Next Article in Journal
p53 Affects Zeb1 Interactome of Breast Cancer Stem Cells
Next Article in Special Issue
The Pannexin 1 Channel and the P2X7 Receptor Are in Complex Interplay to Regulate the Release of Soluble Ectonucleotidases in the Murine Bladder Lamina Propria
Previous Article in Journal
Advanced Glycation End Products as a Potential Target for Restructuring the Ovarian Cancer Microenvironment: A Pilot Study
Previous Article in Special Issue
The Coming of Age of the P2X7 Receptor in Diagnostic Medicine
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

P2X7 Receptor and Extracellular Vesicle Release

by
Maria Teresa Golia
,
Martina Gabrielli
and
Claudia Verderio
*
National Research Council of Italy, Institute of Neuroscience, Via Raoul Follereau 3, 20854 Vedano al Lambro, Italy
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2023, 24(12), 9805; https://doi.org/10.3390/ijms24129805
Submission received: 29 April 2023 / Revised: 21 May 2023 / Accepted: 1 June 2023 / Published: 6 June 2023
(This article belongs to the Special Issue The Role of P2X7 Receptor in Human Health and Diseases)

Abstract

:
Extensive evidence indicates that the activation of the P2X7 receptor (P2X7R), an ATP-gated ion channel highly expressed in immune and brain cells, is strictly associated with the release of extracellular vesicles. Through this process, P2X7R-expressing cells regulate non-classical protein secretion and transfer bioactive components to other cells, including misfolded proteins, participating in inflammatory and neurodegenerative diseases. In this review, we summarize and discuss the studies addressing the impact of P2X7R activation on extracellular vesicle release and their activities.

1. Introduction

P2X7 receptor (P2X7R) is an ATP-gated ion channel belonging to the purinergic P2X family. It is highly expressed by cells of the innate immune system, especially macrophages [1], dendritic cells [2], mast cells [3], and microglia [4], where it promotes inflammasome formation and the release of inflammatory cytokines [5,6,7]. P2X7R is also present in adaptive immune cells (T cells), where it regulates cell development and function [8], and in many other cell types [9] including nervous system cells [10,11], epithelial and endothelial cells [12,13], bone cells [14], fibroblasts [15], and smooth muscle cells [16], as well as in tumor cells, where its expression often correlates with a worse diagnosis [17,18].
Among the P2XR family, P2X7R exhibits peculiar features including a low affinity for ATP, a lack of desensitization and unique structural domains, i.e., a “C-cysteine anchor” intra-cytoplasmic motif and a long C-terminal cytoplasmic domain that contains several protein–protein interaction motifs [19,20]. The receptor also exhibits a characteristic dual gating state depending on extracellular ATP (eATP) concentration. At micromolar eATP concentration, P2X7R opens a cation-selective channel that mediates the cellular influx of Na+ and Ca2+ ions and an efflux of K+ [21]; at higher eATP concentrations (above 100 mM) and upon prolonged exposure, the receptor functions as a non-selective membrane pore permeable to hydrophilic molecules [21], generally leading to cytotoxicity and apoptotic cell death [22]. Ion alterations also induce the opening of the pannexin-1 channel, which, by releasing ATP into the extracellular space, perpetuates P2X7R stimulation [23]. Therefore, whether pannexin-1, as a P2X7R-associated protein, forms the large pore itself or mediates its formation is still a matter of debate.
Channel opening increases cell proliferation and survival [24,25] whereas large pore opening induces the activation of inflammasome (as reviewed in [26]), a cytoplasmic multiprotein complex that, in response to pathogens/cell damage, triggers cytokine release and pyroptosis, a lytic form of programmed cell death [27].
The inflammasome consists of a sensor protein (e.g., NLR family CARD domain containing 4 (NLRC4), NLR Family Pyrin Domain Containing 1 (NLRP1), NLR Family Pyrin Domain Containing 3 (NLRP3) that is activated by ATP, absent-in-melanoma 2 (AIM2), and pyrin), an inflammatory caspase, and in some cases an adaptor protein, such as ASC (apoptosis-associated speck-like protein containing a CARD) [28]. Once assembled and activated in response to ATP, the NLRP3 inflammasome triggers pro-caspase-1 cleavage, which generates active caspase-1 that, in turn, drives the enzymatic activation of the leaderless cytokines Interleukin (IL)-1β and IL-18, initiating an inflammatory response [29,30].
In addition to ATP, the P2X7R cation channel can be opened by non-ATP nucleotides, such as NAD+ (nicotinamide adenine dinucleotide). This ATP-independent pathway consists of receptor ADP-ribosylation by ADP-ribosyltransferases ART2.1 and ART2.2, which catalyze the transfer of ribose from NAD+ to arginine 125 in the ectodomain of the P2X7R close to the ATP binding site [31]. The P2X7R opening by ADP-ribosylation enables Ca2+ and Na+ influx and K+ efflux, phosphatidylserine externalization, membrane pore formation, mitochondrial membrane breakdown, and ultimately cell death [32,33,34].
Interestingly, both ATP and NAD+ concentrations are low (in the submicromolar range) in the extracellular space, due to the activities of the ectoenzymes CD39 and CD38 that degrade them, respectively [8,35]. Therefore, P2X7R activation occurs at inflammatory or damaged sites, as well as in the tumor microenvironment, where ATP and NAD+ are released in substantial amounts [36,37]. Accordingly, P2X7R-mediated signaling is activated in a large variety of Central Nervous System (CNS) disorders (i.e., Alzheimer’s, Parkinson’s and Huntington’s disease, multiple sclerosis, Amyotrophic Lateral Sclerosis, stroke, neurotrauma, neuropathic pain, epilepsy, and neuropsychiatric disorders), and P2X7R antagonists are under intense investigation as a therapy for these conditions (as reviewed in [38]).
One of the main consequences of P2X7R activation is the formation of blebs at the cell surface and the release of extracellular vesicle (EVs) into the microenvironment. EVs are a heterogeneous group of cell-derived membranous structures which directly bud from the plasma membrane (microvesicles) or originate in the endocytic compartment as intraluminal vesicles (ILVs) which are released through the fusion of multivesicular bodies (MVBs) to the plasma membrane (exosomes) [39]. Due to technical limitations in isolating and distinguishing EVs based on their biogenesis, the currently recognized nomenclature identifies EVs according to their physical properties and dimensions, distinguishing medium-large/large EVs (>200 nm) and small EVs (<200 nm) [40]. Accordingly, here we use the terms large and small EVs to refer to the two main populations of EVs. EVs act as carriers of bioactive molecules (proteins, lipids, genetic materials, and metabolites) and convey their bioactive cargoes between cells, playing a fundamental role in cell-to-cell communication in both physiological conditions and during inflammatory and degenerative diseases [41,42].
In the present review, we will first discuss the impact of P2X7R activation on EV release from the cell surface and the endocytic compartment. Then, we will summarize the current knowledge about the role of P2X7R activation in the sorting of proteins into EVs, the secretion of inflammatory cytokines, and the dissemination of misfolded proteins.

