Publications
& News

Extracellular Vesicles Tune the Immune System in Renal Disease: A Focus on Systemic Lupus Erythematosus, Antiphospholipid Syndrome, Thrombotic Microangiopathy and ANCA-Vasculitis

1. January 2021

International Journal o f
Molecular Sciences
Review
Extracellular Vesicles Tune the Immune System in Renal
Disease: A Focus on Systemic Lupus Erythematosus,
Antiphospholipid Syndrome, Thrombotic Microangiopathy
and ANCA-Vasculitis
Martina Mazzariol, Giovanni Camussi and Maria Felice Brizzi *


Citation: Mazzariol, M.; Camussi, G.;
Brizzi, M.F. Extracellular Vesicles
Tune the Immune System in Renal
Disease: A Focus on Systemic Lupus
Erythematosus, Antiphospholipid
Syndrome, Thrombotic
Microangiopathy and ANCAVasculitis.
Int. J. Mol. Sci. 2021, 22,

  1. https://doi.org/10.3390/
    ijms22084194
    Academic Editor: Leonora Balaj
    Received: 29 March 2021
    Accepted: 16 April 2021
    Published: 18 April 2021
    Publisher’s Note: MDPI stays neutral
    with regard to jurisdictional claims in
    published maps and institutional affiliations.
    Copyright: © 2021 by the authors.
    Licensee MDPI, Basel, Switzerland.
    This article is an open access article
    distributed under the terms and
    conditions of the Creative Commons
    Attribution (CC BY) license (https://
    creativecommons.org/licenses/by/
    4.0/).
    Department of Medical Sciences, University of Turin, Corso Dogliotti 14, 10126 Turin, Italy;
    marti.mazzariol@gmail.com (M.M.); giovanni.camussi@unito.it (G.C.)
  • Correspondence: mariafelice.brizzi@unito.it; Tel.: +39-01-1670-6653
    Abstract: Extracellular vesicles (EV) are microparticles released in biological fluids by different cell
    types, both in physiological and pathological conditions. Owing to their ability to carry and transfer
    biomolecules, EV are mediators of cell-to-cell communication and are involved in the pathogenesis
    of several diseases. The ability of EV to modulate the immune system, the coagulation cascade, the
    angiogenetic process, and to drive endothelial dysfunction plays a crucial role in the pathophysiology
    of both autoimmune and renal diseases. Recent studies have demonstrated the involvement of
    EV in the control of renal homeostasis by acting as intercellular signaling molecules, mediators
    of inflammation and tissue regeneration. Moreover, circulating EV and urinary EV secreted by
    renal cells have been investigated as potential early biomarkers of renal injury. In the present
    review, we discuss the recent findings on the involvement of EV in autoimmunity and in renal
    intercellular communication. We focused on EV-mediated interaction between the immune system
    and the kidney in autoimmune diseases displaying common renal damage, such as antiphospholipid
    syndrome, systemic lupus erythematosus, thrombotic microangiopathy, and vasculitis. Although
    further studies are needed to extend our knowledge on EV in renal pathology, a deeper investigation
    of the impact of EV in kidney autoimmune diseases may also provide insight into renal biological
    processes. Furthermore, EV may represent promising biomarkers of renal diseases with potential
    future applications as diagnostic and therapeutic tools.
    Keywords: renal disease; autoimmune diseases; HUS; TTP; APS; antiphospholipid syndrome; vasculitis;
    systemic lupus erythematosus; lupus nephritis; pathogenesis; microparticles; extracellular
    vesicles; microvesicles; exosomes
  1. Introduction
    Extracellular vesicles (EV) are extracellular structures bounded by a phospholipid
    bilayer and released by different cell types in biological fluids (blood, urine, synovial
    fluids) through various mechanisms. First described as “platelet dust” by Peter Wolf
    in 1967, in recent years EV have gained interest as a commonly recognized important
    player in cell-to-cell communication, both in physiological and pathological conditions.
    This depends on their ability to transfer their cargo, consisting of proteins, lipids, or
    nucleic acids. Interestingly, EV have a role in different biological processes including cell
    proliferation and differentiation, inflammation, immune signaling, angiogenesis, and stress
    responses [1]. In this review, we will summarize the most recent data on EV biological
    features and their pathogenic role in immune-associated renal disease, and their potential
    use as disease biomarkers.
    Int. J. Mol. Sci. 2021, 22, 4194. https://doi.org/10.3390/ijms22084194 https://www.mdpi.com/journal/ijms
    Int. J. Mol. Sci. 2021, 22, 4194 2 of 20
    1.1. EV Nomenclature
    The most recently updated guidelines of the International Society for Extracellular
    Vesicles (ISEV) recommend using the term “extracellular vesicles” as “the generic term for
    particles naturally released from the cell that are delimited by a lipid bilayer and cannot
    replicate” [2]. EV were historically classified into three main subtypes according to their
    size, biogenesis and biological features: exosomes (the smallest EV generated from the
    invagination of the endosomal membrane) [1], microvesicles (or EV which are directly
    released from the plasma membrane) [3] and apoptotic bodies (the largest EV released by
    apoptotic cells) [4]. However, since a consensus has not yet emerged on specific markers
    defining each subtype, the ISEV guidelines recommend defining EV according to [2]:
     physical characteristics such as size (small EV and medium/large EV, with ranges
    defined, for instance, respectively, <100 nm or <200 nm (small), or >200 nm (large
    and/or medium)) or density (low, middle, high);
     biochemical composition (CD63+/CD81+􀀀 EV, Annexin A5-stained EV, etc.);
     descriptions of conditions or cell of origin (podocyte EV, hypoxic EV, large oncosomes,
    apoptotic bodies) [2].
    1.2. EV Isolation and Detection
    EV can be detected in almost all human fluids (blood samples, cerebrospinal fluid,
    synovial fluid, urine, bile, saliva, bronchoalveolar fluid) using different techniques depending
    on the EV population. In the pre-analytical phase, body sample collection, handling,
    and storage may impact EV concentration, composition, and function. Thus, to follow a
    preanalytical protocol tailored to the specific body fluid, the cell of origin of EV is crucial [5].
    After collection and isolation, EV can be analyzed through various methods. The most
    commonly exploited and recommended are the following:
     flow cytometry which detects EV passing through a laser beam. Modern flow cytometers
    may have many lasers and fluorescence detectors, which allow to label them with
    multiple conjugated antibodies using the same sample. Although widely used, the
    analysis using flow cytometry has limitations in detecting the smallest EV whose number
    and surface expression may be underestimated [3]. To circumvent this limitation,
    an alternative bead-based technique has been developed using specifically activated
    beads that capture EV with a cocktail of different exosome marker epitopes allowing
    subsequent simultaneous detection of multiple antigens [6];
     nanoparticle tracking analysis (NTA) which visualizes EV in the liquid phase by
    light scattering using a light microscope. A video is taken and the NTA software
    tracks the Brownian motion of individual vesicles and calculates their size and total
    concentration. NTA with fluorescent mode detects labeled vesicles and provides
    quantitative and qualitative analysis. NTA can detect vesicles smaller than those
    distinguished by conventional flow cytometry [3].
    1.3. EV and Cell-to-Cell Communication
    EV are shed into the extracellular environment under physiological and pathological
    conditions, and their release is increased by cellular stress conditions as inflammation,
    hypoxia, oxidative stress or infections [7]. Once released into the circulation, EV have a
    half-life ranging from a few minutes to a few hours, during which they can be taken up
    from cells by different mechanisms such as endocytosis or fusion with the membrane of the
    recipient cell [3]. Circulating EV mainly originate from platelets, erythrocytes, leukocytes,
    and endothelial cells; while urinary EV mainly derive from podocytes, tubular cells, and
    epithelial cells [8]. EV may transfer to the target cell receptors, allowing cell signaling
    in cells that are originally devoid of receptors or by enhancing their number. Moreover,
    EV may release or transfer proteins as cytokines and growth factors translating in the
    modulation of target cells and their extracellular environment. Furthermore, EV may
    transfer messenger RNAs (mRNAs) and microRNAs (miRNAs) to target cells, modulating
    their function or changing their phenotype [3].
    Int. J. Mol. Sci. 2021, 22, 4194 3 of 20
  2. EV in Physiological and Pathological Settings
    Based on their crucial role in cell-to-cell communication, EV have been involved in
    several processes such as coagulation and thrombosis, endothelial dysfunction, angiogenesis
    and immune modulation. Since all of these processes impact renal diseases, a brief
    discussion will be reported (Figure 1).
    Int. J. Mol. Sci. 2021, 22, x FOR PEER REVIEW 3 of 21
    may transfer messenger RNAs (mRNAs) and microRNAs (miRNAs) to target cells,
    modulating their function or changing their phenotype [3].
  3. EV in Physiological and Pathological Settings
    Based on their crucial role in cell-to-cell communication, EV have been involved in
    several processes such as coagulation and thrombosis, endothelial dysfunction,
    angiogenesis and immune modulation. Since all of these processes impact renal diseases,
    a brief discussion will be reported (Figure 1).
    Figure 1. EV in physiological and pathological processes. (A) EV and coagulation: EV enriched in phosphatidylserine (PS)
    and tissue factor (TF) can promote the activation of the coagulation cascade as well as platelet aggregation. (B) EV and
    endothelial activation: EV can modulate vasodilatation and vasoconstriction by activating endothelial cells, releasing
    cytokines, reducing nitric oxide (NO) production and increasing reactive oxygen species (ROS) production. (C) EV and
    angiogenesis: EV can modulate angiogenesis by increasing ROS production, downregulating the VEGF pathway,
    modulating the migration and proliferation of endothelial cells and matrix degradation. This figure has been created using
    Servier Medical Art templates, which are licensed under a Creative Commons Attribution 3.0 Unported License;
    https://smart.servier.com.
    2.1. EV and Coagulation
    Several autoimmune and renal diseases are associated with increased thrombotic risk
    and thrombotic events. EV are involved in the coagulation cascade as carriers of
    phosphatidylserine (PS) and tissue factor (TF) and are able to promote the activation of
    the coagulation cascade. PS can act as a binding site for the coagulation factors II, Va and
    Xa, enhancing the association between factor Xa and Va and the subsequent conversion
    of prothrombin to thrombin [9]. TF expressed on the EV surface can activate the extrinsic
    coagulation pathway by binding factor VIIa [10]. Interestingly, platelet-derived EV have
    Figure 1. EV in physiological and pathological processes. (A) EV and coagulation: EV enriched in
    phosphatidylserine (PS) and tissue factor (TF) can promote the activation of the coagulation cascade
    as well as platelet aggregation. (B) EV and endothelial activation: EV can modulate vasodilatation
    and vasoconstriction by activating endothelial cells, releasing cytokines, reducing nitric oxide (NO)
    production and increasing reactive oxygen species (ROS) production. (C) EV and angiogenesis:
    EV can modulate angiogenesis by increasing ROS production, downregulating the VEGF pathway,
    modulating the migration and proliferation of endothelial cells and matrix degradation. This figure
    has been created using Servier Medical Art templates, which are licensed under a Creative Commons
    Attribution 3.0 Unported License; https://smart.servier.com (accessed on 28 March 2021).
    2.1. EV and Coagulation
    Several autoimmune and renal diseases are associated with increased thrombotic
    risk and thrombotic events. EV are involved in the coagulation cascade as carriers of
    phosphatidylserine (PS) and tissue factor (TF) and are able to promote the activation of
    the coagulation cascade. PS can act as a binding site for the coagulation factors II, Va and
    Xa, enhancing the association between factor Xa and Va and the subsequent conversion
    of prothrombin to thrombin [9]. TF expressed on the EV surface can activate the extrinsic
    coagulation pathway by binding factor VIIa [10]. Interestingly, platelet-derived EV have
    higher pro-coagulant activity compared with activated platelets, possibly due to their
    enrichment in phosphatidylserine, glycoprotein IIb/IIIa, factor Xa, and P-selectin [11].
