- [101].Katsiougiannis S. Extracellular Vesicles: Evolving Contributors in Autoimmunity. Forum on immunopathological diseases and therapeutics, 2015;6:163–70. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [102].Zhao Y, Wei W, Liu ML. Extracellular vesicles and lupus nephritis - New insights into pathophysiology and clinical implications. Journal of autoimmunity, 2020;115:102540. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [103].Martin KR, Kantari-Mimoun C, Yin M, Pederzoli-Ribeil M, Angelot-Delettre F, Ceroi A et al. Proteinase 3 Is a Phosphatidylserine-binding Protein That Affects the Production and Function of Microvesicles. The Journal of biological chemistry, 2016;291:10476–89. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [104].Manojlovic M, Juto A, Jonasdottir A, Colic J, Vojinovic J, Nordin A et al. Microparticles expressing myeloperoxidase as potential biomarkers in anti-neutrophil cytoplasmic antibody (ANCA)-associated vasculitides (AAV). J Mol Med (Berl), 2020;98:1279–86. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [105].Pitanga TN, de Aragão França L, Rocha VC, Meirelles T, Borges VM, Gonçalves MS et al. Neutrophil-derived microparticles induce myeloperoxidase-mediated damage of vascular endothelial cells. BMC Cell Biol, 2014;15:21. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [106].Antovic A, Mobarrez F, Manojlovic M, Soutari N, De Porta Baggemar V, Nordin A et al. Microparticles Expressing Myeloperoxidase and Complement C3a and C5a as Markers of Renal Involvement in Antineutrophil Cytoplasmic Antibody-associated Vasculitis. The Journal of rheumatology, 2020;47:714–21. [DOI] [PubMed] [Google Scholar]
- [107].Fousert E, Toes R, Desai J. Neutrophil Extracellular Traps (NETs) Take the Central Stage in Driving Autoimmune Responses. Cells, 2020;9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [108].Brinkmann V, Reichard U, Goosmann C, Fauler B, Uhlemann Y, Weiss DS et al. Neutrophil extracellular traps kill bacteria. Science (New York, NY), 2004;303:1532–5. [DOI] [PubMed] [Google Scholar]
- [109].Yamashita K, Takaori-Kondo A, Mizugishi K. Exaggerated neutrophil extracellular trap formation in Kawasaki disease: a key phenomenon behind the outbreak in western countries? Annals of the rheumatic diseases, 2020. [DOI] [PubMed] [Google Scholar]
- [110].Yoshida Y, Takeshita S, Kawamura Y, Kanai T, Tsujita Y, Nonoyama S. Enhanced formation of neutrophil extracellular traps in Kawasaki disease. Pediatric research, 2020;87:998–1004. [DOI] [PubMed] [Google Scholar]
- [111].Akiyama M, Zeisbrich M, Ibrahim N, Ohtsuki S, Berry GJ, Hwang PH et al. Neutrophil Extracellular Traps Induce Tissue-Invasive Monocytes in Granulomatosis With Polyangiitis. Frontiers in immunology, 2019;10:2617. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [112].Le Joncour A, Martos R, Loyau S, Lelay N, Dossier A, Cazes A et al. Critical role of neutrophil extracellular traps (NETs) in patients with Behcet’s disease. Annals of the rheumatic diseases, 2019;78:1274–82. [DOI] [PubMed] [Google Scholar]
- [113].Whittall-García LP, Torres-Ruiz J, Zentella-Dehesa A, Tapia-Rodríguez M, Alcocer-Varela J, Mendez-Huerta N et al. Neutrophil extracellular traps are a source of extracellular HMGB1 in lupus nephritis: associations with clinical and histopathological features. Lupus, 2019;28:1549–57. [DOI] [PubMed] [Google Scholar]
- [114].Wang C, Chang DY, Chen M, Zhao MH. HMGB1 contributes to glomerular endothelial cell injury in ANCA-associated vasculitis through enhancing endothelium-neutrophil interactions. Journal of cellular and molecular medicine, 2017;21:1351–60. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [115].Wang C, Deng H, Gong Y, You R, Chen M, Zhao MH. Effect of high mobility group box 1 on Toll-like receptor 9 in B cells in myeloperoxidase-ANCA-associated vasculitis. Autoimmunity, 2020;53:28–34. [DOI] [PubMed] [Google Scholar]