2. P2X7R Activation and EV Release

Among stimuli that promote EV release (cytokines, LPS, capsaicin, serotonin, Wnt3a, and a-synuclein) [43], eATP is the classical trigger that, through P2X7R activation, massively increases the shedding of EVs from the plasma membrane of immune cells including dendritic cells [44,45], microglia [46], and macrophages [6,47]. Of note, not only millimolar concentration of eATP but also ATP endogenously released by astrocytes could induce P2X7R-dependent EV release in microglia-astrocyte co-cultures [46].
The first evidence implicating P2X7R activation in the release of EVs dates back to 2001, when MacKenzie and colleagues showed that within the first few minutes of P2X7R activation, bleb formation occurs at the surface of monocytes and large EVs with externalized phosphatidylserine (PS) are released into the extracellular space as a result of bleb detachment from the membrane [48]. Notably, bleb formation and the externalization of PS, a typical marker of apoptosis, are reversible processes under brief P2X7R stimulation, dissociating ATP-induced bleb formation and EV release from apoptosis [48].
Subsequent studies have clarified the mechanism by which P2X7R activation drives large EV biogenesis. The pathway involves the activation of p38 MAP kinase and Rho-associated protein kinases (ROCK) [49,50,51,52] (Figure 1). Specifically, following P2X7R activation, a Src kinase interacts with the C-terminus of the receptor and phosphorylates p38 MAP kinase, inducing the translocation of acid sphingomyelinase (A-SMase) from the luminal lysosomal compartment to the plasma membrane outer leaflet [49]. This enzyme hydrolyzes sphingomyelin to ceramide, thereby creating ceramide-enriched microdomains that, by perturbing membrane curvature/fluidity, facilitate the formation of plasma membrane blebs and large EV shedding from microglia and astrocytes [49]. The key role of A-SMase in EV release was indicated by the genetic inactivation and pharmacological inhibition of the A-SMase. Both approaches strongly abolished EV release from LPS-primed microglia and astrocytes [49] and alveolar macrophages [7] (see below). The in vivo validation of the role of P2X7R and A-SMase in EV release was suggested by the immunohistochemical quantification of EV-like particles immunoreactive for the P2X7R in the cerebral cortex of rats administered intraperitoneally with the P2X7R antagonist A804598 or the A-SMase inhibitor FTY720, immediately after traumatic brain injury (TBI), a condition inducing P2X7R expression and EV release from microglia [53]. Both drugs reduced the number of P2X7R positive particles surrounding microglia, but the particles were not unequivocally identified as EVs [53]. Beyond lipids, the P2X7R-dependent activation of ROCK is another key intracellular signaling mechanism in plasma membrane bleb formation. By interacting with LIM-kinases (LIMK) and the myosin light chain (MLC) [54,55], ROCK regulates cytoskeletal reorganization, favoring the shedding of large EV. The role of ROCK in EV release was supported by the complete abrogation of P2X7R-dependent blebbing in HEK293 cells pre-incubated with ROCK inhibitor Y-27632 [50].
In addition to large EVs shed from the plasma membrane, P2X7R activation promotes the release of small EVs generated in the endosomal compartment of innate immune cells [56,57,58]. Interestingly, in human macrophages the ATP-dependent small vesicles’ release was shown to be a consequence of NLRP3/ASC/procaspase-1 inflammasome assembling, as evidenced by the suppression of small EVs secretion under the genetic deletion of ASC or NLPR3 [57]. These findings suggest that inflammasome activation may regulate the membrane trafficking pathways that control MVBs fusion to the plasma membrane. The involvement of the NLRP3 inflammasome in small EV secretion was further indicated by a study showing that LPS/ATP-induced inflammasome and caspase-1 activation promotes the loading of specific miRNAs into small EVs and their release via the cleavage of the Rab-interacting lysosomal protein RILP in a human monocytic cell line. RILP is part of the complex that links the trafficking GTPase Rab7 to the dynein motor complex; cleaved RILP does not make the link to the dynein complex and promotes the movement of MVBs toward the cell surface (Figure 1). In addition, it induces selective miRNA cargo sorting via interaction with Hrs (hepatocyte growth factor-regulated tyrosine kinase substrate), a component of the ESCRT-0 complex, and the RNA-binding protein FMRP that acts as a chaperone to package specific AAUGC motif-containing miRNAs into ILV [59]. Accordingly, the inhibition of caspase-1 blocked small EV secretion from the monocytic cells activated with LPS/ATP [59]. Further advances in the mechanism driving the ATP-induced release of small EVs were made in 2022, when Ruan and colleagues identified Sepp1, Mcfd2, and Sdc1 as critical molecules for the release of CD63 positive small EVs from microglia using a genome-wide shRNA library screening [60]. The identified molecules may represent interesting targets for inhibiting small EV release and limiting the pathogenic contribution of EVs and their inflammatory cargo to neuroinflammatory disorders.

