1,412
Views
0
CrossRef citations to date
0
Altmetric
Editorial

Microparticles - culprits in the pathogenesis of systemic lupus erythematosus?

&
Pages 443-445 | Received 14 Feb 2018, Accepted 04 May 2018, Published online: 11 May 2018

Membrane-derived extracellular vesicles include exosomes, microvesicles, and apoptotic bodies. The two latter vesicle subsets are produced during cell activation and apoptosis, respectively. Microvesicles are typically defined by sizes within the range of 0.1–1 µm in diameter [Citation1,Citation2]. Apoptotic bodies may be included in the microvesicle size range but do also exceed this depending on stage of apoptosis [Citation3]. Despite differences in genesis and size, microvesicles and apoptotic bodies are often collectively referred to as microparticles (MPs). First described as ‘platelet dust’ by Peter Wolf in 1967, these tiny circulators were initially dismissed as inert cell artifacts. However, in recent years, such outlook has subsided in favor of evidence of great diversity, enrolling MPs in many physiological processes, including coagulation, inflammation, and intercellular communication [Citation4,Citation5]. This recognition has doubtlessly nurtured the growing interest of MPs as potential pathogenic players not least in the context of the systemic autoimmune disease, systemic lupus erythematosus (SLE). SLE is characterized by hyperproduction of autoantibodies against especially nuclear antigens and immune complex (IC) deposition in various parts of the body, leading to systemic inflammation and tissue damage. Furthermore, increased type I interferon (IFN) activity is frequent and suspected as a major disease contributor [Citation6,Citation7]. With defective removal of apoptotic material being recognized as a central pathoetiologic element of SLE [Citation6], the suspicion of apoptotically derived MPs to have a role in this matter is not unforeseen.

Apoptotic MPs that derive from nucleated cells may contain chromatin including dsDNA and RNA [Citation8]. It is, however, also of great interest that MPs derived from activated platelets may contain mitochondrial DNA [Citation9] and partake in the formation of MP-ICs [Citation10]. Further, oxidized mitochondrial nucleoids released by neutrophils have been shown to activate type I IFN production in SLE [Citation11]. It could even be speculated that apoptotically derived chromatin in SLE patients may have increased immunogenic potential due to posttranslational modifications occurring during disturbed apoptosis or delayed clearing. In this respect, it is of particular interest that DNase 1L3 seems to restrict the immunogenicity of apoptotic cell-derived MPs in DNASE1L3-deficient mice [Citation12]. SLE-MPs are also characterized by the carriage of immunoglobulins (Igs) [Citation13,Citation14] that in part bind to dsDNA [Citation15]. MP-ICs in SLE could thus be regarded as large ICs with capacity for tissue deposition as well as sources of complexed nucleic acids, which may stimulate type I IFN production by interaction with internal nucleic acid sensors [Citation7]. Clinically, MP-ICs have been associated with disease activity and vascular damage in SLE patients [Citation10,Citation14].

An obvious question to address is whether the MP-profile in SLE patients differs from that of healthy individuals. Several studies have quantitated MPs in plasma but with conflicting results. Whereas some have reported increased occurrence of MPs among SLE patients [Citation16,Citation17], others found it to be similar or even decreased relative to healthy subjects [Citation5,Citation18,Citation19]. These variations may reflect a high degree of dynamism and/or pleiotropic effects of individual disease courses but also strongly depend on pre-analytical parameters and analytical methodologies. Particularly in SLE, the formation of ICs may perturb analysis of MPs due to overlapping biophysical properties [Citation20]. Not only quantitative aberrations are of interest in disclosing potential pathogenic roles of MPs in SLE but also qualitative changes. The increased proportion of MPs not binding phosphatidylserine-binding probes such as annexin V and lactadherin [Citation17] could be explained by reduced clearing of such MPs as externalized phosphatidylserine and the phagocytic phosphatidylserine receptor mediate effective phagocytosis of apoptotic cells by macrophages [Citation21]. However, the safe clearance of apoptotic cells and remnants also implicates a wider range of scavenger receptors and serum opsonins and the redundancy in this system indicates its vital significance. Besides the rare state of C1q deficiency and lupus-like disease [Citation22], the finding of reduced amounts of C3b/iC3b on SLE-MPs molecules supports the notion of diminished opsonization of MPs in SLE patients [Citation19]. Albeit this would at least partly explain an increased load of peripheral MPs in SLE, the investigation did not reveal any quantitative differences in such regard. This could be explained by an increased tissue deposition of MPs in SLE and one may speculate if numeric aberrations are mainly visible in specific subsets of MPs rather than the collective.