    Moreover, EV-mediated thrombotic events can be also promoted by their expression of the
    multimers of vonWillebrand factor and stabilization of platelet aggregation (Figure 1A) [9].
    2.2. EV and Endothelial Dysfunction
    Endothelial dysfunction is frequently associated with the pathogenesis of autoimmune
    diseases: EV contribute to endothelial regulation in physiological and pathological
    conditions. EV can reduce endothelial and macrophage nitric oxide production, controlling
    vasodilation. EV can also carry thromboxane A2 acting as vasoconstrictor and platelet
    Int. J. Mol. Sci. 2021, 22, 4194 4 of 20
    aggregation factor [1]. In addition, EV may increase the production of reactive oxygen
    species (ROS). Finally, as occurs in systemic vasculitis, activated neutrophils release EV expressing
    the myeloperoxidase (MPO) which by activating the myeloperoxidase-hydrogen
    peroxide-chloride system, leads to endothelial damage (Figure 1B) [12].
    2.3. EV and Angiogenesis
    Angiogenesis, defined as the growth of new blood vessels from pre-existing ones, is
    frequently involved in the pathogenesis of autoimmune diseases. EV may be involved
    in several angiogenesis-associated events [13] as matrix degradation, endothelial progenitor
    recruitment, and differentiation, migration and proliferation of endothelial cells.
    Endothelial- and macrophage-derived EV contain pro-angiogenic enzymes (MMP-2, MMP-
    9, MT1-MMP) which promote extracellular matrix remodeling and favor endothelial cell
    invasion [14]. On the contrary, lymphocyte-derived EV can inhibit angiogenesis by generating
    ROS and down-regulating the VEGF pathway (Figure 1C) [15].
    2.4. EV and Immune System Modulation
    EV may affect the immune response by activating innate and adaptive immunity
    and by participating in the formation of the immune complexes (IC), by acting as autoantigens.
    Immune cells release immunocompetent EV, which can modulate the immune
    response by regulating antigen presentation, NK/T cell activation, T cell polarization and
    immunosuppression (Figure 2) [16].
    In the immune system, natural killer (NK) cells have a role in innate immunity, whereas
    B and T cells are an essential part of adaptive immunity.
     Antigen presentation B cells can recognize foreign antigens, while T cells require
    antigen-presenting cells (APCs) for antigen recognition. Major histocompatibility
    complex class I (MHC I) and class II (MHC-II) present antigens to CD8+ and CD4+
    T cells, thereby activating the immune response. APCs- and B cells-derived EV
    express the MHC-I, MHC-II and the T-cell costimulatory molecules, thus may take
    part in the antigen presentation process and in the CD8+ and CD4+ T cell activation
    (Figure 2A) [17].
     Source of self-antigens and IC formation EV participate in the formation of IC. Indeed,
    EV can express both self-antigens and MHC complexes and may activate autoreactive
    T-cells in autoimmune disease. As an example, the synovial fluid of patients with
    rheumatoid arthritis contains IC composed of platelet-derived EV and autoantibodies
    against citrullinated peptides [18]. Similarly, in systemic lupus erythematosus (SLE),
    EV carry nuclear molecules, which represent a potential source of autoantigens and
    participate in IC formation. Furthermore, EV-associated ICs may affect the recognition
    and clearance of EV by phagocytes, leading to the accumulation of cell debris and
    triggering the autoimmune response (Figure 2B,C) [19].
     Role of adjuvants in innate immune response Leukocyte-derived EV activate the
    endothelium by upregulating adhesion molecules and releasing cytokines. This
    leads to leukocyte recruitment via platelet-derived EV, which promotes monocyte
    adhesion to the endothelium [20]. Dendritic cell-derived EV increase the NK cytotoxic
    activity and stimulate the release of proinflammatory cytokines by epithelial cells
    (Figure 2D) [21].
     Role in complement activation When the complement system undergoes activation, the
    membrane attack complex may be set down on blood cells and complement-coated EV
    may be released. C3-positive EV reflect the activation of the alternative pathway of the
    complement, while C1q-positive EV reflect the activation of the classic pathway [22].
    Moreover, EV may express complement regulators on their surface (complement
    receptor type 1, membrane cofactor protein, decay-accelerating factor also denoted as
    CD59), thereby inhibiting the membrane attack complex (Figure 2E) [23].
    Int. J. Mol. Sci. 2021, 22, 4194 5 of 20
    Int. J. Mol. Sci. 2021, 22, x FOR PEER REVIEW 5 of 21
    Figure 2. EV and immune system modulation: (A) Self-antigen presentation: antigen-presenting cells
    (APC) can vehicle self-antigens and release extracellular vesicles (EV) which present autoantigens
    to autoreactive T cells. (B) Self-antigen transfer: EV carrying self-antigens transfer them to APC
    which present them to T cells, triggering an autoimmune response. (C) Immune complex formation
    (IC): EV can participate in IC formation by carrying self-antigens that are bound by circulating
    autoantibodies. (D) Activation of innate immunity: EV can stimulate cytokine release and the upregulation
    of adhesion molecules on endothelial cells, which favor circulating immune cell recruitment.
    (E) Interaction with the complement system: the complement cascade can be activated on circulating
    leukocytes which can then release EV exposing complement molecules (e.g., C3, C1q). EV can also
    convey complement inhibitors and modulate the complement system. This Figure has been created
    using Servier Medical Art templates, which are licensed under a Creative Commons Attribution 3.0
    Unported License; https://smart.servier.com (accessed on 28 March 2021).
    Int. J. Mol. Sci. 2021, 22, 4194 6 of 20
  4. EV and Renal Intercellular Communication
    Recent studies have shown the relevance of EV in renal intercellular communication.
    Although their role has not been yet fully understood, their potential application as diagnostic,
    prognostic, or therapeutic biomarkers for various kidney diseases is currently being
    explored [8].
    In physiological conditions, the glomerular filtration apparatus prevents circulating
    EV from reaching the lumen of renal nephrons, thus circulating EV may stimulate kidney
    cells facing the vascular compartment and the immune cells [24]. However, EV are released
    from renal cells into the renal tubule and can be detected in urine. The release of renal
    EV may vary during renal disease, thus urinary EV have been proposed as potential
    biomarkers of kidney injury. Urinary EV are largely composed of EV released from renal
    cells as confirmed by proteomic analysis. Urinary EV carry specific proteins, mRNAs
    and miRNAs, which reflect their cellular origin. Indeed, EV from glomerular podocytes
    contain podocin and podocalyxin [25], EVs from proximal tubular cells express cubilin,
    megalin, aminopeptidase [26] and aquaporin-1; EVs from the Henle’s loop contain type 2
    Na-K-2Cl cotransporter, CD9 and Tamm–Horsfall protein [27]; EVs from collecting ducts
    carry AQP-2 and mucin-1 [28]. The majority of urinary EV are released from the first part of
    the renal nephron, with a limited contribution from the lower urinary collecting system [29].
    Interestingly, EV may have a role in cell-to-cell communication between the proximal and
    distal renal tubules. EV released from glomerular podocytes, via the urine flux, reach and
    can be taken up by the epithelial cells of the distal tubule and the collecting duct [30]. A
    recent study has investigated a potential hormonal mechanism that regulates EV uptake. It
    has been demonstrated that in vitro stimulation with desmopressin selectively stimulates
    the EV uptake in tubular cells, while a vasopressin antagonist reduced in vivo the uptake
    of EV injected within the renal tissue [31]. Moreover, EV may take part in the regulation of
    renal inflammation. Indeed, it was reported that EV from proximal tubular cells cultured
    in the presence of a dopamine receptor antagonist are able to reduce ROS production in
    distal tubular cells [32]. Interestingly, EV can modulate renal tissue repair and fibrosis
    after renal damage [33–35]. In vitro studies have demonstrated that hypoxic conditions
    increase the release of tubular cell-derived EV, which induced fibroblast activation and
    proliferation by the transfer of TGF mRNA [36]. EV may also control the ion transport, as
    EV from proximal tubule cells can decrease ENaC activity in the distal tubule and collective
    ducts [37]. Moreover, EV can also mediate the transfer of aquaporin 2 from cells of the
    upper collective duct to those of the lower collective duct, increasing water transport in
    the recipient cells [38]. Urinary EV may also exert protection against bacterial infection
    of the urinary tract. As a matter of fact, urinary EV carry antimicrobial peptides and can
    inhibit the growth of the most common urinary pathogen Escherichia coli by inducing their
    lysis [39]. Mesenchymal-to-epithelial transition is a crucial process in kidney development
    and regeneration. It has been reported that this process is mediated by tubular epithelial
    cell-derived EV which carry a specific subset of miRNAs, driving the differentiation of
    mesenchymal stem cells into epithelial cells [40]. Interestingly, mesenchymal stem cells
    and endothelial progenitor cells release EV that induce nephron regeneration and repair by
    promoting tubular proliferation and by inhibiting apoptosis [41,42].
  5. EV in Renal Disease
    4.1. Antiphospholipid Syndrome
    Antiphospholipid syndrome (APS) is a systemic autoimmune disease characterized
    by recurrent arterial or venous thrombosis and/or obstetric complications in the presence
    of antiphospholipid antibodies (aPL). APS is the most common cause of acquired
    thrombophilia and is associated with decreased survival. A severe form of APS, termed
    catastrophic antiphospholipid syndrome, occurs in <1% of patients with aPL and is associated
    with high mortality [43]. Despite the clinical relevance of this syndrome, its
    pathogenesis is not yet fully understood and recent studies have focused on EV as potential
    damage mediators.
    Int. J. Mol. Sci. 2021, 22, 4194 7 of 20
    Various studies have reported an increased level of EV in APS patients. Štok et al. [6]
    have compared APS patients, patients with idiopathic thrombosis and healthy controls and
    found that circulating EV were increased in patients with a history of thrombotic events
    (APS patients and patients with idiopathic thrombosis) compared to healthy subjects,
    suggesting a chronic cell activation even in the absence of an acute thrombotic event.
    This study demonstrated the presence of platelet, endothelial cell, lymphocyte, antigenpresenting
    cell-derived EV in patients with a history of thrombosis as well as in healthy
    controls. These EV express molecules involved in platelet/endothelial function, immune
    regulation extracellular matrix regulation and cell-to-cell adhesion. The analysis of EV
    surface proteins demonstrated an increased expression of CD8, CD44, CD133/1 and CD62P
    in the aPL patients. The increased expression of CD133/1 and CD62P on the EV surface in
    APS patients could reflect the increased endothelial and platelet activation, respectively, and
    their possible contribution to the thrombotic events [6]. Endothelial- and platelet-derived
    EV were increased in aPL patients, suggesting a chronic activation of endothelial cells and
    platelets. Interestingly, a correlation between the level of endothelial-derived EV and the
    level of anti- 2GPI has been demonstrated which closely correlates with thrombosis [44].
    Breen et al. [45] have shown an increased level of endothelial- and platelet-derived EV
    in aPL patients compared with healthy subjects, while no difference between obstetric APS
    or asymptomatic aPL patients was detected. Moreover, plasma from patients with APS
    and from patients with SLE aPL+ or SLE aPL- increased the release of EV from cultured
    endothelial cells compared to the plasma of healthy subjects. Of note, only plasma from
    APS patients caused the release of EV with significant procoagulant activity [46]. aPL
    induces tissue factor (TF) synthesis in endothelial cells in vitro, and it has been reported
    that TF+ EV are elevated in aPL+ patients [44]. In particular, TF expression on endothelialderived
    EV is increased in APS patients compared to healthy subjects [47]. Moreover,
    Willemze et al. [48] have demonstrated that EV from APS patients display a higher TF
    activity compared to asymptomatic aPL+ patients.