3. Role of P2X7R-Induced EVs in Cytokine Release and the Propagation of Inflammation

Cytokines lacking the conventional secretory sequence do not follow the classical endoplasmic-reticulum-to-Golgi pathway for secretion, but are exported via membrane-enclosed vesicles [61].
MacKenzie and colleagues showed that the pro-inflammatory cytokine IL-1β is packaged in EVs shed upon P2X7R-mediated monocyte activation, providing the first evidence that P2X7R-induced EV release represents an unconventional mechanism for the secretion of a leaderless protein [48]. In the following years, the presence of IL-1β was confirmed in EVs released from rat microglia and human dendritic cells [45,46], and the way in which IL-1β passes from the EVs lumen into the extracellular space was clarified. Specifically, it was observed that EVs contain the machinery necessary for IL-1β processing (they carry P2X7R in their membranes and caspase-1 in their lumen), and that P2X7R opening at the EV surface activates caspase-1-dependent IL-1β cleavage in the EVs, similar to the cells [45,46] (Figure 2). In addition, evidence was provided that IL-1β release may occur through the EV membrane as a consequence of P2X7R-dependent pore opening [5], and that pannexin-1 is required for IL-1β processing and secretion from macrophages [23].
Collectively, these results indicated that large EVs released upon P2X7R activation from immune cells carry IL-1β and mediate IL-1β secretion in a P2X7R-dependent manner. Later evidence obtained from macrophages and dendritic cells showed that small EVs also carry inflammasome components, i.e., NLP3, caspase-1, and ASC, that are essential for IL-1β processing within EVs [57,58,62]. These studies also showed that both small and large EVs released from mycobacterium-infected macrophages and dendritic cells upon P2X7R activation are enriched in major histocompatibility complex class II (MHC-II) [45,63,64], thus potentially contributing to the rapid dissemination and presentation of foreign antigens as part of the immune response induced by local inflammation [65,66]. In line with the involvement of EVs in the immune response, large EVs released upon P2X7R activation from LPS-treated microglia were reported to carry the IL-1β transcript and to act as vehicles for the transfer of IL-1β mRNA between immune cells, participating in the propagation of inflammatory signals both in vitro and in vivo, upon EV injection into the mouse corpus callosum [67].
Subsequent studies have revealed that large EVs released upon P2X7R activation mediate the release of other inflammatory cytokines, i.e., IL-18 and Tumor necrosis factor (TNF) [6,7] (Figure 2). Like IL-1β, IL-18 is a leaderless cytokine and its release from human blood-derived macrophages occurs in association with large EVs generated upon P2X7R activation [6]. Conversely, TNF is secreted by the classical endoplasmic-reticulum-to-Golgi pathway in a mature, soluble isoform of 17 kDa. Thus, the presence of TNF in EVs was quite unexpected. Notably, Soni and colleagues demonstrated that ATP stimulation alters the mechanism of TNF secretion from mouse bone-marrow derived macrophages, redirecting TNF release from classical to unconventional secretory pathways [7]. Specifically, ATP inhibits the conventional secretion of soluble TNF and drives the packaging of the transmembrane pro-TNF isoform into large EVs [7]. TNF carried by EVs was biologically more potent than soluble TNF at equal or even higher doses and mediated significant lung inflammation in vivo [7], revealing that ATP-dependent packaging into EVs uniquely protects enclosed TNF, enhancing its biological activity. These findings were confirmed in vivo upon the intratracheal instillation of ATP and the analysis of EV production and TNF quantification in the bronchoalveolar lavage fluid [7].
To conclude, relevant cytokines are expressed in EVs at both mRNA and protein levels and in both transmembrane/pro- and soluble/mature forms. The cytokines can be rapidly released from vesicles in the mature forms (IL-1β and IL-18) at sites of extracellular ATP accumulation via P2X7R opening, or be presented to recipient cells (pro-TNF), promoting acute inflammation. Given that packaging into EVs prevents the degradation and dilution of the inflammatory mediators, cytokines-loaded EVs released by P2X7R-expressing cells can propagate long-distance inflammatory signals to recipient cells and tissues. Cytokines released as part of EVs upon P2X7R activation are listed in Table 1.