- [116].Ma YH, Ma TT, Wang C, Wang H, Chang DY, Chen M et al. High-mobility group box 1 potentiates antineutrophil cytoplasmic antibody-inducing neutrophil extracellular traps formation. Arthritis research & therapy, 2016;18:2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [117].Yu SF, Feng WY, Chai SQ, Meng XB, Dou ZX, Zhu H. Down-Regulation of miR-218–5p Promotes Apoptosis of Human Umbilical Vein Endothelial Cells Through Regulating High-Mobility Group Box-1 in Henoch-Schonlein Purpura. The American journal of the medical sciences, 2018;356:64–71. [DOI] [PubMed] [Google Scholar]
- [118].Chen T, Guo ZP, Wang WJ, Qin S, Cao N, Li MM. Increased serum HMGB1 levels in patients with Henoch-Schönlein purpura. Experimental dermatology, 2014;23:419–23. [DOI] [PubMed] [Google Scholar]
- [119].Qian B, Huang H, Cheng M, Qin T, Chen T, Zhao J. Mechanism of HMGB1-RAGE in Kawasaki disease with coronary artery injury. European journal of medical research, 2020;25:8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [120].Ueno K, Ninomiya Y, Hazeki D, Masuda K, Nomura Y, Kawano Y. Disruption of Endothelial Cell Homeostasis Plays a Key Role in the Early Pathogenesis of Coronary Artery Abnormalities in Kawasaki Disease. Scientific reports, 2017;7:43719. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [121].de Souza AW, Perazzio SF, de França NR, Andrade LE, Bijl M, Westra J et al. High mobility group box 1 serum levels are increased in Behçet’s disease, but not associated with disease activity or disease manifestations. Rheumatology (Oxford, England), 2015;54:2151–5. [DOI] [PubMed] [Google Scholar]
- [122].Cloutier N, Tan S, Boudreau LH, Cramb C, Subbaiah R, Lahey L et al. The exposure of autoantigens by microparticles underlies the formation of potent inflammatory components: the microparticle-associated immune complexes. EMBO molecular medicine, 2013;5:235–49. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [123].Burbano C, Villar-Vesga J, Vásquez G, Muñoz-Vahos C, Rojas M, Castaño D. Proinflammatory Differentiation of Macrophages Through Microparticles That Form Immune Complexes Leads to T- and B-Cell Activation in Systemic Autoimmune Diseases. Frontiers in immunology, 2019;10:2058. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [124].Nielsen CT, Østergaard O, Rekvig OP, Sturfelt G, Jacobsen S, Heegaard NH. Galectin-3 binding protein links circulating microparticles with electron dense glomerular deposits in lupus nephritis. Lupus, 2015;24:1150–60. [DOI] [PubMed] [Google Scholar]
- [125].Menikou S, Langford PR, Levin M. Kawasaki Disease: The Role of Immune Complexes Revisited. Frontiers in immunology, 2019;10:1156. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [126].Li X, Zhang W, Yu HJ, Pan XX, Shen PY, Ren H et al. Clinical and pathological study on patients with primary antineutrophil cytoplasmic autoantibody-associated vasculitis with renal immune complex deposition. Journal of clinical rheumatology : practical reports on rheumatic & musculoskeletal diseases, 2015;21:3–9. [DOI] [PubMed] [Google Scholar]
- [127].Claudy A. Pathogenesis of leukocytoclastic vasculitis. Eur J Dermatol, 1998;8:75–9. [PubMed] [Google Scholar]
- [128].Burbano C, Rojas M, Vásquez G, Castaño D. Microparticles That Form Immune Complexes as Modulatory Structures in Autoimmune Responses. Mediators of inflammation, 2015;2015:267590. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [129].Burbano C, Villar-Vesga J, Orejuela J, Muñoz C, Vanegas A, Vásquez G et al. Potential Involvement of Platelet-Derived Microparticles and Microparticles Forming Immune Complexes during Monocyte Activation in Patients with Systemic Lupus Erythematosus. Frontiers in immunology, 2018;9:322. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [130].Barclay AN, Van den Berg TK. The interaction between signal regulatory protein alpha (SIRPα) and CD47: structure, function, and therapeutic target. Annual review of immunology, 2014;32:25–50. [DOI] [PubMed] [Google Scholar]