    More recently, Mobarrez et al. [19] have compared anti- 2GPI-positive SLE patients,
    aPL-negative SLE patients and healthy controls. They found that SLE patients are depleted
    of 2GPI-positive EV when compared to healthy subjects, and their level is particularly
    low in anti- 2GPI-positive patients. They also found that 2GPI preferentially binds to the
    phosphatidylserine (PS)-positive EV, thus suggesting that anti- 2GPI antibodies may bind
    to the 2GPI-PS complexes on EV resulting in the loss of EV 2GPI expression. 2GPI
    promotes the clearance of PS+ EV, thus the increased number of PS-negative EV may act as
    a possible source of autoantigens and thus trigger the autoimmune response [19].
    Regarding obstetric APS, pregnant women with APS had increased PS+ EV, endoglin+
    EV and endothelium-derived EV compared to healthy controls in the first and second
    trimester of pregnancy. Conversely, in the third trimester, higher levels of TF+ EV and
    platelet-derived EV can be detected. According to the authors, this finding could reflect the
    activation of both endothelial cells and platelets during pregnancy. In particular, high-risk
    APS patients (triple aPL positivity plus vascular thrombosis and/or severe pregnancy
    complications/placental insufficiency) have higher endoglin+ EV, TF+ EV and platelet
    derived-EV in all three trimesters, sustaining a major vascular activation. Interestingly,
    endoglin is expressed by vascular endothelium and by syncytiotrophoblasts and altered
    levels of soluble endoglin are linked to vascular disorders as pre-eclampsia [49].
    In conclusion, patients with aPL have an elevated level of circulating EV, which may
    reflect a state of systemic vascular activation. EV have been shown to act as procoagulant
    and proinflammatory mediators, thus their level may correlate with the thrombotic risk.
    However, at present, a clear relationship between elevated EV levels and thrombotic events
    is still missing (Table 1).
    Int. J. Mol. Sci. 2021, 22, 4194 8 of 20
    Table 1. Extracellular vesicles in antiphospholipid syndrome (APS).
    Study EV Biomarkers Cellular Origin of EV Study Findings Reference
    Štok, U.; et al.
    CD8, CD44, CD133/1,
    CD62P
    Platelets, endothelial
    cells, lymphocytes,
    antigen-presenting cells
    EV increased in patients with thrombotic events
    EV reflect endothelial and platelet chronic
    activation
    [6]
    Chaturvedi, S.; et al.,
    Breen, K.A.; et al.
    CD41, CD61, CD51,
    CD105
    Endothelial cells,
    platelets
    EV increased in aPL+ patients
    EV reflect endothelial and platelet chronic
    activation
    [44,45]
    Chaturvedi, S.; et al.,
    Willemze, R.; et al. Tissue factor (TF) Endothelial cells TF + EV increased in APS
    TF activity increased in EV from aPL+ patients [44,48]
    Mobarrez, F.; et al. 2GPI+ EV 2GPI+ reduced in SLE aPL+
    Anti- 2GPI may bind to 2GPI expressed by EV [19]
    Campello, E.; et al.
    Phosphatidylserine (PS),
    Endoglin,
    Tissue factor (TF)
    Endothelial cells,
    platelets
    PS+ EV, endoglin+ EV and endothelium-derived
    EV increased in 1st and 2nd trimester of
    pregnancy;
    TF+ EV and platelet-derived EV increased in 3rd
    trimester of pregnancy
    Correlation with thrombosis and systemic platelet
    and endothelial activation in obstetric APS
    [49]
    4.2. Systemic Lupus Erythematosus
    Systemic lupus erythematosus (SLE) is a systemic autoimmune disease characterized
    by the production of autoantibodies against nuclear antigens and the deposition of immune
    complex (IC) leading to systemic inflammation and tissue damage. Lupus nephritis (LN)
    is one of the most severe organ manifestations and a primary cause of morbidity and
    mortality [50].
    Studies on circulating EV in SLE patients revealed divergent results regarding their
    circulating level and characterization. EV were found increased in SLE [51,52], or decreased
    compared to healthy controls [53]. Those different results reflect the lack of standardized
    methods for EV evaluation and characterization.
    In SLE, circulating EV expose chromatin on their surface and may represent a source
    of nuclear antigens which can bound both the IgG and the complement, resulting in the
    formation of IC, which correlate with the disease activity and the vascular damage [52,54].
    Circulating EV can bind both IgG and IgM to form immune complexes (EV-ICs) and
    EV-IgG+ were positively correlated with the disease activity. Platelet-derived EV (PEV),
    mainly PEV-IgG+, stimulated monocytes in vitro changing their phenotype and promoting
    their inflammatory response [52]. Several studies have supported the role of the deoxyribonuclease
    DNASE1L3 as a genetic determinant of susceptibility [55,56]. Sisirak et al. [57]
    have found that DNASE1L3 can digest chromatin in apoptotic cell-derived EV and, in the
    absence of DNASE1L3, EV-associated DNA may gather in an extracellular environment
    and promote autoantibody production by autoreactive B cells. DNASE1L3 is produced by
    dendritic cells and macrophages, and its circulating level was inversely correlated with
    anti-dsDNA level [57].
    Platelet activation plays a key role in the pathogenesis of SLE. They promote T and
    B cell activation, NETosis, type I IFN production, and dendritic cell activation resulting
    in systemic organ damage. Platelet-derived EV are the most prevalent circulating EV in
    healthy subjects, while conflicting results have been reported regarding platelet-derived
    EV in SLE. Burbano et al. [52] have found an increased number of circulating EV (mainly
    platelet-derived EV) in patients with SLE compared to healthy controls. However, plateletderived
    EV level was unrelated to disease activity measured with SLEDAI score. Lopez
    et al. [58] have observed an increased level of platelet-, monocyte- and T lymphocytederived
    EV. Interestingly, the authors found that EV level is influenced by the disease
    activity and is related to the activation status of blood parental cells. They also reported
    that glucocorticoid therapy may influence EV production and T cell activation, as EV
    from patients treated with glucocorticoids induced the upregulation of CD25 and the
    Int. J. Mol. Sci. 2021, 22, 4194 9 of 20
    accumulation of IL-10 in T cells [58]. Moreover, circulating platelet-derived EV correlated
    with endothelial-independent vasodilatation in SLE [59].
    Patients with SLE have increased cardiovascular risk due to platelet activation and
    systemic endothelial activation. EV may be involved in this process, as EV and EV-ICs
    from SLE patients can activate endothelial cells by increasing the expression of adhesion
    molecules (CD54, CD102), the production of chemokine (CCL2, CCL5, IL-6) and the
    adherence of monocytes. EV may also mediate endothelial injury by increasing endothelial
    permeability through the alteration of cytoskeletal proteins leading to adhesion and
    migration of monocyte to the inflamed organs [60].
    Polymorphonuclear leukocytes (PMNs) are greatly involved in SLE pathogenesis, as
    PMNs showed generalized hyperactivity, with enhanced apoptosis and increased production
    of neutrophil extracellular traps (NETs) [61]. Moreover, increased oxidative stress
    has been observed in SLE, which may contribute to immune dysregulation [62]. Winberg
    et al. [63] have found that EV from SLE patients induced ROS production in the patient’s
    own PMNs re-suspended in autologous serum, particularly in patients with low circulating
    C3 level, which reflects disease activity. The ROS production partly depends on EV properties,
    serum components (including autoantibodies) and PMN hyper-responsiveness [63].
    In vitro studies have shown the proinflammatory effect of EV on blood-derived plasmacytoid
    dendritic cells and myeloid dendritic cells by increasing the expression of costimulatory
    molecules (CD40, CD80, CD83, CD86) and the release of proinflammatory cytokines
    (IL-6, TNF, IFN ). Moreover, EV enhanced the formation of neutrophil extracellular traps
    (NETs) which may represent a source of nuclear autoantigens thus contributing to the
    pathogenesis of SLE and renal inflammation [64]. EV from patients with active LN contain
    a higher level of acetylated chromatin compared to patients with remissive LN, without
    LN, or healthy controls. Rother et al. [65] have found that the degree of EV acetylated
    chromatin determines their strength to stimulate neutrophils to form NETs.
    According to proteomic and flow cytometry analysis, circulating EV in SLE patients
    have an increased content of IgG and galectin-3 binding protein (G3BP), a glycoprotein
    that may contribute to the pathogenesis of SLE. G3BP is induced by type I IFN and exhibits
    a high binding capacity toward components of the glomerular basement membrane
    (GBM) [66], including collagen IV, fibronectin and galectin-3. Interestingly, patients suffering
    from lupus nephritis show a glomerular G3BP/IgG co-localization pattern specifically
    in the GBM, suggesting the presence of G3BP in the IC delivered either by EV from the
    circulation or locally formed [67]. In SLE patients, in vitro stimulation of peripheral blood
    mononuclear cells (PBMCs) with TLR-9 agonist increases the release of EV expressing both
    G3BP and dsDNA. This was particularly relevant in patients with active LN, compared to
    healthy donors [68].
    Urinary EV may reflect structural damage and renal dysfunction, and they have been
    investigated as potential LN diagnostic and prognostic biomarkers. Urinary podocytederived
    EV reflecting the glomerular podocyte damage are increased in LN and SLE
    patients compared to healthy controls. Interestingly, urinary EV level is correlated to the
    SLE disease activity index (SLEDAI) score, anti-dsDNA antibodies titer, proteinuria and
    histopathological lesions [69]. A recent study demonstrated that urinary EV expressing
    the high-mobility group box 1 molecule (HMGB1) are higher in SLE patients with active
    LN than in those without renal involvement, and correlate with proteinuria. HMGB1 is
    involved in the pathogenesis of several autoimmune diseases and it may be an important
    mediator in LN. Indeed, its expression is increased in glomerular endothelium and
    mesangium, and its blood and urinary level is increased in LN [70].
    Several studies have identified EV-derived miRNA as markers of renal damage which
    can also discriminate active LN [71]; miR-21, miR-150, and miR-29c were correlated to
    renal fibrosis and could predict the progression to the end-stage renal disease [72,73]. A
    unique circulating miRNA expression profile was detected in class IV LN [74] and urinary
    EV-derived miRNA have been proposed as peculiar biomarkers of class IV LN [75].
    Int. J. Mol. Sci. 2021, 22, 4194 10 of 20
    Recently, Garcia-Vives et al. investigated the miRNA expression profile of urinary EV
    in proliferative LN as a new potential prognostic biomarker. Patients with clinical responses
    to therapy are characterized by an increased level of miR-31, miR-107, and miR-135b-5p
    in urine and in renal tissue (mostly localized in epithelial tubular cells), compared to nonresponder
    patients. In vitro stimulation of tubular epithelial cells with proinflammatory
    cytokines increases the release of these miRNA, which can be taken up by endothelial cells
    and mesangial cells in responder patients [76].
    Recent studies have evaluated the role of mitochondria in autoimmune diseases.
    Activated cells can release EV containing mitochondria or free mitochondria which may
    stimulate immunity. Mobarretz et al. [77] have demonstrated the presence of circulating
    particles (approximately 3 m), and among them, a population of large EV carrying
    mitochondrial molecules (mitoEV) were found that both increased and associated with
    the disease activity in SLE patients. Furthermore, patients suffering from active LN have
    higher levels of mitoEV and IgG-coated mitoEV, suggesting that they may contribute to
    the formation of IC and thus be involved in renal damage [77].