4. P2X7R Activation Influences the Proteome of EVs

Distinct EV populations are released by cells in response to various stimuli that influence the cellular activation state [43], with EV composition often mirroring that of donor cells. As already mentioned, P2X7R activation influences the miRNA selectivity of small EV cargo loading through interactions with the RNA-binding protein FMR1 [59]. Furthermore, a few studies have identified proteins that are released as part of EVs via a P2X7R-dependent mechanism (Table 1). These molecules seem to share the ability to control the inflammatory response.
Takenouchi and coworkers showed that only under P2X7R activation was glyceraldehyde-3-phosphate dehydrogenase (GAPDH), a key glycolytic enzyme and a leaderless cytoplasmic protein, sorted in small and large EVs released by LPS-treated microglial cells [69]. Once in the extracellular space, GAPDH might be involved in the regulation of neuroinflammation by favoring the phosphorylation of p38 MAP kinase in microglia [69]. CD14 is another abundant protein cargo of EVs released upon P2X7R activation from macrophages [68]. P2X7R-induced CD14 release in EVs ensures the maintenance of elevated concentrations of circulating CD14 which, by acting as a co-receptor for LPS, is fundamental to controlling infection and increasing survival during sepsis [68].
Further studies associated P2X7R activation with the release of proteins modulating or amplifying the inflammatory response. The release of Interleukin-1 receptor antagonist (IL-Ra) occurs via a P2X7R-dependent large EV-shedding mechanism in macrophages [70]. Since IL-Ra functionally inhibits IL-1-dependent cellular activation, maintaining a balance between IL-1 and IL-1ra may be an important mechanism for regulating the overall inflammatory response [81]. Conversely, the mature form of the TNF converting enzyme (TACE), which is part of the small EVs produced by LPS-primed macrophages under P2X7R activation, by processing the membrane-bound TNF into a soluble cytokine may amplify the pro-inflammatory responses [71]. Finally, other studies have linked P2X7R activation to an increased release of tissue factor (TF) containing large EVs from human dendritic cells and macrophages, producing an enhanced pro-thrombotic response [44,72].
To the best of our knowledge, only one label-free proteomic study systematically explored how P2X7R activation influences the protein composition of EVs. This work showed that the proteome composition of EVs (large and small) released from microglia under ATP activation is largely distinct from that of constitutively released EVs [56]. Specifically, it revealed that EVs released under ATP stimulation contain an increased number of proteins involved in antigen processing and presentation which, along with inflammatory cytokines and MHC-II (see chapter above), can participate in the immune response. In addition, EVs released under P2X7R activation show an increased number of autophagy-lysosomal proteins (i.e., Cathepsin D and C, Lamp1, Vcp, and CD68), suggesting an enhancement of the degradative pathways, and are enriched in proteins implicated in adhesion/extracellular matrix organization (Fibulin 1, Comp, Plasminogen and the matricellular proteins thrombospondin 1 and 4, Vinculin, and Fermt3), which likely account for the stronger EV adhesion to astrocytic target cells compared to constitutive EVs. Interestingly, ATP also drives the sorting in EVs of a set of proteins involved in energy metabolism (i.e., Gpi, Ldha, Mdh2, Tranketolase, Glutamate dehydrogenase 1, Acacb, and others), which together with the glycolytic enzyme GAPDH identified by Takenouchi and colleagues may influence the metabolism of receiving cells. Finally, a unique set of cytoskeletal proteins and proteins regulating the dynamics of actin filaments have been detected in EVs released upon P2X7R activation, i.e., the capping actin protein Capzb, Cap1, and ARP2 actin related protein. These proteins, by controlling the organization of actin filaments present in a fraction of large EVs [82], may favor changes in the EV morphology and promote the capacity of a small fraction of glial EVs to actively move at the surface of target cells [73,82]. Interestingly, some of these cytoskeletal proteins interact with the C-terminus of the P2X7R [83], thus supporting a direct role for the receptor in the sorting of the protein cargo.
Further studies are necessary to define whether changes occurring in microglia-derived EVs under P2X7R activation may be common to EVs released by other cells following receptor stimulation.

5. P2X7R Activation and Misfolded Protein Release in EVs: Implications in Neurodegeneration

Among the bioactive cargo of EVs released upon P2X7R activation, there are pathological misfolded proteins including beta amyloid (Aβ) [73,74,75], tau protein [76,77,78], and α-synuclein [79,80] (for an extensive review, see [43] and Table 1).
By spreading throughout the brain in association with EVs, Aβ and tau protein contribute to the progression of neurodegeneration in AD and tauopathies (reviewed in [84]). Specifically, it has been demonstrated that EV-associated tau released by microglia after ATP stimulation, but not an equal amount of free tau, are able to mediate efficient tau propagation in the mouse hippocampus [76]. The pivotal involvement of P2X7R in this process has been proven by recent findings showing that the administration of the orally applicable and CNS-penetrant P2X7R selective antagonist GSK1482160, which inhibits EV secretion from microglia, blocks tau propagation and rescues memory impairment in the P301S mouse model of tauopathy [77]. Furthermore, the suppression of tau accumulation in the hippocampal region has been indicated in P301S mice lacking P2X7R (P2X7R−/−:P301S mice) [85]. Although for the EV-mediated propagation of Aβ no direct proof of P2X7R involvement by in vivo inhibition/depletion is currently available, large EVs released upon ATP activation by Aβ-exposed microglia, and injected in the mouse brain parenchyma, were shown to cause amyloid-related impairment of synaptic plasticity and propagate deficits to synaptically connected regions [73]. Again, free oligomeric Aβ was not able to propagate synaptic alterations [73].
Small EVs released upon ATP stimulation can also transfer α-synuclein, a key molecule in Parkinson’s disease pathogenesis, from microglia to neurons, where they act as seeds to aggregate the native protein [79]. Once injected in the striatum of healthy mice, microglial small EVs carrying α-synuclein, but not free α-synuclein, cause the aggregation of the protein at the injection site and in anatomically connected regions, and the loss of dopaminergic neurons in the nigrostriatal pathway associated with movement disorders months later [80].
Interestingly, P2X7R’s expression and function have been found to be altered in both AD/tauopathies patients and mouse models, especially in microglia and astrocytes surrounding amyloid plaques, while its genetic or pharmacological inhibition ameliorated the pathology in mice, mitigating inflammation and improving cognitive defects [86,87,88,89]. For these reasons, P2X7R has been implicated in both Aβ and tau-mediated neurodegeneration [87,89] and recognized as a promising pharmacological target for AD [90]. This also applies to Parkinson’s disease, as the pharmacological inhibition of P2X7R signaling limited hemi-Parkinsonism symptoms in a rat model of 6-hydroxydopamine-induced nigrostriatal lesion [91,92,93]. The involvement of the receptor in the EV-mediated propagation of misfolded proteins strengthened its potential as a therapeutic target for neurodegenerative diseases.
The presence of misfolded proteins in EVs released upon P2X7R activation indicates that EV release represents a mechanism exploited by cells to dispose of toxic material, which cannot be degraded in the cells, an old hypothesis formulated many years ago when EVs were still considered cellular debris or culture artefacts, and currently supported by many findings [73,75,79,94].