Proteomic studies show that plasma MPs from SLE patients hold a characteristic signature that deviates markedly from normal [Citation23,Citation24]. These MPs tentatively designated ‘luposomes’ are characterized by lack of or reduced mitochondrial function, indications of disrupted cytoskeletons, and/or an excess of glycolytic proteins, immunoglobulins, ficolin 2, and galectin-3 binding protein (G3BP). Complementary to these findings are flow cytometric analyses, in which circulating MPs with surface-bound Ig [Citation25] or G3BP [Citation18] were quantitated and found to be highly increased among SLE patients relative to healthy individuals. G3BP is a glycoprotein with properties that may have bearings on the pathogenesis of SLE. It belongs to the scavenger receptor cysteine-rich domain family and is widely expressed in extracellular fluids [Citation26]. Furthermore, it is type I IFN-inducible [Citation18] and exhibits a high binding capacity toward components of the glomerular basement membrane (GBM) including collagen IV and nidogen [Citation27]. Considering the association between G3BP and SLE, the mentioned qualities of G3BP permit an intriguing scenario. Do G3BP-positive MPs have a strong affinity for the GBM? And if so, do these particles possess a particular pathogenic potential? Although apoptotic MPs are known to carry DNA/chromatin that is accessible to autoantibodies [Citation8,Citation15], the antigenic availability of activation-derived MPs is less explored. However, a study on murine macrophages showed that MPs released from cells stimulated with Toll-like receptor (TLR)-3 and TLR-4 ligands possessed several similarities with apoptotic MPs, including binding of anti-DNA and anti-nucleosome antibodies [Citation28]. Since G3BP is a type 1 IFN-inducible protein, MPs deriving from type 1 IFN activated cells may have the potential of expressing G3BP. To this end, it is of interest that lupus nephritis (LN) patients exhibit a glomerular G3BP/IgG colocalization pattern specifically in the GBM – a pattern that seems to be absent in non-inflammatory kidney tissue [Citation18]. This finding is somewhat supportive of two circumstances, namely the presence of G3BP-positive MPs in the glomeruli of LN patients and the containment of ICs on these particles. Hence, MPs rich in G3BP might act as IC-carrying vehicles that mediate the deposition of ICs in the kidneys of SLE patients [Citation29]. Yet, the source of such ‘luposomes’ remains to be identified.

MPs may have other sinister roles in SLE besides being perceived as mere ‘passive’ IC carriers. Dieker et al. demonstrated that MPs from SLE patients have strong proinflammatory effects on pDCs and myeloid dendritic cells [Citation5]. Moreover, a comparable response has been observed on granulocytes where SLE MPs had the ability to initiate degranulation and production of reactive oxygen species [Citation30]. The formation of neutrophil extracellular traps is considered to partake in the pathogenesis of SLE and lupus nephritis [Citation31], and this phenomenon has been shown to be triggered by SLE-MPs independently of the formation of reactive oxygen species [Citation32]. It was even found that this mechanism was dependent on the acetylation levels of histones in the SLE MPs, which brings us back to the notion of apoptosis-related modifications of MP constituents.

Besides potential effector roles of MPs in SLE, it seems plausible that some MPs are immunogenic while others are IC formators or both. Further, MPs may also carry significant information regarding the apoptotic process of the parental cells, their potential for clearance, their immunogenic potential, and they may even reflect the metabolic state of their parental cells [Citation24]. This may hopefully leave us with many clues to help us solve the complex mystery of SLE pathogenesis in which there certainly may be several culprits. Based on current evidence, future studies will hopefully establish if and to what extent MPs are one of them.

Declaration of interest

The authors have no relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript. This includes employment, consultancies, honoraria, stock ownership or options, expert testimony, grants or patents received or pending, or royalties. Peer reviewers on this manuscript have no relevant financial or other relationships to disclose.

Additional information

Funding

This work has been in part been funded by The Danish Rheumatism Association (A3865) and the Lundbeck Foundation (R208-2015-4018).