    In conclusion, EV may play a crucial role in the pathogenesis of SLE, and particularly
    in LN-associated renal injury. For these reasons, EV have been proposed as potential
    biomarkers of disease in SLE patients as well as early biomarkers of renal damage in LN
    (Table 2).
    Table 2. Extracellular vesicles in systemic lupus erythematosus.
    Study EV Concentration Cellular Origin of EV EV Pathological Significance Reference
    Burbano, C.; et al.
    Increased in SLE
    compared to healthy
    controls
    platelet
    Formation of immune complexes,
    source of nuclear antigens,
    correlation with disease activity
    [52]
    López, P.; et al.
    Increased in SLE
    compared to healthy
    controls
    platelet, monocyte,
    T lymphocyte
    EV level correlated with:
    disease activity,
    glucocorticoid therapy,
    endothelial vasodilatation
    [58]
    Atehortúa, L.; et al. Endothelial cell activation,
    endothelial injury, [60]
    Winberg, L.-K.; et al.,
    Dieker, J.J.; et al.,
    Rother, N.; et al.
    In vitro stimulation of
    polymorphonuclear leukocytes with EV
    from SLE patients increased ROS
    production
    EV promote neutrophil activation and
    NETs production
    [63–65]
    Nielsen, C.T.; et al.,
    Rasmussen, N.S.; et al.
    IgG/galectin-3 binding protein (G3BP)+
    EV are involved in the pathogenesis of
    lupus nephritis
    [67,68]
    Lu, J.; et al.,
    Vanegas-García, A.; et al.
    Urinary podocyte-derived
    EV increased in SLE Urinary EV
    Urinary podocyte-derived EV level
    correlated with systemic disease activity
    and renal injury
    Urinary EV high-mobility group box 1
    molecule (HMGB1)+ were found to be
    higher in lupus nephritis
    [69,70]
    Felip, M.L.; et al.,
    Solé, C.; et al.,
    Navarro-Quiroz, E.; et al.,
    Li, Y.; et al.,
    Garcia-Vives, E.; et al.
    EV derived miRNA
    miR-21, miR-150, and miR-29c, miR-31,
    miR-107, and miR-135b-5p correlated
    with renal injury in lupus nephritis
    [72–76]
    Mobarrez, F.; et al.
    EV containing
    mitochondrial molecules
    (mitoEV)
    mitoEV were associated with disease
    activity, immune complex formation and
    renal damage
    [77]
    Int. J. Mol. Sci. 2021, 22, 4194 11 of 20
    4.3. Thrombotic Microangiopathies
    Thrombotic microangiopathies include different diseases characterized by microangiopathic
    hemolytic anemia and thrombocytopenia also potentially involving the kidney.
    Thrombotic microangiopathies include Shiga toxin-producing Escherichia coli (STEC-HUS)
    and thrombotic thrombocytopenic purpura (TTP).
    Haemolytic uremic syndrome (HUS) is characterized by nonimmune microangiopathic
    haemolytic anemia, thrombocytopenia and acute kidney injury. Typical HUS is
    subordinate to Shiga toxin-producing Escherichia coli (STEC-HUS) infection, which first
    colonizes the intestine and produces the toxin which enters the bloodstream and causes
    renal injury.
    In STEC-HUS, EV mainly derive from platelets, monocytes, neutrophils and red blood
    cells. Ståhl et al. have found EV expressing TF and phosphatidylserine potentially involved
    in the formation of microthrombi. Moreover, in the acute phase of the disease, circulating
    EV derived from platelets, monocytes, and neutrophils show deposition of C3 and C9
    on their surface. Interestingly, EV also express phosphatidylserine, which activates the
    coagulation factor V and X, thus enhancing and promoting thrombosis [78]. Those findings
    may reflect the systemic complement activation and the role of EV in the inflammatory and
    thrombogenic events in HUS [10].
    In vitro experiments showed that whole blood incubated with Shiga-toxin and/or
    STEC-lipopolysaccharide increased the release of TF-positive EV, C3- and C9-positive EV
    derived from platelets, monocytes and red blood cells [10,22]. It has been speculated that
    activated complement factors carried by EV can be transferred to recipient cells, driving
    cell damage [79].
    Since STEC are non-invasive bacteria, a small amount of Shiga toxin is present in the
    circulation; however, EV may transfer the toxin to the kidneys via peritubular capillaries.
    EV containing Shiga toxin were found within the kidney into renal cells, and in vivo
    experiments showed that EV enriched in Shiga toxin can reach renal cells through the
    glomerular and tubular basement membranes. In vitro studies also demonstrated that EV
    undergo endocytosis in glomerular endothelial cells, leading to cell damage [80].
    Shiga toxin interaction with circulating cells is mediated by two different receptors:
    the globotriaosylceramide (Gb3) toxin receptor and TLR4. Shiga toxin can be taken up
    by cells after binding to Gb3 or by cellular uptake of EV carrying the toxin derived from
    different host cells. EV expressing Shiga toxin are taken up by both Gb3-positive and
    Gb3-negative recipient cells. However, only Gb3-positive host cells are susceptible to
    toxin-induced cellular damage, reduced cellular metabolism and protein synthesis [81].
    Of note, renal endothelial cells express both Gb3 and TLR4. In vitro experiments using
    soluble Shiga toxin have shown that TLR4 acts as a Gb3 coreceptor, thus facilitating renal
    cell injury [79].
    TTP is a rare microangiopathic hemolytic anemia in which mutations of vWF protease
    (ADAMTS13) or autoantibodies against ADAMTS13 lead to the deposition of von
    Willebrand factor (vWF) multimers within capillaries. Systemic endothelial cell injury and
    platelet aggregation activate systemic microthrombosis mainly involving the brain and
    the kidney.
    A comparison of circulating EV in TTP patients and healthy subjects has shown
    that platelet-derived EV are the most represented EV subtype in both groups. Moreover,
    in TTP patients the platelet-derived EV level is significantly higher, as are EV express
    markers of platelet activation such as CD62p. Interestingly, platelet-derived EV have a
    procoagulant activity greater than platelets due to phospholipid expression on their surface
    which triggers the coagulation cascade [82].
    Other studies have found higher levels of platelet-derived and endothelial-derived
    EV in TTP. Endothelial-derived EV have shown procoagulant and proadhesive roles as
    they express CD62E (E-selectin), VWF, intercellular adhesion molecule 1 (ICAM-1), platelet
    endothelial cell adhesion molecules (PECAM-1; CD31) and endoglin (CD105) [83]. Tati
    et al. [84] have shown that circulating endothelial-derived EV are coated with C3 and
    Int. J. Mol. Sci. 2021, 22, 4194 12 of 20
    C9 and complement activated on platelets and glomerular endothelium. Complement
    activation may represent an ancillary phenomenon to platelet activation and endothelial
    injury, and thus may drive the microangiopathic process [84].
    Unfortunately, only a few studies have investigated the role of EV in TTP, and future
    research is needed to better understand the pathogenesis of TTP (Table 3).
    Table 3. Extracellular vesicles in thrombotic microangiopathies.
    Disease Study Cellular Origin of EV EV Biomarkers EV Pathological Significance Reference
    STEC-HUS Ståhl, A.-L.; et al.,
    Arvidsson, I.; et al.
    platelets, monocytes,
    neutrophils
    Tissue factor,
    phosphatidylserine
    (PS), C3, C9
    Promotion of thrombosis
    EV reflect complement
    activation
    [22,78]
    STEC-HUS
    Varrone, E.; et al.,
    Ståhl, A.-L.; et al.,
    Johansson, K.; et al.
    EV carrying Shiga
    toxin
    Delivery system of Shiga toxin
    to the kidney
    involvement in renal cell injury
    [79–81]
    TTP Tahmasbi, L.; et al.,
    Jimenez, J.J.; et al.
    platelets,
    endothelial
    cells
    CD62E (E-selectin),
    VWF, intercellular
    adhesion molecule 1
    (ICAM-1), platelet
    endothelial cell
    adhesion molecule
    (PECAM-1; CD31)
    and endoglin
    (CD105)
    Pro-coagulant and pro-adhesive
    roles [82,83]
    TTP Tati, R.; et al. Endothelial cells C3, C9 EV reflect complement
    activation [84]
    4.4. ANCA-Associated Vasculitis
    Systemic vasculitis consists of different syndromes characterized by blood vessel
    inflammation and multiple organ involvement. Small vessel vasculitis is often associated
    with anti-neutrophil cytoplasmic antibodies (ANCA), predominantly IgG autoantibodies
    directed against neutrophil cytoplasmatic constituents such as proteinase 3 (PR3, named
    cANCA) and myeloperoxidase (MPO, named pANCA). ANCA-associated vasculitis (AAV)
    comprises microscopic polyangiitis (MPA), granulomatosis with polyangiitis (Wegener’s)
    (GPA), and eosinophilic granulomatosis with polyangiitis (Churg-Strauss) (EGPA). AAV
    can affect small vessels in different organs resulting in pauci-immune glomerulonephritis,
    vasculitis involving the respiratory tract, and is associated with an increased risk of systemic
    thrombosis [85]. Despite new treatment options that have recently improved AAV
    prognosis, early diagnostic and prognostic biomarkers are still an unmet need.
    Patients with AAV have an increased risk of thromboembolic events due to the hypercoagulable
    state [86]. ANCA can induce neutrophil activation, neutrophil degranulation
    and release of neutrophil extracellular traps (NETs), which are involved in the development
    of vasculitis lesions [87]. Moreover, complement activation via the alternative pathway
    is crucial for the development of the disease [88]. In vitro experiments have found that
    both pANCA IgG and cANCA IgG can stimulate C5a-primed neutrophils to produce TFexpressing
    EV and TF-expressing NETs which, in turn, promote thrombin generation and
    the activation of the coagulation cascade [89]. Notably, Mendoza et al. [90] have measured
    the TF activity of EV and found higher TF activity in patients with AAV and associated
    venous thromboembolism, compared to patients without thrombotic events [90].
    Recent studies have demonstrated a potential role of platelet- and neutrophil-derived
    EV in the pathogenesis of vasculitis. Polyclonal ANCAs isolated from patients and chimeric
    PR3–ANCA induces the release of EV from primed neutrophils in vitro. Moreover, these EV
    are enriched in PR3 and MPO, exhibit thrombin-generating capacity, can bind the endothelium
    and induce its activation, induce ROS production and the release of proinflammatory
    cytokines (IL-6, IL-8) [91].
    An increased level of circulating platelet-, neutrophil- and endothelial-derived EV in
    patients with vasculitis has also been found [92]. In particular, a high level of neutrophil-EV
    Int. J. Mol. Sci. 2021, 22, 4194 13 of 20
    during the acute phase of the disease which decreased after steroid treatment has been
    reported, reflecting the key role of neutrophil activation. Of note, endothelial-EV level
    correlated with the Birmingham Activity Vasculitis Score (BVAS) and acute phase reactants,
    suggesting their potential application as disease biomarkers [92].
    The kinin system contributes to the inflammatory response and the development of
    vasculitis. The kinins regulate local blood pressure, promote inflammation, and capillary
    leakage. The kinins achieve their effect through B2 and B1 receptors. The B2 receptor is
    constitutively expressed and involved in inflammation and hyperalgesia, while the B1
    is upregulated during chronic inflammation (such as vasculitis), and controls neutrophil
    migration. Kahn et al. [93] have found increased circulating leukocyte-derived EV bearing
    the B1-kinin receptor during vasculitis. In particular, neutrophil-derived EV bearing the B1
    receptor were found docking to glomerular endothelial cells in kidney biopsies of patients
    with AAV. In vitro experiments showed that neutrophil-derived EV transfer functional B1-
    receptors to wild-type human embryonic kidney cells, which suggests a similar mechanism
    in vivo and could potentially promote kinin-associated inflammation. The observation
    that the main inhibitor of the kinin system, named the C1-inhibitor, is also involved in the
    inhibition of the release of EV enriched in B1-receptor has suggested a novel therapeutic
    target in vasculitis [93].