- [131].Johansson Å C, Ohlsson S, Pettersson Å, Bengtsson AA, Selga D, Hansson M et al. Impaired phagocytosis and reactive oxygen species production in phagocytes is associated with systemic vasculitis. Arthritis research & therapy, 2016;18:92. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [132].Owens AP 3rd, Mackman N. Microparticles in hemostasis and thrombosis. Circ Res, 2011;108:1284–97. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [133].Niccolai E, Emmi G, Squatrito D, Silvestri E, Emmi L, Amedei A et al. Microparticles: Bridging the Gap between Autoimmunity and Thrombosis. Seminars in thrombosis and hemostasis, 2015;41:413–22. [DOI] [PubMed] [Google Scholar]
- [134].Tripisciano C, Weiss R, Eichhorn T, Spittler A, Heuser T, Fischer MB et al. Different Potential of Extracellular Vesicles to Support Thrombin Generation: Contributions of Phosphatidylserine, Tissue Factor, and Cellular Origin. Scientific reports, 2017;7:6522. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [135].Wang GH, Lu J, Ma KL, Zhang Y, Hu ZB, Chen PP et al. The Release of Monocyte-Derived Tissue Factor-Positive Microparticles Contributes to a Hypercoagulable State in Idiopathic Membranous Nephropathy. Journal of atherosclerosis and thrombosis, 2019;26:538–46. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [136].Liu ML, Reilly MP, Casasanto P, McKenzie SE, Williams KJ. Cholesterol enrichment of human monocyte/macrophages induces surface exposure of phosphatidylserine and the release of biologically-active tissue factor-positive microvesicles. Arteriosclerosis, thrombosis, and vascular biology, 2007;27:430–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [137].Kasthuri RS, Glover SL, Jonas W, McEachron T, Pawlinski R, Arepally GM et al. PF4/heparin-antibody complex induces monocyte tissue factor expression and release of tissue factor positive microparticles by activation of FcγRI. Blood, 2012;119:5285–93. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [138].Rosas M, Slatter DA, Obaji SG, Webber JP, Alvarez-Jarreta J, Thomas CP et al. The procoagulant activity of tissue factor expressed on fibroblasts is increased by tissue factor-negative extracellular vesicles. PloS one, 2020;15:e0240189. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [139].Ahamed J, Versteeg HH, Kerver M, Chen VM, Mueller BM, Hogg PJ et al. Disulfide isomerization switches tissue factor from coagulation to cell signaling. Proceedings of the National Academy of Sciences of the United States of America, 2006;103:13932–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [140].Aharon A, Tamari T, Brenner B. Monocyte-derived microparticles and exosomes induce procoagulant and apoptotic effects on endothelial cells. Thrombosis and haemostasis, 2008;100:878–85. [DOI] [PubMed] [Google Scholar]
- [141].Holnthoner W, Bonstingl C, Hromada C, Muehleder S, Zipperle J, Stojkovic S et al. Endothelial Cell-derived Extracellular Vesicles Size-dependently Exert Procoagulant Activity Detected by Thromboelastometry. Scientific reports, 2017;7:3707. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [142].Hosseinkhani B, van den Akker NMS, Molin DGM, Michiels L. (Sub)populations of extracellular vesicles released by TNF-α -triggered human endothelial cells promote vascular inflammation and monocyte migration. Journal of extracellular vesicles, 2020;9:1801153. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [143].Wolf P. The nature and significance of platelet products in human plasma. British journal of haematology, 1967;13:269–88. [DOI] [PubMed] [Google Scholar]
- [144].Eleftheriou D, Hong Y, Klein NJ, Brogan PA. Thromboembolic disease in systemic vasculitis is associated with enhanced microparticle-mediated thrombin generation. Journal of thrombosis and haemostasis : JTH, 2011;9:1864–7. [DOI] [PubMed] [Google Scholar]
- [145].Macey M, Hagi-Pavli E, Stewart J, Wallace GR, Stanford M, Shirlaw P et al. Age, gender and disease-related platelet and neutrophil activation ex vivo in whole blood samples from patients with Behçet’s disease. Rheumatology (Oxford, England), 2011;50:1849–59. [DOI] [PubMed] [Google Scholar]
- [146].Mejía JC, Ortiz T, Tàssies D, Solanich X, Vidaller A, Cervera R et al. Procoagulant microparticles are increased in patients with Behçet’s disease but do not define a specific subset of clinical manifestations. Clinical rheumatology, 2016;35:695–9. [DOI] [PubMed] [Google Scholar]
