Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 -- 2744 2023-09-02 18:14:18 |
2 format Meta information modification 2744 2023-09-04 08:26:02 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Riitano, G.; Recalchi, S.; Capozzi, A.; Manganelli, V.; Misasi, R.; Garofalo, T.; Sorice, M.; Longo, A. Exosomes and Microvesicles in Rheumatoid Arthritis Pathogenesis. Encyclopedia. Available online: https://encyclopedia.pub/entry/48765 (accessed on 27 July 2024).
Riitano G, Recalchi S, Capozzi A, Manganelli V, Misasi R, Garofalo T, et al. Exosomes and Microvesicles in Rheumatoid Arthritis Pathogenesis. Encyclopedia. Available at: https://encyclopedia.pub/entry/48765. Accessed July 27, 2024.
Riitano, Gloria, Serena Recalchi, Antonella Capozzi, Valeria Manganelli, Roberta Misasi, Tina Garofalo, Maurizio Sorice, Agostina Longo. "Exosomes and Microvesicles in Rheumatoid Arthritis Pathogenesis" Encyclopedia, https://encyclopedia.pub/entry/48765 (accessed July 27, 2024).
Riitano, G., Recalchi, S., Capozzi, A., Manganelli, V., Misasi, R., Garofalo, T., Sorice, M., & Longo, A. (2023, September 02). Exosomes and Microvesicles in Rheumatoid Arthritis Pathogenesis. In Encyclopedia. https://encyclopedia.pub/entry/48765
Riitano, Gloria, et al. "Exosomes and Microvesicles in Rheumatoid Arthritis Pathogenesis." Encyclopedia. Web. 02 September, 2023.
Exosomes and Microvesicles in Rheumatoid Arthritis Pathogenesis
Edit

Rheumatoid arthritis (RA) is a chronic systemic autoimmune disease, characterized by persistent joint inflammation, leading to cartilage and bone destruction. Autoantibody production is directed to post-translational modified (PTM) proteins, i.e., citrullinated or carbamylated. Autophagy may be the common feature in several types of stress (smoking, joint injury, and infections) and may be involved in post-translational modifications (PTMs) in proteins and the generation of citrullinated and carbamylated peptides recognized by the immune system in RA patients, with a consequent breakage of tolerance. 

rheumatoid arthritis post-translational modifications extracellular vesicles

1. The Role of Autophagy on Post-Translational Modifications of Proteins in RA Patients

The autophagy process could be involved in post-translational changes of proteins and in the generation of citrullinated [1] and carbamylated [2] peptides recognized by the immune system in RA, with a consequent breakage of tolerance [3]. In this way, autophagy may represent a key processing event, creating a substrate for autoreactivity.
Autophagy is described as a regulated process inside almost every cell type activated against various stress conditions, such as starvation, protein aggregation, hypoxia, oxidative stress, and endoplasmic reticulum (ER) stress [4]. At the basal level, autophagy contributes to control biological process, the quality of proteins, and organelles, and eventually leads to a safe environment for cells [5]. Thus, damaged organelles, impaired and misfolded proteins, protein aggregates, and intracellular pathogens are encapsulated into autophagosomes and then fused with lysosomes for subsequent degradation [6]. Autophagy is a stress response that allows unicellular eukaryotic organisms to survive during harsh conditions, probably by regulating energy homeostasis and/or by protein quality control [5]. Alterations in autophagy machinery may be implicated in autoimmune diseases [7][8]. In particular, a significant difference in autophagic propensity between T lymphocytes from healthy donors and patients with SLE has been observed, demonstrating that lymphocytes from SLE patients are more resistant to autophagy induction [9]. Defective autophagy was also observed in the chondrocytes of patients with Kashin-Beck disease [10], in which ATG4C was identified as a susceptibility gene [11].
Moreover, RA synovium exhibits a highly increased ER stress-associated gene signature [12] and TNF-α further increases the expression of ER stress markers in fibroblast-like synoviocytes (FLSs) [13]. Kato et al. [14] identified a dual role for autophagy in the regulation of stress-induced cell death in RA FLSs. Interestingly, citrullination in the autophagosomes may increase the catabolism of the proteins, as charged residues of the proteins are eliminated. Thus, a key role for autophagy in the citrullination of peptides by antigen-presenting cells has been hypothesized [15][16][17].
As reported above, a number of environmental conditions, including smoking and infections, are associated with RA [17]. Autophagy may be the common feature by which, in several types of stress (including smoking, joint injury, and infections), autophagy vesicles of antigen-presenting cells may drive a response to citrullinated peptides recognized by the immune system [17].