    Prikryl et al. [94] recently performed proteomic profiling of urinary EV isolated from
    patients with AAV and renal involvement and in healthy controls. The study showed
    different levels of proteins potentially involved in AAV pathogenesis. As an example, they
    found a significantly decreased level of Golgi mannosidases, such as MAN1A1, both in
    urinary EV and in the whole urine in active AAV. Interestingly, MAN1A1 is involved in
    protein glycosylation, which is considered to be involved in autoimmune disease and T cell
    activation, supporting its role in the pathogenesis of AAV. Moreover, they showed different
    levels of proteins related to neutrophil activation and degranulation, platelet regulation
    and podocyte-associated proteins, to name a few [94].
    Several studies have also shown that EV may contain enzymes linked to lipid metabo
    lism. Indeed, a comparison between EV in active GPA and healthy controls showed an
    increased content of leukotriene (LT)B4 and 5-oxo-eicosatetraenoic acid (5-oxo-ETE) in
    EV from GPA patients. Moreover, neutrophils primed with GM-CSF, and stimulated
    with EV recovered from GPA patients, generate ROS and release dsDNA. Interestingly,
    in vitro-primed neutrophils were stimulated by LTB4 and 5-oxo-ETE, thus EV carrying
    lipid enzymes may contribute to AAV pathogenesis [95]. In a recent study, Surmiak
    et al. [96] stimulated human umbilical endothelial cells (HUVEC) with EV from anti-
    PR3-activated neutrophils and analyzed their miRNA and mRNA content. They found a
    miRNA/mRNA profile consistent with the release of proinflammatory cytokines, which
    may be involved in endothelial injury in vasculitis. The most increased cytokines were
    IL-8, IL-33, Dickkopf-related protein 1 (DKK-1), soluble interleukin (IL)-1 like receptor-1
    (ST2) and angiopoietin-2. Interestingly this cytokine profile is similar to the circulating
    cytokine profile in GPA patients [96].
    Recent studies have investigated the role of EV in mediating endothelial injury in MPA.
    EV can be taken up by glomerular endothelial cells in vitro and can increase the release of
    soluble cellular adhesion molecules (sICAM-1 and sVCAM-1) leading to the injury of the
    glomerular endothelial barrier. A sequencing analysis of EV miRNA cargo in MPA patients
    revealed a different miRNA profile in MPA patients. In particular, a correlation between
    miR-185-3p, miR-125a-3p and clinical parameters, such as BVAS and 24-h urine proteins,
    was reported. Thus, the EV-miRNA content has been proposed as a biomarker of renal
    involvement in MPA [97].
    Circulating EV expressing MPO are elevated in AAV patients compared with healthy
    subjects. Interestingly, MPO+ EV expressing inflammatory biomarkers such as PTX3 and
    HMGB1 are associated with disease activity. Of interest, PTX3 is released by neutrophils
    during the inflammatory process, while HMGB1 may be associated with renal injury in
    AAV [98]. HMGB1 enhanced neutrophil activation and migration towards glomerular
    Int. J. Mol. Sci. 2021, 22, 4194 14 of 20
    endothelial cells in the presence of ANCA, leading to glomerular cell injury and the release
    of TF-positive EV and endothelin-1, which is involved in the fibrogenesis [99].
    The comparison of circulating EV expressing MPO in AAV patients and healthy
    controls reveals an increased expression of the complement components C3a and C5a on
    EV from AAV patients. Moreover, among AAV patients, C3a and C5a expression is higher
    in patients with active renal involvement compared to non-renal disease. Interestingly,
    the level of C3a and C5a expression on EV correlated with disease activity evaluated by
    BVAS [100].
    Platelet-derived EV were also found to be increased in AAV patients, particularly
    in MPO-positive patients with active disease in whom EV also expressed higher levels
    of chemokines, adhesion molecules, growth and apoptotic factors. Moreover, EV level
    correlated with the disease activity and the renal involvement, with serum creatinine and
    glomerular histologic lesions [101].
    Taken together, these results may provide insight into the role of EV in the pathogenesis
    of renal injury in AAV (Table 4).
    Table 4. Extracellular vesicles in ANCA-associated vasculitis.
    Study EV Biomarkers EV Cellular Origin Study Findings Reference
    Daniel, L.; et al. proteinase 3 (PR3),
    myeloperoxidase (MPO)
    EV released from primed
    neutrophils in vitro
    EV can induce endothelial activation,
    ROS production, cytokines release [91]
    Brogan, P.A.; et al.
    Platelets,
    neutrophils, endothelial
    cells
    EV level increased in vasculitis
    Decrease of neutrophil-derived EV
    after treatment
    Endothelial-derived EV correlated
    with disease activity
    [92]
    Kahn, R.; et al. B1 kinin receptor Leukocytes
    EV level increased in vasculitis
    Neutrophil-derived B1+ EV found
    on glomerular endothelial cells and
    renal injury
    [93]
    Prikryl, P.; et al. Urinary EV
    Proteomic EV profiling showed
    different regulation of proteins
    potentially involved in vasculitis
    pathogenesis
    [94]
    Surmiak, M.; et al.
    leukotriene (LT)B4,
    5-oxo-eicosatetraenoic
    acid (5-oxo-ETE)
    EV enriched in LTB4 and 5-oxo-ETE
    in granulomatosis with polyangiitis [95]
    Wang, Y.; et al.
    Sequencing analysis of EV miRNA
    cargo in microscopic polyangiitis
    identified a correlation between
    miR-185-3p, miR-125a-3p and both
    the clinical activity score and
    proteinuria
    [97]
    Manojlovic, M.; et al.
    myeloperoxidase (MPO),
    PTX3, high mobility group
    box 1 (HMGB1)
    PTX3+ and HMGB1+ EV correlated
    with disease activity
    HMGB1 potentially associated with
    renal injury
    [98]
    Antovic, A.; et al. myeloperoxidase (MPO),
    C3a, C5a
    MPO C3a+ and C5a+ EV increased
    in vasculitis, particularly in patients
    with renal involvement
    C3a and C5a expressed on EV
    correlated with disease activity
    [100]
    Miao, D.; et al.
    chemokines, adhesion
    molecules, growth and
    apoptotic factors
    Platelets
    Increased EV in vasculitis
    EV correlate with disease activity
    and renal injury
    [101]
    Int. J. Mol. Sci. 2021, 22, 4194 15 of 20
  6. Conclusions
    Recent studies have demonstrated the relevance of EV in physiological and pathological
    processes [7]. Due to their role in immune system modulation, EV are considered
    crucial actors in the pathogenesis of autoimmune diseases [1].
    Although current treatment options have improved their prognosis, autoimmune
    conditions remain rare diseases with high mortality and morbidity, particularly when
    the kidney is involved. Currently, kidney biopsy is the most important diagnostic and
    prognostic tool; however, recent investigations have focused on the identification and
    development of non-invasive biomarkers for early diagnosis and for the assessment of the
    disease activity.
    Recently, the impact of EV in autoimmune disease with renal involvement has been
    investigated [3]. EV numbers and features seem to correlate with pathological processes.
    In addition, EV have been reported to enhance the autoimmune process and promote
    renal injury. In APS, the EV circulating level has been shown to reflect endothelial and
    platelet chronic activation and may correlate with thrombotic events [6,44,49]. Moreover,
    EV can actively contribute to SLE pathogenesis as an autoantigen source, part of the
    immune complexes, endothelial and leukocytes activation promoters [52,63]. Of interest,
    the galectin-3 binding protein (G3BP)+ EV and urinary high-mobility group box 1 molecule
    (HMGB1)+ EV seem to be involved in lupus nephritis pathogenesis [67,68,70]. Likewise,
    EV contribute to renal injury in STEC-HUS [79–81]. In ANCA-associated vasculitis, EV are
    involved in endothelial activation and correlate with disease activity [91,94]. Of interest,
    HMGB1+ EV and B1-receptor + EV can be directly involved in renal cell injury and
    have been proposed as a novel therapeutic target [93,98]. Moreover, miRNA carried
    by circulating EV can be investigated as novel diagnostic biomarkers for both lupus
    nephritis [72,74,76] and ANCA-associated vasculitis [97].
    Encouraging discoveries on EV as diagnostic tools have been provided; however, being
    a recent research field, contradictory results reflecting the lack of standardized evaluation
    methods make their application in the clinical practice still challenging. Future studies
    should focus on defining standardized methods of EV collection and cargo evaluation. Of
    note, interesting observations have been reported in the research field, which need further
    investigation to be transferred in clinical settings.
    However, the rareness of these diseases made it difficult to investigate the potential
    clinical impact of EV as diagnostic and prognostic tools in large clinical studies. For this
    reason, multicenter studies are needed to collect relevant data.
    Despite these limitations, future studies on the role of EV in renal pathology should
    be pursued to better identify new targets in autoimmune diseases. Overall, the future
    challenge is to develop tools exploiting EV as diagnostic biomarkers, therapeutic targets or
    drug vectors for novel treatment options in autoimmune renal diseases.
    Author Contributions: M.M. contributed to data curation and writing the original draft; G.C. contributed
    to editing; M.F.B. contributed to writing, visualization, founding and editing. All authors
    have read and agreed to the published version of the manuscript.
    Funding: This work has been supported by grants obtained by MFB from Ministero dell’Istruzione,
    Università e Ricerca (MIUR) ex 60%.
    Institutional Review Board Statement: Not applicable.
    Informed Consent Statement: Not applicable.
    Data Availability Statement: Not applicable.
    Conflicts of Interest: G.C. is a component of the Scientific Advisory Board of UNICYTE. The other
    authors have no conflict of interest.
    Int. J. Mol. Sci. 2021, 22, 4194 16 of 20
    References
  7. Turpin, D.; Truchetet, M.-E.; Faustin, B.; Augusto, J.-F.; Contin-Bordes, C.; Brisson, A.; Blanco, P.; Duffau, P. Role of extracellular
    vesicles in autoimmune diseases. Autoimmun. Rev. 2016, 15, 174–183. [CrossRef]
  8. Théry, C.; Witwer, K.W.; Aikawa, E.; Alcaraz, M.J.; Anderson, J.D.; Andriantsitohaina, R.; Antoniou, A.; Arab, T.; Archer, F.;
    Atkin-Smith, G.K.; et al. Minimal information for studies of extracellular vesicles 2018 (MISEV2018): A position statement of the
    International Society for Extracellular Vesicles and update of the MISEV2014 guidelines. J. Extracell. Vesicles 2018, 7, 1535750.
    [CrossRef] [PubMed]
  9. Ståhl, A.-L.; Johansson, K.; Mossberg, M.; Kahn, R.; Karpman, D. Exosomes and microvesicles in normal physiology, pathophysiology,
    and renal diseases. Pediatr. Nephrol. 2019, 34, 11–30. [CrossRef] [PubMed]
  10. Yáñez-Mó, M.; Siljander, P.R.M.; Andreu, Z.; Zavec, A.B.; Borràs, F.E.; Buzas, E.I.; Buzas, K.; Casal, E.; Cappello, F.; Carvalho,
    J.; et al. Biological Properties of Extracellular Vesicles and their Physiological Functions. J. Extracell. Vesicles 2015, 4, 27066.