- [147].Khan E, Ambrose NL, Ahnström J, Kiprianos AP, Stanford MR, Eleftheriou D et al. A low balance between microparticles expressing tissue factor pathway inhibitor and tissue factor is associated with thrombosis in Behçet’s Syndrome. Scientific reports, 2016;6:38104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [148].Huang YM, Wang H, Wang C, Chen M, Zhao MH. Promotion of hypercoagulability in antineutrophil cytoplasmic antibody-associated vasculitis by C5a-induced tissue factor-expressing microparticles and neutrophil extracellular traps. Arthritis & rheumatology (Hoboken, NJ), 2015;67:2780–90. [DOI] [PubMed] [Google Scholar]
- [149].Kambas K, Chrysanthopoulou A, Vassilopoulos D, Apostolidou E, Skendros P, Girod A et al. Tissue factor expression in neutrophil extracellular traps and neutrophil derived microparticles in antineutrophil cytoplasmic antibody associated vasculitis may promote thromboinflammation and the thrombophilic state associated with the disease. Ann Rheum Dis, 2014;73:1854–63. [DOI] [PubMed] [Google Scholar]
- [150].Mendoza CE, Brant EJ, McDermott ML, Froment A, Hu Y, Hogan SL et al. Elevated Microparticle Tissue Factor Activity Differentiates Patients With Venous Thromboembolism in Antineutrophil Cytoplasmic Autoantibody Vasculitis. Kidney international reports, 2019;4:1617–29. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [151].Bagot CN, Arya R. Virchow and his triad: a question of attribution. British journal of haematology, 2008;143:180–90. [DOI] [PubMed] [Google Scholar]
- [152].Brogan P, Eleftheriou D. Vasculitis update: pathogenesis and biomarkers. Pediatric nephrology (Berlin, Germany), 2018;33:187–98. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [153].Erdbruegger U, Grossheim M, Hertel B, Wyss K, Kirsch T, Woywodt A et al. Diagnostic role of endothelial microparticles in vasculitis. Rheumatology (Oxford, England), 2008;47:1820–5. [DOI] [PubMed] [Google Scholar]
- [154].Tian J, Lv HT, An XJ, Ling N, Xu F. Endothelial microparticles induce vascular endothelial cell injury in children with Kawasaki disease. European review for medical and pharmacological sciences, 2016;20:1814–8. [PubMed] [Google Scholar]
- [155].Ding YY, Ren Y, Feng X, Xu QQ, Sun L, Zhang JM et al. Correlation between brachial artery flow-mediated dilation and endothelial microparticle levels for identifying endothelial dysfunction in children with Kawasaki disease. Pediatric research, 2014;75:453–8. [DOI] [PubMed] [Google Scholar]
- [156].Kümpers P, Erdbrügger U, Grossheim M, Meyer GP, Hiss M, Gwinner W et al. Endothelial microparticles as a diagnostic aid in Churg-Strauss vasculitis-induced cardiomyopathy. Clinical and experimental rheumatology, 2008;26:S86–9. [PubMed] [Google Scholar]
- [157].Clarke LA, Hong Y, Eleftheriou D, Shah V, Arrigoni F, Klein NJ et al. Endothelial injury and repair in systemic vasculitis of the young. Arthritis and rheumatism, 2010;62:1770–80. [DOI] [PubMed] [Google Scholar]
- [158].Brodsky SV, Zhang F, Nasjletti A, Goligorsky MS. Endothelium-derived microparticles impair endothelial function in vitro. American journal of physiology Heart and circulatory physiology, 2004;286:H1910–5. [DOI] [PubMed] [Google Scholar]
- [159].Njock MS, Cheng HS, Dang LT, Nazari-Jahantigh M, Lau AC, Boudreau E et al. Endothelial cells suppress monocyte activation through secretion of extracellular vesicles containing antiinflammatory microRNAs. Blood, 2015;125:3202–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [160].Tesse A, Martínez MC, Hugel B, Chalupsky K, Muller CD, Meziani F et al. Upregulation of proinflammatory proteins through NF-kappaB pathway by shed membrane microparticles results in vascular hyporeactivity. Arteriosclerosis, thrombosis, and vascular biology, 2005;25:2522–7. [DOI] [PubMed] [Google Scholar]