2. The Interplay between Autophagy and Exosomes

Several studies have been undertaken to understand the crosstalk between endomembrane organelles and the molecular mechanisms involved in vesicular trafficking. Vesicular processes are highly dynamic and closely depending on subcellular compartments. Among them, the well-known vesicular processes are autophagy-related vesicles [18] and endosome-derived vesicles, i.e., exosomes [19]. The latter represent a class of nano-sized EVs, which derive from endosomal compartments and share several lines of linkages with endocytosis, lysosomal degradation, and autophagocytosis.
The capacity of EVs derived from the endosomal system to interact with the autophagic process has been extensively reported [20]. In this regard, biochemical studies support the evidence that autophagy shares with the molecular machinery of EVs, which include autophagy-related proteins and key proteins for EV biogenesis and secretion pathways [21][22]. In fact, unlike degradative autophagy, the autophagic machinery, including ATG factors, may lead to a form of unconventional secretion/expulsion of cytosolic proteins instead of their degradation; this mechanism appears to be of particular importance for protein secretion, immune surveillance, and cell signaling [23][24].
Thus, whether under physiological or pathological conditions, the crosstalk between exosome–autophagy networks ensure the cellular homeostasis via the lysosomal degradative pathway and/or the secretion of cargo into the extracellular space [25].
Exosomes, which are small EVs, have emerged as key players in the development and progression of RA-related joint inflammation. These unique EVs perform essential functions by facilitating the transportation of autoantigens and mediators between distant cells within affected joints [26].
Exosome biogenesis is a tightly regulated process. The molecular machinery includes four multiprotein complexes, known as the endosomal sorting complexes responsible for transport (ESCRT-0, -I, -II, and -III), in addition to auxiliary molecules. The cascade of interaction among ESCRT subunits and accessory molecules leads to the budding of vesicles into endosomes [27]. In mammalian cells, multivesicular body (MVB) generation is affected by autophagic machinery.
Recently, three different forms of autophagy have been extensively investigated: (i) macroautophagy refers to the formation of double membrane vesicles named autophagosomes, which enclose proteins and/or organelles, delivering them to the lysosome for degradation; (ii) microautophagy refers to the direct engulfment of cellular components to be degraded by lysosomes; (iii) chaperone-mediated autophagy (CMA) refers to the transport of target proteins to lysosomes in a lysosome-associated membrane protein-dependent manner. Macroautophagy is essential for the regulation of cellular function, organelle degradation, and adaptation to stress. The others are more directly involved in the fine regulation of cellular function.
To date, over 30 proteins encoded by specific Autophagy-related genes (Atg) are mandatory for macroautophagy, the most studied type of autophagy (hereafter referred to as autophagy). Among these, Autophagy-related-5 (ATG5), Autophagy-related-7 (ATG7), Autophagy-related-12 (ATG12), in association with Autophagy-related-16-like-1 (Atg16L1), participate in an enzymatic cascade that drives the nucleation, expansion, and closure of the phagophore in response to various stress stimuli. In particular, the macromolecular ATG12-ATG5-ATG16 complex, is responsible for the covalent modification of LC3-I (microtubule-associated protein 1 light chain 3, ATG8) with the amine part of phosphatidylethanolamine to form LC3-II, which is essential for autophagosome formation. Next, autophagosomes can fuse with lysosomes enabling their cargo to be degraded by acidic hydrolases. Alternatively, autophagosomes can also be fused with endosome-derived cell structures, such as MVBs. The key role of autophagy-related proteins, including ATG16L1 and ATG5, in exosome biogenesis in normal and pathological conditions has been well-determined [28]. For instance, ATG5 promotes the process leading to the fusion of MVBs with the plasma membrane in breast cancer cells; the inhibition of the ATG16L1 and ATG5-ATG12 complex markedly affects exosome biogenesis or their secretion, in addition to the sorting of LC3, a well-known autophagic marker, into exosomes [29]. Taken together, ATG5 and ATG16L1 protect MVBs from lysosomal degradation and direct them into the secretory pathway instead of the lysosomal pathway. Interestingly, the interaction of ATG12 with ATG3, which is responsible for LC3β conjugation, regulates exosome biogenesis through interaction with apoptosis-linked gene 2-interacting protein X (ALIX), a protein that cooperates with the ESCRT-III complex. Interestingly, the inhibition of ALIX decreases the autophagy flux, indicating a regulatory cross-link between exosome biogenesis and autophagy pathways [30].
In addition, ESCRT-independent machinery, including several lipids (i.e., ceramide), tetraspanins (CD9, CD63, and CD81), and other proteins, plays a pivotal role in the biogenesis of MVBs and exosome sorting [31].