6. Conclusions

At inflammatory or damaged sites, P2X7R activation by extracellular ATP or NAD+ promotes the massive shedding of large EVs from the plasma membrane, via the translocation of acid sphingomyelinase and the release of small EVs from multivesicular bodies via inflammasome activation. The generated EVs expose MHCII on their surface and specific inflammatory miRNAs cargo in their lumen, and carry and release inflammatory cytokines into the extracellular space, promoting a local acute inflammatory response. Encapsulation into EVs can enhance cytokine activity, as shown for TNF, and by preventing cytokine degradation can deliver inflammatory signals to distant cells and tissues.
P2X7R-dependent EV release also represents a mechanism for the cells to dispose of unwanted materials, such as misfolded proteins (aβ, tau, and a-synuclein), which are resistant to degradation, and to disseminate them throughout the brain. Encapsulation into EVs can also increase the activity of misfolded proteins. In fact, Aβ, tau, and α–synuclein induce/propagate pathology more efficiently when associated with EVs, indicating that a higher activity of EV-associated proteins compared to free soluble forms is not a mere consequence of protection from degradation.
Further research is needed to better characterize the molecules modulating or amplifying the inflammatory/degenerative responses that are released as part of EVs upon P2X7R activation, in light of the emerging role of P2X7R inhibitors as promising therapeutic tools for limiting neurodegenerative and inflammatory processes.

Author Contributions

M.T.G., M.G., C.V.; original draft preparation, C.V.; review and editing, All authors have read and agreed to the published version of the manuscript.

Funding

This work was funded by the Ministry of Health, grant number RF-2018-12365333.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

AIM2, Absent-in-melanoma 2; ASC, Apoptosis-associated speck-like protein containing a CARD; A-SMase, Acid sphingomyelinase; Aβ, Beta amyloid; CNS, Central Nervous System; eATP, Extracellular ATP; EVs, Extracellular vesicles; GAPDH, Glyceraldehyde-3-phosphate dehydrogenase; Hrs, Hepatocyte growth-factor-regulated tyrosine kinase substrate; IL, Interleukin; IL-Ra, Interleukin-1 receptor antagonist; ILV, Intraluminal vesicle; LIMK, LIM-kinases; MHC-II, Major histocompatibility complex class II; MLC, myosin light chain; MVBs, multivesicular bodies; NAD+, Nicotinamide adenine dinucleotide; NLRC4, NLR Family CARD Domain Containing 4; NLRP1, NLR Family Pyrin Domain Containing 1; NLRP3, NLR Family Pyrin Domain Containing 3; P2X7R, P2X7 receptor; PS, Phosphatidylserine; RILP, Rab-interacting lysosomal protein; ROCK, Rho-associated protein kinases; TACE, TNF converting enzyme; TBI, Traumatic brain injury; TF, Tissue factor; TNF, Tumor necrosis factor.