- [161].Lopatko Fagerström I, Ståhl AL, Mossberg M, Tati R, Kristoffersson AC, Kahn R et al. Blockade of the kallikrein-kinin system reduces endothelial complement activation in vascular inflammation. EBioMedicine, 2019;47:319–28. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [162].Marks RM, Todd RF 3rd, Ward PA. Rapid induction of neutrophil-endothelial adhesion by endothelial complement fixation. Nature, 1989;339:314–7. [DOI] [PubMed] [Google Scholar]
- [163].Ohlsson S, Holm L, Hansson C, Ohlsson SM, Gunnarsson L, Pettersson Å et al. Neutrophils from ANCA-associated vasculitis patients show an increased capacity to activate the complement system via the alternative pathway after ANCA stimulation. PloS one, 2019;14:e0218272. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [164].Watanabe-Kusunoki K, Nakazawa D, Kusunoki Y, Kudo T, Hattanda F, Nishio S et al. Recombinant thrombomodulin ameliorates autoimmune vasculitis via immune response regulation and tissue injury protection. Journal of autoimmunity, 2020;108:102390. [DOI] [PubMed] [Google Scholar]
- [165].Kumar SV, Kulkarni OP, Mulay SR, Darisipudi MN, Romoli S, Thomasova D et al. Neutrophil Extracellular Trap-Related Extracellular Histones Cause Vascular Necrosis in Severe GN. Journal of the American Society of Nephrology : JASN, 2015;26:2399–413. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [166].Pérez-Cremades D, Bueno-Betí C, García-Giménez JL, Ibañez-Cabellos JS, Hermenegildo C, Pallardó FV et al. Extracellular histones disarrange vasoactive mediators release through a COX-NOS interaction in human endothelial cells. Journal of cellular and molecular medicine, 2017;21:1584–92. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [167].Bianchi B, Katsambas A, Lotti TM. Neuropeptides and endothelial injury in cutaneous vasculitis. Clinics in dermatology, 1999;17:625–7. [DOI] [PubMed] [Google Scholar]
- [168].Bodolay E, Koch AE, Kim J, Szegedi G, Szekanecz Z. Angiogenesis and chemokines in rheumatoid arthritis and other systemic inflammatory rheumatic diseases. J Cell Mol Med, 2002;6:357–76. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [169].Maruotti N, Cantatore FP, Nico B, Vacca A, Ribatti D. Angiogenesis in vasculitides. Clinical and experimental rheumatology, 2008;26:476–83. [PubMed] [Google Scholar]
- [170].Bassyouni IH, Sharaf M, Wali IE, Mansour HM. Clinical significance of Angiopoietin-1 in Behcet’s disease patients with vascular involvement. Heart Vessels, 2016;31:918–24. [DOI] [PubMed] [Google Scholar]
- [171].Pulsatelli L, Boiardi L, Assirelli E, Pazzola G, Muratore F, Addimanda O et al. Imbalance between angiogenic and anti-angiogenic factors in sera from patients with large-vessel vasculitis. Clinical and experimental rheumatology, 2020;38 Suppl 124:23–30. [PubMed] [Google Scholar]
- [172].van Sleen Y, Sandovici M, Abdulahad WH, Bijzet J, van der Geest KSM, Boots AMH et al. Markers of angiogenesis and macrophage products for predicting disease course and monitoring vascular inflammation in giant cell arteritis. Rheumatology (Oxford, England), 2019;58:1383–92. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [173].Todorova D, Simoncini S, Lacroix R, Sabatier F, Dignat-George F. Extracellular Vesicles in Angiogenesis. Circulation research, 2017;120:1658–73. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [174].Carmeliet P, Jain RK. Molecular mechanisms and clinical applications of angiogenesis. Nature, 2011;473:298–307. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [175].Taraboletti G, D’Ascenzo S, Borsotti P, Giavazzi R, Pavan A, Dolo V. Shedding of the matrix metalloproteinases MMP-2, MMP-9, and MT1-MMP as membrane vesicle-associated components by endothelial cells. The American journal of pathology, 2002;160:673–80. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [176].Nawaz M, Shah N, Zanetti BR, Maugeri M, Silvestre RN, Fatima F et al. Extracellular Vesicles and Matrix Remodeling Enzymes: The Emerging Roles in Extracellular Matrix Remodeling, Progression of Diseases and Tissue Repair. Cells, 2018;7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [177].Sun Y, Liu XL, Zhang D, Liu F, Cheng YJ, Ma Y et al. Platelet-Derived Exosomes Affect the Proliferation and Migration of Human Umbilical Vein Endothelial Cells Via miR-126. Current vascular pharmacology, 2019;17:379–87. [DOI] [PubMed] [Google Scholar]