As reported above, the exosome cargo may also contain molecules sorted from vesicles generated during the autophagic process named autophagosomes [32], though other intracellular vesicular systems, such as Golgi apparatus/vesicles, in the endocytosis pathway are not excluded. Crosslink between exosome biogenesis and autophagy pathways that engages the vesicular system has been supported by the biogenesis of hybrid vesicles inside cells referred to as amphisomes [33]. These vesicles are generated through the fusion of MVBs with autophagosomes, which finally combine with lysosomes for the hydrolysis and degradation of cargo; alternatively, they fuse with the plasma membrane for releasing intraluminal vesicles (ILVs) in extracellular space [34]. Based on this evidence, it can be assumed that MVBs represent transient structures, where cellular conditions affect their fate for degradation versus secretion. In addition, it is interesting to note that the fate of autophagosomes can also shift from a conventional degradation pathway to a secretory one depending on cellular conditions.

3. Role of Exosomes and Microvesicles in RA Pathogenesis

Numerous papers have demonstrated a role for exosomes in RA pathogenesis [35][36]. Exosomes are involved in intercellular communications, and some reports [37][38] have found upregulated proteins and RNAs inside them that contribute to the progression of RA. Some studies have shown that the total number of exosomes in both plasma and synovial fluid is increased in RA patients compared to healthy individuals [36].
Exosomes are present in the synovial fluid of inflamed joints, which originate from FLSs and cells infiltrated in the synovial joint, including platelets, granulocytes, monocytes, neutrophils, and T and B cells [39][40]. Exosomes from RA FLSs were shown to promote their abnormal proliferation and synovial hyperplasia [41]. These exosomes contain membrane-bound forms of TNF-α that in turn promote the activation of NF-kB and the induction of membrane-type matrix metalloproteinase (MMP)-1 in RA FLSs [42]. RA FLSs release transforming growth factor beta (TGF-β), enhancing RA FLS proliferation and angiogenesis. In addition, the increase of the 24- and the 17–18-kDa Toll-like receptor (TLR) 3 fragments has been observed in serum exosomes of RA patients, which may reflect the hyperactive state of RA [43]. The TLR3 signal activates NF-κB and Interferon Regulatory Factor (IRF) 3 transcription factors, which lead to the secretion of type I interferons and proinflammatory cytokines, such as IL-6 and IL-8 [44]. Interestingly, four microRNA (miRNAs), i.e., miR-155-5p, miR-146a-5p, miR-323a-5p, and mir-1307-3p, were upregulated upon TNF-α stimulation in the exosomes derived from FLSs, and different studies have shown the role of these miRNAs in the pathogenesis of RA [45]. Furthermore, post-translationally modified proteins, mainly citrullinated proteins, known as autoantigens in RA, were detected in exosomes purified from the synovial fluids of RA patients [46]. These citrullinated proteins, such as the Spα receptor, the fibrin α-chain fragment, the fibrin β-chain, the fibrinogen β-chain precursor, the fibrinogen D fragment, and vimentin enhance the production of pro-inflammatory cytokines and initiate pro-inflammatory responses characterized by Th1 and Th17 proliferations [46][47]. According to many reports [26][46][47][48], circulating exosomes have shown an ability to present citrullinated peptides to the effector cells in the form of an MHC-peptide complex. In contrast, some exosomes like those derived from mesenchymal stem cells (MSCs) can decrease joint destruction, suppressing FLS proliferation and promoting cartilage regeneration, such as exosomes containing miR-150-5p [49]. Furthermore, T cell-derived exosomes containing miR-204-5p could contribute to the inhibition of FLS proliferation [50]. Exosomes from different sources can affect RA progression by inducing the proliferation of CD4+ T cells and their differentiation towards Th17 cells, a pro-inflammatory cell population, in RA. For example, miR-424 in exosomes derived from RA FLSs significantly induced Th17 differentiation and inhibited Treg cell differentiation under hypoxic conditions [51]. On the other hand, miR-146a and miR-155 in MSC exosomes suppress T- and B-cell immune responses and increase Treg in vitro [52]. Moreover, exosomal miR-155 and miR-146a can be used for the early diagnosis of RA. Additionally, miRNA17 was upregulated in exosomes purified from RA patients’ plasma, which can suppress Treg induction by inhibiting the expression of transforming growth factor-beta receptor II (TGFBR II) in RA patients [52][53].
Another important factor in RA pathogenesis is bone resorption, and some exosomes may promote this process. Previous studies have shown that the levels of RANKL in exosomes isolated from the synovial fluid of patients with RA were significantly higher than those of patients with several other types of arthritis and induced higher numbers of osteoclasts involved in bone destruction [54]. In addition, exosomes from FLSs contain increased levels of miR-221-3p and mir-92a that can induce bone destruction in RA patients [55]. Correspondingly, the expression level of Hotair, a kind of long non-coding (Lnc) RNA leading to the migration of active macrophages, was greater in the exosome from RA patients. Hotair induces the release of MMP-2 and MMP-13 by osteoclasts and synoviocytes. Furthermore, Hotair is quite stable and easily detected in blood and urine and could be used as a diagnostic marker for RA [56]. In theory, the selective elimination of these exosomes would be beneficial to arthritis therapy.