    [CrossRef] [PubMed]
  11. Coumans, F.A.W.; Brisson, A.R.; Buzas, E.I.; Dignat-George, F.; Drees, E.E.E.; El-Andaloussi, S.; Emanueli, C.; Gasecka, A.;
    Hendrix, A.; Hill, A.F.; et al. Methodological Guidelines to Study Extracellular Vesicles. Circ. Res. 2017, 120, 1632–1648. [CrossRef]
    [PubMed]
  12. Štok, U.; Blokar, E.; Lenassi, M.; Holcar, M.; Frank-Bertoncelj, M.; Erman, A.; Resnik, N.; Sodin-Šemrl, S.; Cˇ ucˇnik, S.; Pirkmajer,
    K.P.; et al. Characterization of Plasma-Derived Small Extracellular Vesicles Indicates Ongoing Endothelial and Platelet Activation
    in Patients with Thrombotic Antiphospholipid Syndrome. Cells 2020, 9, 1211. [CrossRef] [PubMed]
  13. Buzas, E.I.; György, B.; Nagy, G.; Falus, A.; Gay, S. Emerging role of extracellular vesicles in inflammatory diseases. Nat. Rev.
    Rheumatol. 2014, 10, 356–364. [CrossRef]
  14. Pomatto, M.A.C.; Gai, C.; Bussolati, B.; Camussi, G. Extracellular Vesicles in Renal Pathophysiology. Front. Mol. Biosci. 2017, 4, 37.
    [CrossRef]
  15. Owens, A.P., III; Mackman, N.; Weber, C.; Mause, S. Microparticles in hemostasis and thrombosis. Circ. Res. 2011, 108, 1284–1297.
    [CrossRef]
  16. Ståhl, A.-L.; Sartz, L.; Nelsson, A.; Békássy, Z.D.; Karpman, D. Shiga Toxin and Lipopolysaccharide Induce Platelet-Leukocyte
    Aggregates and Tissue Factor Release, a Thrombotic Mechanism in Hemolytic Uremic Syndrome. PLoS ONE 2009, 4, e6990.
    [CrossRef]
  17. Sinauridze, E.; Kireev, D.; Popenko, N.Y.; Pichugin, A.V.; Panteleev, M.; Krymskaya, O.V.; Ataullakhanov, F. Platelet microparticle
    membranes have 50- to 100-fold higher specific procoagulant activity than activated platelets. Thromb. Haemost. 2007, 97, 425–434.
  18. Pitanga, T.N.; França, L.D.A.; Rocha, V.C.J.; Meirelles, T.; Borges, V.M.; Gonçalves, M.S.; Pontes-De-Carvalho, L.C.; Noronha-Dutra,
    A.A.; Dos-Santos, W.L.C. Neutrophil-derived microparticles induce myeloperoxidase-mediated damage of vascular endothelial
    cells. BMC Cell Biol. 2014, 15, 21. [CrossRef]
  19. Lombardo, G.; Dentelli, P.; Togliatto, G.; Rosso, A.; Gili, M.; Gallo, S.; Deregibus, M.C.; Camussi, G.; Brizzi, M.F. Activated Stat5
    trafficking Via Endothelial Cell-derived Extracellular Vesicles Controls IL-3 Pro-angiogenic Paracrine Action. Sci. Rep. 2016,
    6, 25689. [CrossRef]
  20. 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. Am. J. Pathol. 2002, 160, 673–680.
    [CrossRef]
  21. Yang, C.; Mwaikambo, B.R.; Zhu, T.; Gagnon, C.; LaFleur, J.; Seshadri, S.; Lachapelle, P.; Lavoie, J.-C.; Chemtob, S.; Hardy,
    P. Lymphocytic microparticles inhibit angiogenesis by stimulating oxidative stress and negatively regulating VEGF-induced
    pathways. Am. J. Physiol. Regul. Integr. Comp. Physiol. 2008, 294, R467–R476. [CrossRef] [PubMed]
  22. Ezhang, B.; Eyin, Y.; Elai, R.C.; Elim, S.K. Immunotherapeutic Potential of Extracellular Vesicles. Front. Immunol. 2014, 5, 518.
    [CrossRef]
  23. Anel, A.; Gallego-Lleyda, A.; De Miguel, D.; Naval, J.; Martínez-Lostao, L. Role of Exosomes in the Regulation of T-cell Mediated
    Immune Responses and in Autoimmune Disease. Cells 2019, 8, 154. [CrossRef] [PubMed]
  24. Cloutier, N.; Tan, S.; Boudreau, L.H.; Cramb, C.; Subbaiah, R.; Lahey, L.; Albert, A.; Shnayder, R.; Gobezie, R.; Nigrovic,
    P.A.; et al. The exposure of autoantigens by microparticles underlies the formation of potent inflammatory components: The
    microparticle-associated immune complexes. EMBO Mol. Med. 2012, 5, 235–249. [CrossRef] [PubMed]
  25. Mobarrez, F.; Gunnarsson, I.; Svenungsson, E. Altered 2-glycoprotein I expression on microparticles in the presence of
    antiphospholipid antibodies. J. Thromb. Haemost. 2017, 15, 1799–1806. [CrossRef]
  26. Mesri, M.; Altieri, D.C. Endothelial cell activation by leukocyte microparticles. J. Immunol. 1998, 161, 4382–4387.
  27. Munich, S.; Sobo-Vujanovic, A.; Buchser, W.J.; Beer-Stolz, D.; Vujanovic, N.L. Dendritic cell exosomes directly kill tumor cells and
    activate natural killer cells via TNF superfamily ligands. OncoImmunology 2012, 1, 1074–1083. [CrossRef]
  28. Ståhl, A.-L.; Sartz, L.; Karpman, D. Complement activation on platelet-leukocyte complexes and microparticles in enterohemorrhagic
    Escherichia coli–induced hemolytic uremic syndrome. Blood 2011, 117, 5503–5513. [CrossRef]
  29. Clayton, A.; Harris, C.L.; Court, J.; Mason, M.D.; Morgan, B.P. Antigen-presenting cell exosomes are protected from complementmediated
    lysis by expression of CD55 and CD59. Eur. J. Immunol. 2003, 33, 522–531. [CrossRef] [PubMed]
  30. Van Balkom, B.W.; Pisitkun, T.; Verhaar, M.C.; Knepper, M.A. Exosomes and the kidney: Prospects for diagnosis and therapy of
    renal diseases. Kidney Int. 2011, 80, 1138–1145. [CrossRef]
    Int. J. Mol. Sci. 2021, 22, 4194 17 of 20
  31. Hogan, M.C.; Johnson, K.L.; Zenka, R.M.; Charlesworth, M.C.; Madden, B.J.; Mahoney, D.W.; Oberg, A.L.; Huang, B.Q.;
    Leontovich, A.A.; Nesbitt, L.L.; et al. Subfractionation, characterization, and in-depth proteomic analysis of glomerular membrane
    vesicles in human urine. Kidney Int. 2014, 85, 1225–1237. [CrossRef]
  32. Moon, P.-G.; Lee, J.-E.; You, S.; Kim, T.-K.; Cho, J.-H.; Kim, I.-S.; Kwon, T.-H.; Kim, C.-D.; Park, S.-H.; Hwang, D.; et al. Proteomic
    analysis of urinary exosomes from patients of early IgA nephropathy and thin basement membrane nephropathy. Proteom. 2011,
    11, 2459–2475. [CrossRef] [PubMed]
  33. Ranghino, A.; DiMuccio, V.; Papadimitriou, E.; Bussolati, B. Extracellular vesicles in the urine: Markers and mediators of tissue
    damage and regeneration. Clin. Kidney J. 2015, 8, 23–30. [CrossRef] [PubMed]
  34. Gonzales, P.A.; Pisitkun, T.; Hoffert, J.D.; Tchapyjnikov, D.; Star, R.A.; Kleta, R.;Wang, N.S.; Knepper, M.A. Large-Scale Proteomics
    and Phosphoproteomics of Urinary Exosomes. J. Am. Soc. Nephrol. 2009, 20, 363–379. [CrossRef] [PubMed]
  35. Pisitkun, T.; Shen, R.-F.; Knepper, M.A. Identification and proteomic profiling of exosomes in human urine. Proc. Natl. Acad. Sci.
    USA 2004, 101, 13368–13373. [CrossRef]
  36. Salih, M.; Zietse, R.; Hoorn, E.J. Urinary extracellular vesicles and the kidney: Biomarkers and beyond. Am. J. Physiol. Physiol.
    2014, 306, F1251–F1259. [CrossRef]
  37. Oosthuyzen, W.; Scullion, K.M.; Ivy, J.R.; Morrison, E.E.; Hunter, R.W.; Lewis, P.J.S.; O’Duibhir, E.; Street, J.M.; Caporali, A.;
    Gregory, C.D.; et al. Vasopressin Regulates Extracellular Vesicle Uptake by Kidney Collecting Duct Cells. J. Am. Soc. Nephrol.
    2016, 27, 3345–3355. [CrossRef] [PubMed]
  38. Gildea, J.J.; Seaton, J.E.; Victor, K.G.; Reyes, C.M.;Wang, D.B.; Pettigrew, A.C.; Courtner, C.E.; Shah, N.; Tran, H.T.; Van Sciver,
    R.E.; et al. Exosomal transfer from human renal proximal tubule cells to distal tubule and collecting duct cells. Clin. Biochem.
    2014, 47, 89–94. [CrossRef] [PubMed]
  39. Kholia, S.; Sanchez, M.B.H.; Cedrino, M.; Papadimitriou, E.; Tapparo, M.; Deregibus, M.C.; Bruno, S.; Antico, F.; Brizzi, M.F.;
    Quesenberry, P.J.; et al. Mesenchymal Stem Cell Derived Extracellular Vesicles Ameliorate Kidney Injury in Aristolochic Acid
    Nephropathy. Front. Cell Dev. Biol. 2020, 8, 1–17. [CrossRef] [PubMed]
  40. Ranghino, A.; Bruno, S.; Bussolati, B.; Moggio, A.; DiMuccio, V.; Tapparo, M.; Biancone, L.; Gontero, P.; Frea, B.; Camussi, G. The
    effects of glomerular and tubular renal progenitors and derived extracellular vesicles on recovery from acute kidney injury. Stem
    Cell Res. Ther. 2017, 8, 24. [CrossRef]
  41. Grange, C.; Papadimitriou, E.; Dimuccio, V.; Pastorino, C.; Molina, J.; O’Kelly, R.; Niedernhofer, L.J.; Robbins, P.D.; Camussi, G.;
    Bussolati, B. Urinary Extracellular Vesicles Carrying Klotho Improve the Recovery of Renal Function in an Acute Tubular Injury
    Model. Mol. Ther. 2020, 28, 490–502. [CrossRef] [PubMed]
  42. Borges, F.T.; Melo, S.A.; Özdemir, B.C.; Kato, N.; Revuelta, I.; Miller, C.A.; Ii, V.H.G.; LeBleu, V.S.; Kalluri, R. TGF- 1–Containing
    Exosomes from Injured Epithelial Cells Activate Fibroblasts to Initiate Tissue Regenerative Responses and Fibrosis. J. Am. Soc.
    Nephrol. 2013, 24, 385–392. [CrossRef]
  43. Jella, K.K.; Yu, L.; Yue, Q.; Friedman, D.; Duke, B.J.; Alli, A.A. Exosomal GAPDH from Proximal Tubule Cells Regulate ENaC
    Activity. PLoS ONE 2016, 11, e0165763. [CrossRef]
  44. Street, J.M.; Birkhoff,W.; Menzies, R.I.;Webb, D.J.; Bailey, M.A.; Dear, J.W. Exosomal transmission of functional aquaporin 2 in
    kidney cortical collecting duct cells. J. Physiol. 2011, 589, 6119–6127. [CrossRef]
  45. Hiemstra, T.F.; Charles, P.D.; Gracia, T.; Hester, S.S.; Gatto, L.; Al-Lamki, R.; Floto, R.A.; Su, Y.; Skepper, J.N.; Lilley, K.S.; et al.