- [178].Hellström M, Kalén M, Lindahl P, Abramsson A, Betsholtz C. Role of PDGF-B and PDGFR-beta in recruitment of vascular smooth muscle cells and pericytes during embryonic blood vessel formation in the mouse. Development, 1999;126:3047–55. [DOI] [PubMed] [Google Scholar]
- [179].Brill A, Dashevsky O, Rivo J, Gozal Y, Varon D. Platelet-derived microparticles induce angiogenesis and stimulate post-ischemic revascularization. Cardiovasc Res, 2005;67:30–8. [DOI] [PubMed] [Google Scholar]
- [180].Yang C, Mwaikambo BR, Zhu T, Gagnon C, Lafleur J, Seshadri S et al. Lymphocytic microparticles inhibit angiogenesis by stimulating oxidative stress and negatively regulating VEGF-induced pathways. American journal of physiology Regulatory, integrative and comparative physiology, 2008;294:R467–76. [DOI] [PubMed] [Google Scholar]
- [181].Yang Y, Luo H, Zhou C, Zhang R, Liu S, Zhu X et al. Regulation of capillary tubules and lipid formation in vascular endothelial cells and macrophages via extracellular vesicle-mediated microRNA-4306 transfer. The Journal of international medical research, 2019;47:453–69. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [182].Wang XW, He XJ, Lee KC, Huang C, Hu JB, Zhou R et al. MicroRNA-221 sponge therapy attenuates neointimal hyperplasia and improves blood flows in vein grafts. International journal of cardiology, 2016;208:79–86. [DOI] [PubMed] [Google Scholar]
- [183].Ye B, Wu ZH, Tsui TY, Zhang BF, Su X, Qiu YH et al. lncRNA KCNQ1OT1 Suppresses the Inflammation and Proliferation of Vascular Smooth Muscle Cells through IκBa in Intimal Hyperplasia. Molecular therapy Nucleic acids, 2020;20:62–72. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [184].Weyand CM, Goronzy JJ. Immune mechanisms in medium and large-vessel vasculitis. Nature reviews Rheumatology, 2013;9:731–40. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [185].Hid Cadena R, Abdulahad WH, Hospers GAP, Wind TT, Boots AMH, Heeringa P et al. Checks and Balances in Autoimmune Vasculitis. Frontiers in immunology, 2018;9:315. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [186].Liu H, Dong S, Xiong W, Liu Q, Liao B. Effects of high mobility group box 1 and nuclear factor κB on neointimal hyperplasia after common carotid artery balloon injury. International journal of clinical and experimental medicine, 2015;8:13323–8. [PMC free article] [PubMed] [Google Scholar]
- [187].Cai J, Yuan H, Wang Q, Yang H, Al-Abed Y, Hua Z et al. HMGB1-Driven Inflammation and Intimal Hyperplasia After Arterial Injury Involves Cell-Specific Actions Mediated by TLR4. Arteriosclerosis, thrombosis, and vascular biology, 2015;35:2579–93. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [188].Kim EJ, Park SY, Baek SE, Jang MA, Lee WS, Bae SS et al. HMGB1 Increases IL-1β Production in Vascular Smooth Muscle Cells via NLRP3 Inflammasome. Front Physiol, 2018;9:313. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [189].Eun SY, Ko YS, Park SW, Chang KC, Kim HJ. IL-1β enhances vascular smooth muscle cell proliferation and migration via P2Y2 receptor-mediated RAGE expression and HMGB1 release. Vascular pharmacology, 2015;72:108–17. [DOI] [PubMed] [Google Scholar]
- [190].Wang JG, Williams JC, Davis BK, Jacobson K, Doerschuk CM, Ting JP et al. Monocytic microparticles activate endothelial cells in an IL-1β-dependent manner. Blood, 2011;118:2366–74. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [191].Vats R, Brzoska T, Bennewitz MF, Jimenez MA, Pradhan-Sundd T, Tutuncuoglu E et al. Platelet Extracellular Vesicles Drive Inflammasome-IL-1β-Dependent Lung Injury in Sickle Cell Disease. Am J Respir Crit Care Med, 2020;201:33–46. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [192].Zhu J, Liu B, Wang Z, Wang D, Ni H, Zhang L et al. Exosomes from nicotine-stimulated macrophages accelerate atherosclerosis through miR-21–3p/PTEN-mediated VSMC migration and proliferation. Theranostics, 2019;9:6901–19. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [193].Li X, Ballantyne LL, Yu Y, Funk CD. Perivascular adipose tissue-derived extracellular vesicle miR-221–3p mediates vascular remodeling. FASEB journal : official publication of the Federation of American Societies for Experimental Biology, 2019;33:12704–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [194].Liu R, Shen H, Ma J, Sun L, Wei M. Extracellular Vesicles Derived from Adipose Mesenchymal Stem Cells Regulate the Phenotype of Smooth Muscle Cells to Limit Intimal Hyperplasia. Cardiovasc Drugs Ther, 2016;30:111–8. [DOI] [PubMed] [Google Scholar]