Macrophages are relevant in the pathogenesis of RA and activated macrophages found in RA synovia are an early hallmark of RA. The effect of exosomes on macrophages in RA is still relatively limited. The secretion of miR-let-7b-containing exosomes promotes the differentiation of M1-type pro-inflammatory macrophages in RA joint inflammation [41]. Serum-derived exosomes miR-6089 and miR-548a-3p can regulate macrophage proliferation and differentiation [41][57].
These observations show the presence of disease-contributing exosomes, which could be useful inflammation markers of arthritis diseases. Furthermore, it has been shown that a subset of RA patients contains IgM RF associated with EVs, among which are exosomes, which can be used to distinguish between active and inactive RA [58].
Several proteomic studies have been performed to identify the exosomal components and their potential functions in the development of inflammatory arthritis. In RA, most of the exosomes are produced by the leukocytes and synoviocytes, and they are loaded with inflammatory molecules and enzymes that might be implicated in RA pathogenesis and the inflammatory process; therefore, they could be used as markers for RA subsets.
In addition to exosomes, MVs derived from cell plasma membrane may also play a role in the immunopathogenesis of RA [59]. The mechanisms by which MVs originate from plasma membrane are not fully known; external stimuli such as calcium ionophore, collagen, and epinephrine, as well as stress and mechanical factors, lead to the release of MVs. An influx of Ca2+ as an exogenous stimulus and the release of calcium from the endoplasmic reticulum leads to the activation of calpain, which plays an important role in the formation of MVs by participating in the reorganization of the cytoskeleton, which in turn participates in the shedding of MVs. The formation of MVs and their subsequent release is very often linked to the translocation of phosphatidylserine to the outer membrane of the cells. Moreover, the release of MVs can occur in specialized microdomains of the plasma membrane, i.e., lipid rafts, areas enriched in cholesterol and specialized in signal transduction, as they are rich in proteins involved in cell activation. MVs can transfer a series of information from one cell to another; they themselves can carry different molecules depending on the cell type from which they originate, influencing the functions of the cells they meet [60][61].
The presence of MVs has been demonstrated in biological fluids in both health and disease conditions. They can influence different functions in different diseases, contributing to their pathogenic mechanisms. Following cell activation or cell death (apoptosis, necroptosis, pyroptosis, and NETosis), large numbers of MVs are released into the blood, such as in the case of autoimmune diseases, including SLE and APS. Autoantigens generated during apoptosis are redistributed into the membrane surface of MVs or apoptotic bodies [62].
In rheumatic disorders, MVs isolated from synovial fluid have been shown to negatively impact osteoarthritis (OA) and RA disease progression [47]. Transmission electron microscopy observations demonstrated the occurrence of large multilamellar synovial MVs that are altered in synovial fluid from OA and patients with RA. There is also a difference in the biochemical properties of the synovial fluid of patients with OA and RA joints as compared to human samples collected from healthy volunteers [63]. In particular, the protein amount present in the synovial fluid is a greater than two-fold increase in OA samples and a greater than 2.5-fold increase in RA samples, as compared to healthy volunteers [63]. In addition, it has been shown that the number of MVs from the plasma of RA patients is significantly higher than in healthy donors [62].
A large presence of autoantigens typical of RA, including carbamylated or citrullinated proteins, are contained in the MVs released by patients, together with proinflammatory cytokines, which contribute to endothelial activation, such as adhesion molecules and chemokines. Moreover, it has been observed that MVs released by patients with RA promote an M1 macrophage profile, with a consequent amplification of the pro-inflammatory clinical picture [41]. Citrullinated neoepitopes have been described as key triggers of ACPA synthesis in patients with RA.
Platelets are the main source of MVs in blood, and their presence improves communication within the immune system; furthermore, platelets are involved in the crosstalk between the immune system and the coagulation system. In RA, platelets and platelet-derived MVs have been detected in both blood and synovial fluid samples [64].
Expression of both the enzyme PAD-4 and citrullinated proteins was demonstrated for the first time in human platelets and platelet-derived products (PDPs). In addition, ACPA-mediated platelet activation has been observed in RA patients. Both platelet aggregates and microparticles released as a consequence of platelet activation have been observed in joints in patients with RA [64].

References

  1. Sorice, M.; Iannuccelli, C.; Manganelli, V.; Capozzi, A.; Alessandri, C.; Lococo, E.; Garofalo, T.; Di Franco, M.; Bombardieri, M.; Nerviani, A.; et al. Autophagy generates citrullinated peptides in human synoviocytes: A possible trigger for anti-citrullinated peptide antibodies. Rheumatology 2016, 55, 1374–1385.