References

  1. Steinberg, T.H.; Newman, A.S.; Swansonq, J.A.; Silverstein, S.C. ATP4-Permeabilizes the Plasma Membrane of Mouse Macrophages to Fluorescent Dyes. Chemists 1987, 262, 88–89. [Google Scholar] [CrossRef]
  2. Mutini, C.; Falzoni, S.; Ferrari, D.; Chiozzi, P.; Morelli, A.; Baricordi, O.R.; Collo, G.; Ricciardi-Castagnoli, P.; Di Virgilio, F. Mouse Dendritic Cells Express the P2X7 Purinergic Receptor: Characterization and Possible Participation in Antigen Presentation. J. Immunol. 1999, 163, 1958–1965. [Google Scholar] [CrossRef] [PubMed]
  3. Cockcroft, S.; Gomperts, B.D. ATP induces nucleotide permeability in rat mast cells. Nature 1979, 279, 541–542. [Google Scholar] [CrossRef] [PubMed]
  4. Visentin, S.; Renzi, M.; Frank, C.; Greco, A.; Levi, G. Two different ionotropic receptors are activated by ATP in rat microglia. J. Physiol. 1999, 519, 723. [Google Scholar] [CrossRef]
  5. Ferrari, D.; Pizzirani, C.; Adinolfi, E.; Lemoli, R.M.; Curti, A.; Idzko, M.; Panther, E.; Di Virgilio, F. The P2X7 receptor: A key player in IL-1 processing and release. J. Immunol. 2006, 176, 3877–3883. [Google Scholar] [CrossRef] [Green Version]
  6. Gulinelli, S.; Salaro, E.; Vuerich, M.; Bozzato, D.; Pizzirani, C.; Bolognesi, G.; Idzko, M.; Di Virgilio, F.; Ferrari, D. IL-18 associates to microvesicles shed from human macrophages by a LPS/TLR-4 independent mechanism in response to P2X receptor stimulation. Eur. J. Immunol. 2012, 42, 3334–3345. [Google Scholar] [CrossRef]
  7. Soni, S.; O’Dea, K.P.; Tan, Y.Y.; Cho, K.; Abe, E.; Romano, R.; Cui, J.; Ma, D.; Sarathchandra, P.; Wilson, M.R.; et al. ATP redirects cytokine trafficking and promotes novel membrane TNF signaling via microvesicles. FASEB J. 2019, 33, 6442–6455. [Google Scholar] [CrossRef] [Green Version]
  8. Grassi, F. The P2X7 Receptor as Regulator of T Cell Development and Function. Front. Immunol. 2020, 11, 1179. [Google Scholar] [CrossRef]
  9. Carotti, V.; Rigalli, J.P.; van Asbeck-van der Wijst, J.; Hoenderop, J.G.J. Interplay between purinergic signalling and extracellular vesicles in health and disease. Biochem. Pharmacol. 2022, 203, 115192. [Google Scholar] [CrossRef]
  10. Sperlágh, B.; Vizi, E.S.; Wirkner, K.; Illes, P. P2X7 receptors in the nervous system. Prog. Neurobiol. 2006, 78, 327–346. [Google Scholar] [CrossRef]
  11. Zhao, Y.F.; Tang, Y.; Illes, P. Astrocytic and Oligodendrocytic P2X7 Receptors Determine Neuronal Functions in the CNS. Front. Mol. Neurosci. 2021, 14, 9. [Google Scholar] [CrossRef] [PubMed]
  12. Welter-Stahl, L.; da Silva, C.M.; Schachter, J.; Persechini, P.M.; Souza, H.S.; Ojcius, D.M.; Coutinho-Silva, R. Expression of purinergic receptors and modulation of P2X7 function by the inflammatory cytokine IFNγ in human epithelial cells. Biochim. Biophys. Acta-Biomembr. 2009, 1788, 1176–1187. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Ray, F.R.; Huang, W.; Slater, M.; Barden, J.A. Purinergic receptor distribution in endothelial cells in blood vessels: A basis for selection of coronary artery grafts. Atherosclerosis 2002, 162, 55–61. [Google Scholar] [CrossRef] [PubMed]
  14. Agrawal, A.; Gartland, A. P2X7 receptors: Role in bone cell formation and function. J. Mol. Endocrinol. 2015, 54, R75–R88. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Solini, A.; Chiozzi, P.; Falzoni, S.; Morelli, A.; Fellin, R.; Di Virgilio, F. High glucose modulates P2X7 receptor-mediated function in human primary fibroblasts. Diabetologia 2000, 43, 1248–1256. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Li, X.; Hu, B.; Wang, L.; Xia, Q.; Ni, X. P2X7 receptor-mediated phenotype switching of pulmonary artery smooth muscle cells in hypoxia. Mol. Biol. Rep. 2021, 48, 2133–2142. [Google Scholar] [CrossRef]
  17. Scarpellino, G.; Genova, T.; Munaron, L. Purinergic P2X7 Receptor: A Cation Channel Sensitive to Tumor Microenvironment. Recent Pat. Anticancer Drug Discov. 2019, 14, 32–38. [Google Scholar] [CrossRef]
  18. Di Virgilio, F. P2X7 is a cytotoxic receptor….maybe not: Implications for cancer. Purinergic Signal. 2021, 17, 55–61. [Google Scholar] [CrossRef]
  19. Jiang, L.H.; Caseley, E.A.; Muench, S.P.; Roger, S. Structural basis for the functional properties of the P2X7 receptor for extracellular ATP. Purinergic Signal. 2021, 17, 331–344. [Google Scholar] [CrossRef]
  20. North, R.A. Molecular physiology of P2X receptors. Physiol. Rev. 2002, 82, 1013–1067. [Google Scholar] [CrossRef] [Green Version]
  21. Khadra, A.; Tomić, M.; Yan, Z.; Zemkova, H.; Sherman, A.; Stojilkovic, S.S. Dual Gating Mechanism and Function of P2X7 Receptor Channels. Biophys. J. 2013, 104, 2612–2621. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Di Virgilio, F.; Pizzo, P.; Zanovello, P.; Bronte, V.; Collavo, D. Extracellular ATP as a possible mediator of cell-mediated cytotoxicity. Immunol. Today 1990, 11, 274–277. [Google Scholar] [CrossRef] [PubMed]