- [195].Qu Q, Pang Y, Zhang C, Liu L, Bi Y. Exosomes derived from human umbilical cord mesenchymal stem cells inhibit vein graft intimal hyperplasia and accelerate reendothelialization by enhancing endothelial function. Stem Cell Res Ther, 2020;11:133. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [196].Nakazawa D, Masuda S, Tomaru U, Ishizu A. Pathogenesis and therapeutic interventions for ANCA-associated vasculitis. Nature reviews Rheumatology, 2019;15:91–101. [DOI] [PubMed] [Google Scholar]
- [197].Binda V, Moroni G, Messa P. ANCA-associated vasculitis with renal involvement. Journal of nephrology, 2018;31:197–208. [DOI] [PubMed] [Google Scholar]
- [198].Miao D, Ma TT, Chen M, Zhao MH. Platelets release proinflammatory microparticles in antineutrophil cytoplasmic antibody-associated vasculitis. Rheumatology (Oxford, England), 2019. [DOI] [PubMed] [Google Scholar]
- [199].Daniel L, Fakhouri F, Joly D, Mouthon L, Nusbaum P, Grunfeld JP et al. Increase of circulating neutrophil and platelet microparticles during acute vasculitis and hemodialysis. Kidney international, 2006;69:1416–23. [DOI] [PubMed] [Google Scholar]
- [200].Jennette JC, Nachman PH. ANCA Glomerulonephritis and Vasculitis. Clinical journal of the American Society of Nephrology : CJASN, 2017;12:1680–91. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [201].Zhao L, David MZ, Hyjek E, Chang A, Meehan SM. M2 macrophage infiltrates in the early stages of ANCA-associated pauci-immune necrotizing GN. Clin J Am Soc Nephrol, 2015;10:54–62. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [202].Yoshida M, Sasaki M, Sugisaki K, Yamaguchi Y, Yamada M. Neutrophil extracellular trap components in fibrinoid necrosis of the kidney with myeloperoxidase-ANCA-associated vasculitis. Clinical kidney journal, 2013;6:308–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [203].Nakaoka H, Hirono K, Yamamoto S, Takasaki I, Takahashi K, Kinoshita K et al. MicroRNA-145–5p and microRNA-320a encapsulated in endothelial microparticles contribute to the progression of vasculitis in acute Kawasaki Disease. Scientific reports, 2018;8:1016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [204].Shah V, Christov G, Mukasa T, Brogan KS, Wade A, Eleftheriou D et al. Cardiovascular status after Kawasaki disease in the UK. Heart (British Cardiac Society), 2015;101:1646–55. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [205].Guiducci S, Ricci L, Romano E, Ceccarelli C, Distler JH, Miniati I et al. Microparticles and Kawasaki disease: a marker of vascular damage? Clinical and experimental rheumatology, 2011;29:S121–5. [PubMed] [Google Scholar]
- [206].Tan Z, Yuan Y, Chen S, Chen Y, Chen TX. Plasma Endothelial Microparticles, TNF-a and IL-6 in Kawasaki Disease. Indian pediatrics, 2013;50:501–3. [DOI] [PubMed] [Google Scholar]
- [207].Zhang L, Wang W, Bai J, Xu YF, Li LQ, Hua L et al. Proteomic analysis associated with coronary artery dilatation caused by Kawasaki disease using serum exosomes. Revista portuguesa de cardiologia : orgao oficial da Sociedade Portuguesa de Cardiologia = Portuguese journal of cardiology : an official journal of the Portuguese Society of Cardiology, 2016;35:265–73. [DOI] [PubMed] [Google Scholar]