  2. Manganelli, V.; Recalchi, S.; Capozzi, A.; Riitano, G.; Mattei, V.; Longo, A.; Di Franco, M.; Alessandri, C.; Bombardieri, M.; Valesini, G.; et al. Autophagy induces protein carbamylation in fibroblast-like synoviocytes from patients with rheumatoid arthritis. Rheumatology 2018, 57, 2032–2041.
  3. Valesini, G.; Gerardi, M.C.; Iannuccelli, C.; Pacucci, V.A.; Pendolino, M.; Shoenfeld, Y. Citrullination and autoimmunity. Autoimmun. Rev. 2015, 14, 490–497.
  4. Ravanan, P.; Srikumar, I.F.; Talwar, P. Autophagy: The spotlight for cellular stress responses. Life Sci. 2017, 188, 53–67.
  5. Pohl, C.; Dikic, I. Cellular quality control by the ubiquitin-proteasome system and autophagy. Science 2019, 366, 818–822.
  6. Rashid, H.O.; Yadav, R.K.; Kim, H.R.; Chae, H.J. ER stress: Autophagy induction, inhibition and selection. Autophagy 2015, 11, 1956–1977.
  7. Pierdominici, M.; Vomero, M.; Barbati, C.; Colasanti, T.; Maselli, A.; Vacirca, D.; Giovannetti, A.; Malorni, W.; Ortona, E. Role of autophagy in immunity and autoimmunity, with a special focus on systemic lupus erythematosus. FASEB J. 2012, 26, 1400–1412.
  8. Ciccia, F.; Accardo-Palumbo, A.; Rizzo, A.; Guggino, G.; Raimondo, S.; Giardina, A.; Cannizzaro, A.; Colbert, R.A.; Alessandro, R.; Triolo, G. Evidence that autophagy, but not the unfolded protein response, regulates the expression of IL-23 in the gut of patients with ankylosing spondylitis and subclinical gut inflammation. Ann. Rheum. Dis. 2014, 73, 1566–1574.
  9. Alessandri, C.; Barbati, C.; Vacirca, D.; Piscopo, P.; Confaloni, A.; Sanchez, M.; Maselli, A.; Colasanti, T.; Conti, F.; Truglia, S.; et al. T lymphocytes from patients with systemic lupus erythematosus are resistant to induction of autophagy. FASEB J. 2012, 26, 4722–4732.
  10. Wu, C.; Zheng, J.; Yao, X.; Shan, H.; Li, Y.; Xu, P.; Guo, X. Defective autophagy in chondrocytes with Kashin-Beck disease but higher than osteoarthritis. Osteoarthr. Cartil. 2014, 22, 1936–1946.
  11. Wu, C.; Wen, Y.; Guo, X.; Yang, T.; Shen, H.; Chen, X.; Tian, Q.; Tan, L.; Deng, H.W.; Zhang, F. Genetic association, mRNA and protein expression analysis identify ATG4C as a susceptibility gene for Kashin-Beck disease. Osteoarthr. Cartil. 2017, 25, 281–286.
  12. Connor, A.M.; Mahomed, N.; Gandhi, R.; Keystone, E.C.; and Berger, S.A. TNFa modulates protein degradation pathways in rheumatoid arthritis synovial fibroblasts. Arthritis Res. Ther. 2012, 14, R62.
  13. Clausen, T.H.; Lamark, T.; Isakson, P.; Finley, K.; Larsen, K.B.; Brech, A.; Øvervatn, A.; Stenmark, H.; Bjørkøy, G.; Simonsen, A.; et al. p62/SQSTM1 and ALFY interact to facilitate the formation of p62 bodies/ ALIS and their degradation by autophagy. Autophagy 2010, 6, 330–344.
  14. Kato, M.; Ospelt, C.; Gay, R.E.; Gay, S.; Klein, K. Dual role of autophagy in stress-induced cell death in rheumatoid arthritis synovial fibroblasts. Arthritis Rheumatol. 2014, 66, 40–48.
  15. Ireland, J.M.; Unanue, E.R. Autophagy in antigen-presenting cells results in presentation of citrullinated peptides to CD4 T cells. J. Exp. Med. 2011, 208, 2625–2632.
  16. Suzuki, A.; Yamada, R.; Yamamoto, K. Citrullination by peptidylarginine deiminase in rheumatoid arthritis. Ann. N. Y. Acad. Sci. 2007, 1108, 323–339.
  17. Ireland, J.M.; Unanue, E.R. Processing of proteins in autophagy vesicles of antigen-presenting cells generates citrullinated peptides recognized by the immune system. Autophagy 2012, 8, 429–430.