    Human Urinary Exosomes as Innate Immune Effectors. J. Am. Soc. Nephrol. 2014, 25, 2017–2027. [CrossRef]
  46. Chiabotto, G.; Bruno, S.; Collino, F.; Camussi, G. Mesenchymal Stromal Cells Epithelial Transition Induced by Renal Tubular
    Cells-Derived Extracellular Vesicles. PLoS ONE 2016, 11, e0159163. [CrossRef]
  47. Biancone, L.; Bruno, S.; Deregibus, M.C.; Tetta, C.; Camussi, G. Therapeutic potential of mesenchymal stem cell-derived
    microvesicles. Nephrol. Dial. Transplant. 2012, 27, 3037–3042. [CrossRef] [PubMed]
  48. Gregorini, M.; Corradetti, V.; Pattonieri, E.F.; Rocca, C.; Milanesi, S.; Peloso, A.; Canevari, S.; De Cecco, L.; Dugo, M.; Avanzini,
    M.A.; et al. Perfusion of isolated rat kidney with Mesenchymal Stromal Cells/Extracellular Vesicles prevents ischaemic injury. J.
    Cell. Mol. Med. 2017, 21, 3381–3393. [CrossRef]
  49. Cervera, R.; Serrano, R.; Pons-Estel, G.; Ceberio-Hualde, L.; Shoenfeld, Y.; De Ramón, E.; Buonaiuto, V.; Jacobsen, S.; Zeher, M.;
    Tarr, T.; et al. Morbidity and mortality in the antiphospholipid syndrome during a 10-year period: A multicentre prospective
    study of 1000 patients. Ann. Rheum. Dis. 2015, 74, 1011–1018. [CrossRef]
  50. Alluri, R.; Chaturvedi, S.; McCrae, K.R. Extracellular Vesicles in the Antiphospholipid Syndrome. Semin. Thromb. Hemost. 2018,
    44, 493–504. [CrossRef]
  51. Breen, K.A.; Sanchez, K.; Kirkman, N.; Seed, P.T.; Parmar, K.; Moore, G.W.; Hunt, B.J. Endothelial and platelet microparticles in
    patients with antiphospholipid antibodies. Thromb. Res. 2014, 135, 368–374. [CrossRef]
  52. Camoin-Jau, L.; Sabatier, F.; Arnoux, D.; Anfosso, F.; Bardin, N.; Veit, V.; Combes, V.; Gentile, S.; Moal, V.; Sanmarco, M.; et al.
    Endothelial microparticles: A potential contribution to the thrombotic complications of the antiphospholipid syndrome. Thromb.
    Haemost. 2004, 91, 667–673. [CrossRef] [PubMed]
  53. Vikerfors, A.; Mobarrez, F.; Bremme, K.; Holmström, M.; Ågren, A.; Eelde, A.; Bruzelius, M.; Antovic, A.;Wallén, H.; Svenungsson,
    E. Studies of microparticles in patients with the antiphospholipid syndrome (APS). Lupus 2012, 21, 802–805. [CrossRef] [PubMed]
  54. Willemze, R.; Bradford, R.L.; Mooberry, M.J.; Roubey, R.A.S.; Key, N.S. Plasma microparticle tissue factor activity in patients with
    antiphospholipid antibodies with and without clinical complications. Thromb. Res. 2013, 133, 187–189. [CrossRef] [PubMed]
    Int. J. Mol. Sci. 2021, 22, 4194 18 of 20
  55. Campello, E.; Radu, C.M.; Tonello, M.; Kuzenko, A.; Bulato, C.; Hoxha, A.; Mattia, E.; Spiezia, L.; Ruffatti, A.; Simioni, P.
    Circulating microparticles in pregnant patients with primary anti-phospholipid syndrome: An exploratory study. Scand. J.
    Rheumatol. 2018, 47, 501–504. [CrossRef] [PubMed]
  56. Hahn, B.H.; McMahon, M.A.;Wilkinson, A.;Wallace,W.D.; Daikh, D.I.; Fitzgerald, J.D.; Karpouzas, G.A.; Merrill, J.T.;Wallace,
    D.J.; Yazdany, J.; et al. American College of Rheumatology guidelines for screening, treatment, and management of lupus
    nephritis. Arthritis Rheum. 2012, 64, 797–808. [CrossRef] [PubMed]
  57. Mobarrez, F.; Vikerfors, A.; Gustafsson, J.T.; Gunnarsson, I.; Zickert, A.; Larsson, A.; Pisetsky, D.S.; Wallén, H.; Svenungsson,
    E. Microparticles in the blood of patients with systemic lupus erythematosus (SLE): Phenotypic characterization and clinical
    associations. Sci. Rep. 2016, 6, 36025. [CrossRef] [PubMed]
  58. Burbano, C.; Villar-Vesga, J.; Orejuela, J.; Muñoz, C.; Vanegas, A.; Vásquez, G.; Rojas, M.; Castaño, D. Potential Involvement of
    Platelet-Derived Microparticles and Microparticles Forming Immune Complexes during Monocyte Activation in Patients with
    Systemic Lupus Erythematosus. Front. Immunol. 2018, 9, 322. [CrossRef]
  59. Nielsen, C.T.; Østergaard, O.; Johnsen, C.; Jacobsen, S.; Heegaard, N.H.H. Distinct features of circulating microparticles and their
    relationship to clinical manifestations in systemic lupus erythematosus. Arthritis Rheum. 2011, 63, 3067–3077. [CrossRef]
  60. Ullal, A.J.; Reich, C.F.; Clowse, M.; Criscione-Schreiber, L.G.; Tochacek, M.; Monestier, M.; Pisetsky, D.S. Microparticles as
    antigenic targets of antibodies to DNA and nucleosomes in systemic lupus erythematosus. J. Autoimmun. 2011, 36, 173–180.
    [CrossRef]
  61. Al-Mayouf, S.M.; Sunker, A.; Abdwani, R.; Al Abrawi, S.; Almurshedi, F.; Alhashmi, N.; Al Sonbul, A.; Sewairi, W.; Qari, A.;
    Abdallah, E.; et al. Loss-of-function variant in DNASE1L3 causes a familial form of systemic lupus erythematosus. Nat. Genet.
    2011, 43, 1186–1188. [CrossRef]
  62. Ueki, M.; Takeshita, H.; Fujihara, J.; Iida, R.; Yuasa, I.; Kato, H.; Panduro, A.; Nakajima, T.; Kominato, Y.; Yasuda, T. Caucasianspecific
    allele in non-synonymous single nucleotide polymorphisms of the gene encoding deoxyribonuclease I-like 3, potentially
    relevant to autoimmunity, produces an inactive enzyme. Clin. Chim. Acta 2009, 407, 20–24. [CrossRef]
  63. Sisirak, V.; Sally, B.; D’Agati, V.; Martinez-Ortiz, W.; Özçakar, Z.B.; David, J.; Rashidfarrokhi, A.; Yeste, A.; Panea, C.; Chida, A.S.;
    et al. Digestion of Chromatin in Apoptotic Cell Microparticles Prevents Autoimmunity. Cell 2016, 166, 88–101. [CrossRef]
  64. López, P.; Rodríguez-Carrio, J.; Martínez-Zapico, A.; Caminal-Montero, L.; Suárez, A. Circulating microparticle subpopulations
    in systemic lupus erythematosus are affected by disease activity. Int. J. Cardiol. 2017, 236, 138–144. [CrossRef]
  65. McCarthy, E.; Moreno-Martinez, D.; Wilkinson, F.; McHugh, N.; Bruce, I.; Pauling, J.; Alexander, M.; Parker, B. Microparticle
    subpopulations are potential markers of disease progression and vascular dysfunction across a spectrum of connective tissue
    disease. BBA Clin. 2017, 7, 16–22. [CrossRef] [PubMed]
  66. Atehortúa, L.; Rojas, M.; Vásquez, G.; Muñoz-Vahos, C.H.; Vanegas-García, A.; Posada-Duque, R.A.; Castaño, D. Endothelial
    activation and injury by microparticles in patients with systemic lupus erythematosus and rheumatoid arthritis. Arthritis Res.
    2019, 21, 1–15. [CrossRef] [PubMed]
  67. Kaplan, M.J. Neutrophils in the pathogenesis and manifestations of SLE. Nat. Rev. Rheumatol. 2011, 7, 691–699. [CrossRef]
    [PubMed]
  68. Perl, A. Oxidative stress in the pathology and treatment of systemic lupus erythematosus. Nat. Rev. Rheumatol. 2013, 9, 674–686.
    [CrossRef]
  69. Winberg, L.K.; Jacobsen, S.; Nielsen, C.H. Microparticles from patients with systemic lupus erythematosus induce production of
    reactive oxygen species and degranulation of polymorphonuclear leukocytes. Arthritis Res. 2017, 19, 230. [CrossRef] [PubMed]
  70. Dieker, J.J.; Tel, J.J.; Pieterse, E.E.; Thielen, A.; Rother, N.N.; Bakker, M.; Fransen, J.J.; Dijkman, H.B.P.M.; Berden, J.H.; De Vries,
    J.M.; et al. Circulating Apoptotic Microparticles in Systemic Lupus Erythematosus Patients Drive the Activation of Dendritic Cell
    Subsets and Prime Neutrophils for NETosis. Arthritis Rheumatol. 2016, 68, 462–472. [CrossRef]
  71. Rother, N.; Pieterse, E.; Lubbers, J.; Hilbrands, L.; Van Der Vlag, J. Acetylated Histones in Apoptotic Microparticles Drive the
    Formation of Neutrophil Extracellular Traps in Active Lupus Nephritis. Front. Immunol. 2017, 8, 1136. [CrossRef]
  72. Sasaki, T.; Brakebusch, C.; Engel, J.; Timpl, R. Mac-2 binding protein is a cell-adhesive protein of the extracellular matrix which
    self-assembles into ring-like structures and binds beta1 integrins, collagens and fibronectin. EMBO J. 1998, 17, 1606–1613.
    [CrossRef]
  73. Nielsen, C.T.; Østergaard, O.; Rekvig, O.P.; Sturfelt, G.; Jacobsen, S.; Heegaard, N.H.H. Galectin-3 binding protein links circulating
    microparticles with electron dense glomerular deposits in lupus nephritis. Lupus 2015, 24, 1150–1160. [CrossRef] [PubMed]
  74. Rasmussen, N.S.; Nielsen, C.T.; Jacobsen, S. Microvesicles in active lupus nephritis show Toll-like receptor 9-dependent coexpression
    of galectin-3 binding protein and double-stranded DNA. Clin. Exp. Immunol. 2021, 204, 64–77. [CrossRef] [PubMed]
  75. Lu, J.; Hu, Z.B.; Chen, P.P.; Lu, C.C.; Zhang, J.X.; Li, X.Q.; Yuan, B.Y.; Huang, S.J.; Ma, K.L. Urinary podocyte microparticles
    are associated with disease activity and renal injury in systemic lupus erythematosus. BMC Nephrol. 2019, 20, 303. [CrossRef]
    [PubMed]
  76. Burbano, C.; Gómez-Puerta, J.; Muñoz-Vahos, C.; Vanegas-García, A.; Rojas, M.; Vásquez, G.; Castaño, D. HMGB1+ microparticles
    present in urine are hallmarks of nephritis in patients with systemic lupus erythematosus. Eur. J. Immunol. 2019, 49, 323–335.