References

  • Distler JH, Pisetsky DS, Huber LC, et al. Microparticles as regulators of inflammation: novel players of cellular crosstalk in the rheumatic diseases. Arthritis Rheum. 2005;52:3337–3348.
  • Arraud N, Linares R, Tan S, et al. Extracellular vesicles from blood plasma: determination of their morphology, size, phenotype and concentration. J Thromb Haemost. 2014;12:614–627.
  • Zirngibl M, Furnrohr BG, Janko C, et al. Loading of nuclear autoantigens prototypically recognized by systemic lupus erythematosus sera into late apoptotic vesicles requires intact microtubules and myosin light chain kinase activity. Clin Exp Immunol. 2015;179:39–49.
  • Hargett LA, Bauer NN. On the origin of microparticles: from “platelet dust” to mediators of intercellular communication. Pulm Circ. 2013;3:329–340.
  • Dieker J, Tel J, Pieterse E, 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.
  • Kaul A, Gordon C, Crow MK, et al. Systemic lupus erythematosus. Nat Rev Dis Primers. 2016;2:16039.
  • Eloranta ML, Ronnblom L. Cause and consequences of the activated type I interferon system in SLE. J Mol Med (Berl). 2016;94:1103–1110.
  • Reich CF 3rd, Pisetsky DS. The content of DNA and RNA in microparticles released by Jurkat and HL-60 cells undergoing in vitro apoptosis. Exp Cell Res. 2009;315:760–768.
  • Boudreau LH, Duchez AC, Cloutier N, et al. Platelets release mitochondria serving as substrate for bactericidal group IIA-secreted phospholipase A2 to promote inflammation. Blood. 2014;124:2173–2183.
  • Burbano C, Villar-Vesga J, Orejuela J, et al. 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.
  • Caielli S, Athale S, Domic B, et al. Oxidized mitochondrial nucleoids released by neutrophils drive type I interferon production in human lupus. J Exp Med. 2016;213:697–713.
  • Sisirak V, Sally B, D’Agati V, et al. Digestion of chromatin in apoptotic cell microparticles prevents autoimmunity. Cell. 2016;166:88–101.
  • Nielsen CT. Circulating microparticles in systemic lupus erythematosus. Dan Med J. 2012;59:B4548.
  • Fortin PR, Cloutier N, Bissonnette V, et al. Distinct subtypes of microparticle-containing immune complexes are associated with disease activity, damage, and carotid intima-media thickness in systemic lupus erythematosus. J Rheumatol. 2016;43:2019–2025.
  • Ullal AJ, Marion TN, Pisetsky DS. The role of antigen specificity in the binding of murine monoclonal anti-DNA antibodies to microparticles from apoptotic cells. Clin Immunol. 2014;154:178–187.
  • Pereira J, Alfaro G, Goycoolea M, et al. Circulating platelet-derived microparticles in systemic lupus erythematosus. Association with increased thrombin generation and procoagulant state. Thromb Haemost. 2006;95:94–99.
  • Mobarrez F, Vikerfors A, Gustafsson JT, et al. Microparticles in the blood of patients with systemic lupus erythematosus (SLE): phenotypic characterization and clinical associations. Sci Rep. 2016;6:36025.
  • Nielsen CT, Ostergaard O, Rekvig OP, et al. Galectin-3 binding protein links circulating microparticles with electron dense glomerular deposits in lupus nephritis. Lupus. 2015;24:1150–1160.
  • Winberg LK, Nielsen CH, Jacobsen S. Surface complement C3 fragments and cellular binding of microparticles in patients with SLE. Lupus Sci Med. 2017;4:e000193.
  • Gyorgy B, Modos K, Pallinger E, et al. Detection and isolation of cell-derived microparticles are compromised by protein complexes resulting from shared biophysical parameters. Blood. 2011;117:e39–e48.
  • Li MO, Sarkisian MR, Mehal WZ, et al. Phosphatidylserine receptor is required for clearance of apoptotic cells. Science. 2003;302:1560–1563.
  • Stegert M, Bock M, Trendelenburg M. Clinical presentation of human C1q deficiency: how much of a lupus? Mol Immunol. 2015;67:3–11.
  • Ostergaard O, Nielsen CT, Iversen LV, et al. Unique protein signature of circulating microparticles in systemic lupus erythematosus. Arthritis Rheum. 2013;65:2680–2690.
  • Ostergaard O, Nielsen CT, Tanassi JT, et al. Distinct proteome pathology of circulating microparticles in systemic lupus erythematosus. Clin Proteomics. 2017;14:23.
  • Nielsen CT, Ostergaard O, Stener L, et al. Increased IgG on cell-derived plasma microparticles in systemic lupus erythematosus is associated with autoantibodies and complement activation. Arthritis Rheum. 2012;64:1227–1236.
  • White MJ, Roife D, Gomer RH. Galectin-3 binding protein secreted by breast cancer cells inhibits monocyte-derived fibrocyte differentiation. J Immunol. 2015;195:1858–1867.
  • Sasaki T, Brakebusch C, Engel J, et al. 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.
  • Spencer DM, Gauley J, Pisetsky DS. The properties of microparticles from RAW 264.7 macrophage cells undergoing in vitro activation or apoptosis. Innate Immun. 2014;20:239–248.
  • Nielsen CT, Ostergaard O, Rasmussen NS, et al. A review of studies of the proteomes of circulating microparticles: key roles for galectin-3-binding protein-expressing microparticles in vascular diseases and systemic lupus erythematosus. Clin Proteomics. 2017;14:11.
  • Winberg LK, Jacobsen S, Nielsen CH. Microparticles from patients with systemic lupus erythematosus induce production of reactive oxygen species and degranulation of polymorphonuclear leukocytes. Arthritis Res Ther. 2017;19:230.
  • Hakkim A, Furnrohr BG, Amann K, et al. Impairment of neutrophil extracellular trap degradation is associated with lupus nephritis. Proc Natl Acad Sci U S A. 2010;107:9813–9818.
  • Rother N, Pieterse E, Lubbers J, et al. Acetylated histones in apoptotic microparticles drive the formation of neutrophil extracellular traps in active lupus nephritis. Front Immunol. 2017;8:1136.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.