  18. Matarrese, P.; Garofalo, T.; Manganelli, V.; Gambardella, L.; Marconi, M.; Grasso, M.; Tinari, A.; Misasi, R.; Malorni, W.; Sorice, M. Evidence for the involvement of GD3 ganglioside in the autophagosome formation and maturation. Autophagy 2014, 10, 750–765.
  19. Van Niel, G.; D'Angelo, G.; Raposo, G. Shedding light on the cell biology of extracellular vesicles. Nat. Rev. Mol. Cell. Biol. 2018, 19, 213–228.
  20. Gudbergsson, J.M.; Johnsen, K.B. Exosomes and autophagy: Rekindling the vesicular waste hypothesis. J. Cell Commun. Signal. 2019, 13, 443–450.
  21. Xu, J.; Camfield, R.; Gorski, S.M. The interplay between exosomes and autophagy—Partners in crime. J. Cell. Sci. 2018, 131, jcs215210.
  22. Salimi, L.; Akbari, A.; Jabbari, N.; Mojarad, B.; Vahhabi, A.; Szafert, S.; Kalashani, S.A.; Soraya, H.; Nawaz, M.; Rezaie, J. Synergies in exosomes and autophagy pathways for cellular homeostasis and metastasis of tumor cells. Cell. Biosci. 2020, 10, 64.
  23. Ponpuak, M.; Mandell, M.A.; Kimura, T.; Chauhan, S.; Cleyrat, C.; Deretic, V. Secretory autophagy. Curr. Opin. Cell Biol. 2015, 35, 106–116.
  24. Buratta, S.; Tancini, B.; Sagini, K.; Delo, F.; Chiaradia, E.; Urbanelli, L.; Emiliani, C. Lysosomal Exocytosis, Exosome Release and Secretory Autophagy: The Autophagic- and Endo-Lysosomal Systems Go Extracellular. Int. J. Mol. Sci. 2020, 21, 2576.
  25. Baixauli, F.; López-Otín, C.; Mittelbrunn, M. Exosomes and Autophagy: Coordinated Mechanisms for the Maintenance of Cellular Fitness. Front. Immunol. 2014, 5, 403.
  26. Alghamdi, M.; Alamry, S.A.; Bahlas, S.M.; Uversky, V.N.; Redwan, E.M. Circulating extracellular vesicles and rheumatoid arthritis: A proteomic analysis. Cell. Mol. Life Sci. 2021, 79, 25.
  27. Colombo, M.; Moita, C.; van Niel, G.; Kowal, J.; Vigneron, J.; Benaroch, P.; Manel, N.; Moita, L.F.; Théry, C.; Raposo, G. Analysis of ESCRT functions in exosome biogenesis, composition and secretion highlights the heterogeneity of extracellular vesicles. J. Cell Sci. 2013, 126, 5553–5565.
  28. Guo, H.; Sadoul, R.; Gibbings, D. Autophagy-independent effects of autophagy-related-5 (Atg5) on exosome production and metastasis. Mol. Cell. Oncol. 2018, 5, e1445941.
  29. Guo, H.; Chitiprolu, M.; Roncevic, L.; Javalet, C.; Hemming, F.J.; Trung, M.T.; Meng, L.; Latreille, E.; Tanese de Souza, C.; McCulloch, D.; et al. Atg5 Disassociates the V1V0-ATPase to Promote Exosome Production and Tumor Metastasis Independent of Canonical Macroautophagy. Dev. Cell. 2017, 43, 716–730.e7.
  30. Murrow, L.; Malhotra, R.; Debnath, J. ATG12–ATG3 interacts with Alix to promote basal autophagic flux and late endosome function. Nat. Cell Biol. 2015, 17, 300–310.
  31. Kowal, J.; Arras, G.; Colombo, M.; Jouve, M.; Morath, J.P.; Primdal-Bengtson, B.; Dingli, F.; Loew, D.; Tkach, M.; Théry, C. Proteomic comparison defines novel markers to characterize heterogeneous populations of extra- cellular vesicle subtypes. Proc. Natl. Acad. Sci. USA 2016, 113, E968–E977.
  32. Griffiths, R.E.; Kupzig, S.; Cogan, N.; Mankelow, T.J.; Betin, V.M.; Trakarnsanga, K.; Massey, E.J.; Lane, J.D.; Parsons, S.F.; Anstee, D.J. Maturing reticulocytes internalize plasma membrane in glycophorin A-containing vesicles that fuse with autophagosomes before exocytosis. Blood 2012, 119, 6296–6306.
  33. Klionsky, D.J.; Eskelinen, E.L.; Deretic, V. Autophagosomes, phagosomes, autolysosomes, phagolysosomes, autophagolysosomes... wait, I’m confused. Autophagy 2014, 10, 549–551.