  23. Pelegrin, P.; Surprenant, A. Pannexin-1 mediates large pore formation and interleukin-1β release by the ATP-gated P2X7 receptor. EMBO J. 2006, 25, 5071–5082. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Bianco, F.; Ceruti, S.; Colombo, A.; Fumagalli, M.; Ferrari, D.; Pizzirani, C.; Matteoli, M.; Di Virgilio, F.; Abbracchio, M.P.; Verderio, C. A role for P2X7 in microglial proliferation. J. Neurochem. 2006, 99, 745–758. [Google Scholar] [CrossRef]
  25. Monif, M.; Reid, C.A.; Powell, K.L.; Smart, M.L.; Williams, D.A. The P2X7 receptor drives microglial activation and proliferation: A trophic role for P2X7R pore. J. Neurosci. 2009, 29, 3781–3791. [Google Scholar] [CrossRef] [Green Version]
  26. Pelegrin, P. P2X7 receptor and the NLRP3 inflammasome: Partners in crime. Biochem. Pharmacol. 2021, 187, 114385. [Google Scholar] [CrossRef]
  27. Orioli, E.; De Marchi, E.; Giuliani, A.L.; Adinolfi, E. P2X7 Receptor Orchestrates Multiple Signalling Pathways Triggering Inflammation, Autophagy and Metabolic/Trophic Responses. Curr. Med. Chem. 2017, 24, 2261–2275. [Google Scholar] [CrossRef]
  28. Sharma, D.; Kanneganti, T.D. The cell biology of inflammasomes: Mechanisms of inflammasome activation and regulation. J. Cell Biol. 2016, 213, 617–629. [Google Scholar] [CrossRef] [Green Version]
  29. Di Virgilio, F. Liaisons dangereuses: P2X7 and the inflammasome. Trends Pharmacol. Sci. 2007, 28, 465–472. [Google Scholar] [CrossRef]
  30. Martinon, F.; Burns, K.; Tschopp, J. The Inflammasome: A molecular platform triggering activation of inflammatory caspases and processing of proIL-β. Mol. Cell 2002, 10, 417–426. [Google Scholar] [CrossRef]
  31. Adriouch, S.; Bannas, P.; Schwarz, N.; Fliegert, R.; Guse, A.H.; Seman, M.; Haag, F.; Koch-Nolte, F. ADP-ribosylation at R125 gates the P2X7 ion channel by presenting a covalent ligand to its nucleotide binding site. FASEB J. 2008, 22, 861–869. [Google Scholar] [CrossRef] [PubMed]
  32. Seman, M.; Adriouch, S.; Scheuplein, F.; Krebs, C.; Freese, D.; Glowacki, G.; Deterre, P.; Haag, F.; Koch-Nolte, F. NAD-Induced T Cell Death: ADP-Ribosylation of Cell Surface Proteins by ART2 Activates the Cytolytic P2X7 Purinoceptor. Immunity 2003, 19, 571–582. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Scheuplein, F.; Schwarz, N.; Adriouch, S.; Krebs, C.; Bannas, P.; Rissiek, B.; Seman, M.; Haag, F.; Koch-Nolte, F. NAD+ and ATP released from injured cells induce P2X7-dependent shedding of CD62L and externalization of phosphatidylserine by murine T cells. J. Immunol. 2009, 182, 2898–2908. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Nishida, K.; Nakatani, T.; Ohishi, A.; Okuda, H.; Higashi, Y.; Matsuo, T.; Fujimoto, S.; Nagasawa, K. Mitochondrial dysfunction is involved in P2X7 receptor-mediated neuronal cell death. J. Neurochem. 2012, 122, 1118–1128. [Google Scholar] [CrossRef]
  35. Pfister, M.; Ogilvie, A.; Da Silva, C.P.; Grahnert, A.; Guse, A.H.; Hauschildt, S. NAD degradation and regulation of CD38 expression by human monocytes/macrophages. Eur. J. Biochem. 2001, 268, 5601–5608. [Google Scholar] [CrossRef]
  36. Adriouch, S.; Hubert, S.; Pechberty, S.; Koch-Nolte, F.; Haag, F.; Seman, M. NAD+ released during inflammation participates in T cell homeostasis by inducing ART2-mediated death of naive T cells in vivo. J. Immunol. 2007, 179, 186–194. [Google Scholar] [CrossRef] [Green Version]
  37. Rissiek, B.; Haag, F.; Boyer, O.; Koch-Nolte, F.; Adriouch, S. P2X7 on mouse T cells: One channel, many functions. Front. Immunol. 2015, 6, 204. [Google Scholar] [CrossRef] [Green Version]
  38. Illes, P. P2X7 Receptors Amplify CNS Damage in Neurodegenerative Diseases. Int. J. Mol. Sci. 2020, 21, 5996. [Google Scholar] [CrossRef]
  39. van Niel, G.; Raposo, G. Shedding light on the cell biology of extracellular vesicles. Nat. Rev. Mol. Cell Biol. 2018, 19, 213–228. [Google Scholar] [CrossRef]
  40. 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. [Google Scholar] [CrossRef] [Green Version]
  41. Quek, C.; Hill, A.F. The role of extracellular vesicles in neurodegenerative diseases. Biochem. Biophys. Res. Commun. 2017, 483, 1178–1186. [Google Scholar] [CrossRef] [PubMed]
  42. 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. [Google Scholar] [CrossRef] [Green Version]
  43. Gabrielli, M.; Raffaele, S.; Fumagalli, M.; Verderio, C. The multiple faces of extracellular vesicles released by microglia: Where are we 10 years after? Front. Cell. Neurosci. 2022, 16, 984690. [Google Scholar] [CrossRef] [PubMed]