- [208].Jia HL, Liu CW, Zhang L, Xu WJ, Gao XJ, Bai J et al. Sets of serum exosomal microRNAs as candidate diagnostic biomarkers for Kawasaki disease. Scientific reports, 2017;7:44706. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [209].Zhang X, Xin G, Sun D. Serum exosomal miR-328, miR-575, miR-134 and miR-671–5p as potential biomarkers for the diagnosis of Kawasaki disease and the prediction of therapeutic outcomes of intravenous immunoglobulin therapy. Experimental and therapeutic medicine, 2018;16:2420–32. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [210].Zhang L, Song QF, Jin JJ, Huang P, Wang ZP, Xie XF et al. Differential protein analysis of serum exosomes post-intravenous immunoglobulin therapy in patients with Kawasaki disease. Cardiology in the young, 2017;27:1786–96. [DOI] [PubMed] [Google Scholar]
- [211].Jin J, Wang J, Lu Y, Fan Z, Huang N, Ma L et al. Platelet-Derived Microparticles: A New Index of Monitoring Platelet Activation and Inflammation in Kawasaki Disease. Indian journal of pediatrics, 2019;86:250–5. [DOI] [PubMed] [Google Scholar]
- [212].Yahata T, Suzuki C, Yoshioka A, Hamaoka A, Ikeda K. Platelet activation dynamics evaluated using platelet-derived microparticles in Kawasaki disease. Circulation journal : official journal of the Japanese Circulation Society, 2014;78:188–93. [DOI] [PubMed] [Google Scholar]
- [213].Kim HJ, Choi EH, Lim YJ, Kil HR. The Usefulness of Platelet-derived Microparticle as Biomarker of Antiplatelet Therapy in Kawasaki Disease. Journal of Korean medical science, 2017;32:1147–53. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [214].Cheng X, Dang A, Lv N, Zhao T. Microparticles from Endothelial Cells and Immune Cells in Patients with Takayasu Arteritis. Journal of atherosclerosis and thrombosis, 2019;26:547–58. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [215].Wang S, Dong X, Gao J, Wang Z. Targeting Inflammatory Vasculature by Extracellular Vesicles. The AAPS journal, 2018;20:37. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [216].Strnad J, Burke JR. IkappaB kinase inhibitors for treating autoimmune and inflammatory disorders: potential and challenges. Trends Pharmacol Sci, 2007;28:142–8. [DOI] [PubMed] [Google Scholar]
- [217].Gao J, Chu D, Wang Z. Cell membrane-formed nanovesicles for disease-targeted delivery. J Control Release, 2016;224:208–16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [218].Kim M, Kim G, Hwang DW, Lee M. Delivery of High Mobility Group Box-1 siRNA Using Brain-Targeting Exosomes for Ischemic Stroke Therapy. Journal of biomedical nanotechnology, 2019;15:2401–12. [DOI] [PubMed] [Google Scholar]
- [219].Pakravan K, Babashah S, Sadeghizadeh M, Mowla SJ, Mossahebi-Mohammadi M, Ataei F et al. MicroRNA-100 shuttled by mesenchymal stem cell-derived exosomes suppresses in vitro angiogenesis through modulating the mTOR/HIF-1α/VEGF signaling axis in breast cancer cells. Cell Oncol (Dordr), 2017;40:457–70. [DOI] [PubMed] [Google Scholar]
- [220].Qu Q, Wang L, Bing W, Bi Y, Zhang C, Jing X et al. miRNA-126–3p carried by human umbilical cord mesenchymal stem cell enhances endothelial function through exosome-mediated mechanisms in vitro and attenuates vein graft neointimal formation in vivo. Stem cell research & therapy, 2020;11:464. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [221].Gupta D, Wiklander O, Görgens A, Conceicao M, Corso G, Liang X et al. Engineering of extracellular vesicles for display of protein biotherapeutics; 2020. [Google Scholar]
- [222].Martínez M, Ricart JM, Ruiz-Aja S, Rus A, Todolí J, Calvo J et al. Platelet activation and red blood cell phosphatidylserine exposure evaluated by flow cytometry in patients with Behçet’s disease: are they related to thrombotic events? Pathophysiology of haemostasis and thrombosis, 2007;36:18–22. [DOI] [PubMed] [Google Scholar]
- [223].Hajj-Ali RA, Major J, Langford C, Hoffman GS, Clark T, Zhang L et al. The interface of inflammation and subclinical atherosclerosis in granulomatosis with polyangiitis (Wegener’s): a preliminary study. Translational research : the journal of laboratory and clinical medicine, 2015;166:366–74. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [224].Dursun I, Düsünsel R, Poyrazoglu HM, Gunduz Z, Patıroglu T, Ulger H et al. Circulating endothelial microparticles in children with Henoch-Schönlein purpura; preliminary results. Rheumatology international, 2011;31:1595–600. [DOI] [PubMed] [Google Scholar]