  34. Sanchez-Wandelmer, J.; Reggiori, F. Amphisomes: Out of the autophagosome shadow? EMBO J. 2013, 32, 3116–3118.
  35. Withrow, J.; Murphy, C.; Liu, Y.; Hunter, M.; Fulzele, S.; Hamrick, M.W. Extracellular vesicles in the pathogenesis of rheumatoid arthritis and osteoarthritis. Arthritis Res. Ther. 2016, 18, 286.
  36. Schioppo, T.; Ubiali, T.; Ingegnoli, F.; Bollati, V.; Caporali, R. The role of extracellular vesicles in rheumatoid arthritis: A systematic review. Clin. Rheumatol. 2021, 40, 3481–3497.
  37. Lee, E.Y.; Bang, J.Y.; Park, G.W.; Choi, D.S.; Kang, J.S.; Kim, H.J.; Park, K.S.; Lee, J.O.; Kim, Y.K.; Kwon, K.H.; et al. Global proteomic profiling of native outer membrane vesicles derived from Escherichia coli. Proteomics 2007, 7, 3143–3153.
  38. Kaparakis-Liaskos, M.; Ferrero, R.L. Immune modulation by bacterial outer membrane vesicles. Nat. Rev. Immunol. 2005, 15, 375–387.
  39. Bartok, B.; Firestein, G.S. Fibroblast-like synoviocytes: Key effector cells in rheumatoid arthritis. Immunol. Rev. 2010, 233, 233–255.
  40. Krajewska-Włodarczyk, M.; Owczarczyk-Saczonek, A.; Żuber, Z.; Wojtkiewicz, M.; Wojtkiewicz, J. Role of microparticles in the pathogenesis of inflammatory joint diseases. Int. J. Mol. Sci. 2019, 20, 5453.
  41. Zhao, J.; Zhang, B.; Meng, W.; Hu, J. Elucidating a fresh perspective on the interplay between exosomes and rheumatoid arthritis. Front. Cell. Dev. Biol. 2023, 11, 1177303.
  42. Zhang, B.; Zhao, M.; Lu, Q. Extracellular vesicles in rheumatoid arthritis and systemic lupus erythematosus: Functions and applications. Front. Immunol. 2021, 11, 575712.
  43. Tsuno, H.; Arito, M.; Suematsu, N.; Sato, T.; Hashimoto, A.; Matsui, T.; Omoteyama, K.; Sato, M.; Okamoto, K.; Tohma, S.; et al. A proteomic analysis of serum-derived exosomes in rheumatoid arthritis. BMC Rheumatol. 2018, 2, 35.
  44. Chen, Y.; Lin, J.; Zhao, Y.; Ma, X.; Yi, H. Toll-like receptor 3 (TLR3) regulation mechanisms and roles in antiviral innate immune responses. J. Zhejiang Univ. Sci. B 2021, 22, 609–632.
  45. Takamura, Y.; Aoki, W.; Satomura, A.; Shibasaki, S.; Ueda, M. Small RNAs detected in exosomes derived from the MH7A synovial fibroblast cell line with TNF-α stimulation. PLoS ONE 2018, 13, e0201851.
  46. Skriner, K.; Adolph, K.; Jungblut, P.R.; Burmester, G.R. Association of citrullinated proteins with synovial exosomes. Arthritis Rheum. 2006, 54, 3809–3814.
  47. Heydari, R.; Koohi, F.; Rasouli, M.; Rezaei, K.; Abbasgholinejad, E.; Bekeschus, S.; Doroudian, M. Exosomes as rheumatoid arthritis diagnostic biomarkers and therapeutic agents. Vaccines 2023, 11, 687.
  48. Shenoda, B.B.; Ajit, S.K. Modulation of Immune Responses by Exosomes Derived from Antigen-Presenting Cells. Clin. Med. Insights Pathol. 2016, 9, 1–8.
  49. Chen, Z.; Wang, H.; Xia, Y.; Yan, F.; Lu, Y. Therapeutic potential of mesenchymal cell-derived miRNA-150-5p-expressing exosomes in rheumatoid arthritis mediated by the modulation of MMP14 and VEGF. J. Immunol. 2018, 201, 2472–2482.
  50. Wu, L.F.; Zhang, Q.; Mo, X.B.; Lin, J.; Wu, Y.L.; Lu, X.; He, P.; Wu, J.; Guo, Y.F.; Wang, M.J.; et al. Identification of novel rheumatoid arthritis-associated MiRNA-204-5p from plasma exosomes. Exp. Mol. Med. 2022, 54, 334–345.
  51. Ding, Y.; Wang, L.; Wu, H.; Zhao, Q.; Wu, S. Exosomes derived from synovial fibroblasts under hypoxia aggravate rheumatoid arthritis by regulating Treg/Th17 balance. Exp. Biol. Med. 2020, 245, 1177–1186.