    [CrossRef] [PubMed]
  77. Perez-Hernandez, J.; Forner, M.J.; Pinto, C.; Chaves, F.J.; Cortes, R.; Redon, J. Increased Urinary Exosomal MicroRNAs in Patients
    with Systemic Lupus Erythematosus. PLoS ONE 2015, 10, e0138618. [CrossRef] [PubMed]
    Int. J. Mol. Sci. 2021, 22, 4194 19 of 20
  78. Solé, C.; Cortés-Hernández, J.; Felip, M.L.; Vidal, M.; Ordi-Ros, J. miR-29c in urinary exosomes as predictor of early renal fibrosis
    in lupus nephritis. Nephrol. Dial. Transplant. 2015, 30, 1488–1496. [CrossRef] [PubMed]
  79. Solé, C.; Moliné, T.; Vidal, M.; Ordi-Ros, J.; Cortés-Hernández, J. An Exosomal Urinary miRNA Signature for Early Diagnosis of
    Renal Fibrosis in Lupus Nephritis. Cells 2019, 8, 773. [CrossRef] [PubMed]
  80. Navarro-Quiroz, E.; Pacheco-Lugo, L.; Navarro-Quiroz, R.; Lorenzi, H.; España-Puccini, P.; Díaz-Olmos, Y.; Almendrales, L.;
    Olave, V.; Gonzalez-Torres, H.; Diaz-Perez, A.; et al. Profiling analysis of circulating microRNA in peripheral blood of patients
    with class IV lupus nephritis. PLoS ONE 2017, 12, e0187973. [CrossRef] [PubMed]
  81. Li, Y.; Xu, X.; Tang, X.; Bian, X.; Shen, B.; Zhao, H.; Luo, S.; Chen, Z.; Zhang, K. MicroRNA expression profile of urinary exosomes
    in Type IV lupus nephritis complicated by cellular crescent. J. Biol. Res. 2018, 25, 1–13. [CrossRef]
  82. Garcia-Vives, E.; Solé, C.; Moliné, T.; Vidal, M.; Agraz, I.; Ordi-Ros, J.; Cortés-Hernández, J. The Urinary Exosomal miRNA
    Expression Profile is Predictive of Clinical Response in Lupus Nephritis. Int. J. Mol. Sci. 2020, 21, 1372. [CrossRef]
  83. Mobarrez, F.; Fuzzi, E.; Gunnarsson, I.; Larsson, A.; Eketjäll, S.; Pisetsky, D.S.; Svenungsson, E. Microparticles in the blood of
    patients with SLE: Size, content of mitochondria and role in circulating immune complexes. J. Autoimmun. 2019, 102, 142–149.
    [CrossRef]
  84. Arvidsson, I.; Ståhl, A.-L.; Hedström, M.M.; Kristoffersson, A.-C.; Rylander, C.; Westman, J.S.; Storry, J.R.; Olsson, M.L.; Karpman,
    D. Shiga Toxin–Induced Complement-Mediated Hemolysis and Release of Complement-Coated Red Blood Cell–Derived
    Microvesicles in Hemolytic Uremic Syndrome. J. Immunol. 2015, 194, 2309–2318. [CrossRef]
  85. Varrone, E.; Carnicelli, D.; Brigotti, M. Extracellular Vesicles and Renal Endothelial Cells. Am. J. Pathol. 2021, 1–10. [CrossRef]
  86. Ståhl, A.-L.; Arvidsson, I.; Johansson, K.E.; Chromek, M.; Rebetz, J.; Loos, S.; Kristoffersson, A.-C.; Békássy, Z.D.; Mörgelin, M.;
    Karpman, D. A Novel Mechanism of Bacterial Toxin Transfer within Host Blood Cell-Derived Microvesicles. PLoS Pathog. 2015,
    11, e1004619. [CrossRef]
  87. Johansson, K.; Willysson, A.; Kristoffersson, A.-C.; Tontanahal, A.; Gillet, D.; Ståhl, A.-L.; Karpman, D. Shiga Toxin-Bearing
    Microvesicles Exert a Cytotoxic Effect on Recipient Cells Only When the Cells Express the Toxin Receptor. Front. Cell. Infect.
    Microbiol. 2020, 10, 212. [CrossRef] [PubMed]
  88. Tahmasbi, L.; Karimi, M.; Kafiabadi, S.A.; Nikougoftar, M.; Haghpanah, S.; Ranjbaran, R.; Moghadam, M. Evaluation of Plasma
    Platelet Microparticles in Thrombotic Thrombocytopenic Purpura. Ann. Clin. Lab. Sci. 2017, 47, 62–67. [PubMed]
  89. Jimenez, J.J.; Jy, W.; Mauro, L.M.; Horstman, L.L.; Soderland, C.; Ahn, Y.S. Endothelial microparticles released in thrombotic
    thrombocytopenic purpura express von Willebrand factor and markers of endothelial activation. Br. J. Haematol. 2003, 123,
    896–902. [CrossRef]
  90. Tati, R.; Kristoffersson, A.-C.; Ståhl, A.-L.; Rebetz, J.; Wang, L.; Licht, C.; Motto, D.; Karpman, D. Complement Activation
    Associated with ADAMTS13 Deficiency in Human and Murine Thrombotic Microangiopathy. J. Immunol. 2013, 191, 2184–2193.
    [CrossRef]
  91. Jennette, J.C.; Falk, R.J.; Bacon, P.A.; Basu, N.; Cid, M.C.; Ferrario, F.; Flores-Suarez, L.F.; Gross,W.L.; Guillevin, L.; Hagen, E.C.;
    et al. 2012 Revised International Chapel Hill Consensus Conference Nomenclature of Vasculitides. Arthritis Rheum. 2013, 65, 1–11.
    [CrossRef] [PubMed]
  92. Weidner, S.; Hafezi-Rachti, S.; Rupprecht, H.D. Thromboembolic events as a complication of antineutrophil cytoplasmic antibody–
    associated vasculitis. Arthritis Rheum. 2006, 55, 146–149. [CrossRef]
  93. Cartin-Ceba, R.; Peikert, T.; Specks, U. Pathogenesis of ANCA-Associated Vasculitis. Curr. Rheumatol. Rep. 2012, 14, 481–493.
    [CrossRef]
  94. Gou, S.-J.; Yuan, J.; Chen, M.; Yu, F.; Zhao, M.-H. Circulating complement activation in patients with anti-neutrophil cytoplasmic
    antibody–associated vasculitis. Kidney Int. 2013, 83, 129–137. [CrossRef]
  95. Huang, Y.-M.; Wang, H.; Wang, C.; Chen, M.; Zhao, M.-H. Promotion of Hypercoagulability in Antineutrophil Cytoplasmic
    Antibody-Associated Vasculitis by C5a-Induced Tissue Factor-Expressing Microparticles and Neutrophil Extracellular Traps.
    Arthritis Rheumatol. 2015, 67, 2780–2790. [CrossRef]
  96. Mendoza, C.E.; Brant, E.J.; McDermott, M.L.; Froment, A.; Hu, Y.; Hogan, S.L.; Jennette, J.C.; Falk, R.J.; Nachman, P.H.; Derebail,
    V.K.; et al. Elevated Microparticle Tissue Factor Activity Differentiates PatientsWith Venous Thromboembolism in Anti-neutrophil
    Cytoplasmic Autoantibody Vasculitis. Kidney Int. Rep. 2019, 4, 1617–1629. [CrossRef]
  97. Daniel, L.; Fakhouri, F.; Joly, D.; Mouthon, L.; Nusbaum, P.; Grunfeld, J.-P.; Schifferli, J.; Guillevin, L.; Lesavre, P.; Halbwachs-
    Mecarelli, L. Increase of circulating neutrophil and platelet microparticles during acute vasculitis and hemodialysis. Kidney Int.
    2006, 69, 1416–1423. [CrossRef] [PubMed]
  98. Brogan, P.A.; Shah, V.; Brachet, C.; Harnden, A.; Mant, D.; Klein, N.; Dillon, M.J. Endothelial and platelet microparticles in
    vasculitis of the young. Arthritis Rheum. 2004, 50, 927–936. [CrossRef] [PubMed]
  99. Kahn, R.; Mossberg, M.; Ståhl, A.-L.; Johansson, K.; Lindman, I.L.; Heijl, C.; Segelmark, M.; Mörgelin, M.; Leeb-Lundberg, L.F.;
    Karpman, D. Microvesicle transfer of kinin B1-receptors is a novel inflammatory mechanism in vasculitis. Kidney Int. 2017, 91,
    96–105. [CrossRef]
  100. Prikryl, P.; Satrapova, V.; Frydlova, J.; Hruskova, Z.; Zima, T.; Tesar, V.; Vokurka, M. Mass spectrometry-based proteomic
    exploration of the small urinary extracellular vesicles in ANCA-associated vasculitis in comparison with total urine. J. Proteom.
    2020, 233, 104067. [CrossRef]
    Int. J. Mol. Sci. 2021, 22, 4194 20 of 20
  101. Surmiak, M.; Gielicz, A.; Stojkov, D.; Szatanek, R.; Wawrzycka-Adamczyk, K.; Yousefi, S.; Simon, H.-U.; Sanak, M. LTB4 and
    5-oxo-ETE from extracellular vesicles stimulate neutrophils in granulomatosis with polyangiitis. J. Lipid Res. 2020, 61, 1–9.
    [CrossRef]
  102. Surmiak, M.; Kosalka-W˛egiel, J.; Pola ´ nski, S.; Sanak, M. Endothelial cells response to neutrophil-derived extracellular vesicles
    miRNAs in anti-PR3 positive vasculitis. Clin. Exp. Immunol. 2021, 1–16. [CrossRef]
  103. Wang, Y.; Bai, Y.; Liu, Y.; Noel, S.W.; Yan, Q.; Thi, H.P.; Sun, X.;Wei,W.; Ma, J.; Zheng, F. Plasma exosomal miRNAs involved in
    endothelial injury in microscopic polyangiitis patients. FASEB J. 2020, 34, 6215–6228. [CrossRef]
  104. Manojlovic, M.; Juto, A.; Jonasdottir, A.; Colic, J.; Vojinovic, J.; Nordin, A.; Bruchfeld, A.; Gunnarsson, I.; Mobarrez, F.; Antovic, A.
    Microparticles expressing myeloperoxidase as potential biomarkers in anti-neutrophil cytoplasmic antibody (ANCA)-associated
    vasculitides (AAV). J. Mol. Med. 2020, 98, 1279–1286. [CrossRef] [PubMed]
  105. Wang, C.; Chang, D.-Y.; Chen, M.; Zhao, M.-H. HMGB1 contributes to glomerular endothelial cell injury in ANCA-associated
    vasculitis through enhancing endothelium-neutrophil interactions. J. Cell. Mol. Med. 2017, 21, 1351–1360. [CrossRef] [PubMed]
  106. Antovic, A.; Mobarrez, F.; Manojlovic, M.; Soutari, N.; Baggemar, V.D.P.; Nordin, A.; Bruchfeld, A.; Vojinovic, J.; Gunnarsson, I.
    Microparticles Expressing Myeloperoxidase and Complement C3a and C5a as Markers of Renal Involvement in Antineutrophil
    Cytoplasmic Antibody–associated Vasculitis. J. Rheumatol. 2020, 47, 714–721. [CrossRef]
  107. Miao, D.; Ma, T.-T.; Chen, M.; Zhao, M.-H. Platelets release proinflammatory microparticles in anti-neutrophil cytoplasmic
    antibody-associated vasculitis. Rheumatology 2019, 58, 1432–1442. [CrossRef] [PubMed]