  52. Peng, X.; Wang, Q.; Li, W.; Ge, G.; Peng, J.; Xu, Y.; Yang, H.; Bai, J.; Geng, D. Comprehensive overview of microRNA function in rheumatoid arthritis. Bone Res. 2023, 11, 8.
  53. Tavasolian, F.; Hosseini, A.Z.; Soudi, S.; Naderi, M. miRNA-146a improves immunomodulatory effects of msc-derived exosomes in rheumatoid arthritis. Curr. Gene Ther. 2020, 20, 297–312.
  54. Song, J.E.; Kim, J.S.; Shin, J.H.; Moon, K.W.; Park, J.K.; Park, K.S.; Lee, E.Y. Role of synovial exosomes in osteoclast differentiation in inflammatory arthritis. Cells 2021, 10, 120.
  55. Mao, G.; Zhang, Z.; Hu, S.; Zhang, Z.; Chang, Z.; Huang, Z.; Liao, W.; Kang, Y. Exosomes derived from miR-92a-3p-overexpressing human mesenchymal stem cells enhance chondrogenesis and suppress cartilage degradation via targeting WNT5A. Stem Cell Res. Ther. 2018, 9, 247.
  56. Tsai, C.Y.; Hsieh, S.C.; Liu, C.W.; Lu, C.H.; Liao, H.T.; Chen, M.H.; Li, K.J.; Wu, C.H.; Shen, C.Y.; Kuo, Y.M.; et al. The expression of non-coding rnas and their target molecules in rheumatoid arthritis: A molecular basis for rheumatoid pathogenesis and its potential clinical applications. Int. J. Mol. Sci. 2021, 22, 5689.
  57. Xu, D.; Song, M.; Chai, C.; Wang, J.; Jin, C.; Wang, X.; Cheng, M.; Yan, S. Exosome-encapsulated miR-6089 regulates inflammatory response via targeting TLR4. J. Cell. Physiol. 2019, 234, 1502–1511.
  58. Arntz, O.J.; Pieters, B.C.H.; Thurlings, R.M.; Wenink, M.H.; van Lent, P.L.E.M.; Koenders, M.I.; van den Hoogen, F.H.J.; van der Kraan, P.M.; van de Loo, F.A.J. Rheumatoid arthritis patients with circulating extracellular vesicles positive for IgM rheumatoid factor have higher disease activity. Front. Immunol. 2018, 9, 2388.
  59. Barbati, C.; Vomero, M.; Colasanti, T.; Ceccarelli, F.; Marcosano, M.; Miranda, F.; Novelli, L.; Pecani, A.; Perricone, C.; Spinelli, F.R.; et al. Microparticles and autophagy: A new frontier in the understanding of atherosclerosis in rheumatoid arthritis. Immunol. Res. 2018, 66, 655–662.
  60. Sapoń, K.; Mańka, R.; Janas, T.; Janas, T. The role of lipid rafts in vesicle formation. J. Cell Sci. 2023, 136, 260887.
  61. Barbat, C.; Trucy, M.; Sorice, M.; Garofalo, T.; Manganelli, V.; Fischer, A.; Mazerolles, F. p56lck, LFA-1 and PI3K but not SHP-2 interact with GM1- or GM3-enriched microdomains in a CD4-p56lck association-dependent manner. Biochem. J. 2007, 402, 471–481.
  62. Ucci, F.M.; Recalchi, S.; Barbati, C.; Manganelli, V.; Capozzi, A.; Riitano, G.; Buoncuore, G.; Garofalo, T.; Ceccarelli, F.; Spinelli, F.M.; et al. Citrullinated and carbamylated proteins in extracellular microvesicles from plasma of patients with rheumatoid arthritis. Rheumatology 2023, 62, 2312–2319.
  63. Ben-Trad, L.; Matei, C.I.; Sava, M.M.; Filali, S.; Duclos, M.E.; Berthier, Y.; Guichardant, M.; Bernoud-Hubac, N.; Maniti, O.; Landoulsi, A.; et al. Synovial extracellular vesicles: Structure and role in synovial fluid tribological performances. Int. J. Mol. Sci. 2022, 23, 11998.
  64. Xu, M.; Du, R.; Xing, W.; Chen, X.; Wan, J.; Wang, S.; Xiong, L.; Nandakumar, K.S.; Holmdahl, R.; Geng, H. Platelets derived citrullinated proteins and microparticles are potential autoantibodies ACPA targets in RA patients. Front. Immunol. 2023, 14, 1084283.
More
Information
Subjects: Pathology
Contributors MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register : , , , , , , ,
View Times: 168
Revisions: 2 times (View History)
Update Date: 04 Sep 2023
1000/1000
Video Production Service