  44. Baroni, M.; Pizzirani, C.; Pinotti, M.; Ferrari, D.; Adinolfi, E.; Calzavarini, S.; Caruso, P.; Bernardi, F.; Di Virgilio, F. Stimulation of P2 (P2X7) receptors in human dendritic cells induces the release of tissue factor-bearing microparticles. FASEB J. 2007, 21, 1926–1933. [Google Scholar] [CrossRef] [PubMed]
  45. Pizzirani, C.; Ferrari, D.; Chiozzi, P.; Adinolfi, E.; Sandonà, D.; Savaglio, E.; Di Virgilio, F. Stimulation of P2 receptors causes release of IL-1-loaded microvesicles from human dendritic cells. Blood 2007, 109, 3856–3864. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Bianco, F.; Pravettoni, E.; Colombo, A.; Schenk, U.; Möller, T.; Matteoli, M.; Verderio, C. Astrocyte-derived ATP induces vesicle shedding and IL-1 beta release from microglia. J. Immunol. 2005, 174, 7268–7277. [Google Scholar] [CrossRef] [Green Version]
  47. Thomas, L.M.; Salter, R.D. Activation of macrophages by P2X7-induced microvesicles from myeloid cells is mediated by phospholipids and is partially dependent on TLR4. J. Immunol. 2010, 185, 3740–3749. [Google Scholar] [CrossRef] [Green Version]
  48. MacKenzie, A.; Wilson, H.L.; Kiss-Toth, E.; Dower, S.K.; North, R.A.; Surprenant, A. Rapid secretion of interleukin-1β by microvesicle shedding. Immunity 2001, 15, 825–835. [Google Scholar] [CrossRef]
  49. Bianco, F.; Perrotta, C.; Novellino, L.; Francolini, M.; Riganti, L.; Menna, E.; Saglietti, L.; Schuchman, E.H.; Furlan, R.; Clementi, E.; et al. Acid sphingomyelinase activity triggers microparticle release from glial cells. EMBO J. 2009, 28, 1043–1054. [Google Scholar] [CrossRef] [Green Version]
  50. Morelli, A.; Chiozzi, P.; Chiesa, A.; Ferrari, D.; Sanz, J.M.; Falzoni, S.; Pinton, P.; Rizzuto, R.; Olson, M.F.; Di Virgilio, F. Extracellular ATP Causes ROCK I-dependent Bleb Formation in P2X7-transfected HEK293 Cells. Mol. Biol. Cell 2003, 14, 2655–2664. [Google Scholar] [CrossRef] [Green Version]
  51. Pfeiffer, Z.A.; Aga, M.; Prabhu, U.; Watters, J.J.; Hall, D.J.; Bertics, P.J. The nucleotide receptor P2X7 mediates actin reorganization and membrane blebbing in RAW 264.7 macrophages via p38 MAP kinase and Rho. J. Leukoc. Biol. 2004, 75, 1173–1182. [Google Scholar] [CrossRef] [PubMed]
  52. Verhoef, P.A.; Estacion, M.; Schilling, W.; Dubyak, G.R. P2X7 receptor-dependent blebbing and the activation of Rho-effector kinases, caspases, and IL-1 beta release. J. Immunol. 2003, 170, 5728–5738. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Liu, X.; Zhao, Z.; Ji, R.; Zhu, J.; Sui, Q.Q.; Knight, G.E.; Burnstock, G.; He, C.; Yuan, H.; Xiang, Z. Inhibition of P2X7 receptors improves outcomes after traumatic brain injury in rats. Purinergic Signal. 2017, 13, 529–544. [Google Scholar] [CrossRef] [Green Version]
  54. Maekawa, M.; Ishizaki, T.; Boku, S.; Watanabe, N.; Fujita, A.; Iwamatsu, A.; Obinata, T.; Ohashi, K.; Mizuno, K.; Narumiya, S. Signaling from Rho to the actin cytoskeleton through protein kinases ROCK and LIM-kinase. Science 1999, 285, 895–898. [Google Scholar] [CrossRef] [PubMed]
  55. Kawano, Y.; Fukata, Y.; Oshiro, N.; Amano, M.; Nakamura, T.; Ito, M.; Matsumura, F.; Inagaki, M.; Kaibuchi, K. Phosphorylation of Myosin-Binding Subunit (Mbs) of Myosin Phosphatase by Rho-Kinase in Vivo. J. Cell Biol. 1999, 147, 1023. [Google Scholar] [CrossRef] [Green Version]
  56. Drago, F.; Lombardi, M.; Prada, I.; Gabrielli, M.; Joshi, P.; Cojoc, D.; Franck, J.; Fournier, I.; Vizioli, J.; Verderio, C. ATP modifies the proteome of extracellular vesicles released by microglia and influences their action on astrocytes. Front. Pharmacol. 2017, 8, 910. [Google Scholar] [CrossRef] [Green Version]
  57. Qu, Y.; Franchi, L.; Nunez, G.; Dubyak, G.R. Nonclassical IL-1 beta secretion stimulated by P2X7 receptors is dependent on inflammasome activation and correlated with exosome release in murine macrophages. J. Immunol. 2007, 179, 1913–1925. [Google Scholar] [CrossRef] [Green Version]
  58. Qu, Y.; Dubyak, G.R. P2X7 receptors regulate multiple types of membrane trafficking responses and non-classical secretion pathways. Purinergic Signal. 2009, 5, 163. [Google Scholar] [CrossRef] [Green Version]
  59. Wozniak, A.L.; Adams, A.; King, K.E.; Dunn, W.; Christenson, L.K.; Hung, W.T.; Weinman, S.A. The RNA binding protein FMR1 controls selective exosomal miRNA cargo loading during inflammation. J. Cell Biol. 2020, 219, e201912074. [Google Scholar] [CrossRef]
  60. Ruan, Z.; Takamatsu-Yukawa, K.; Wang, Y.; Ushman, M.L.; Thomas Labadorf, A.; Ericsson, M.; Ikezu, S.; Ikezu, T. Functional genome-wide short hairpin RNA library screening identifies key molecules for extracellular vesicle secretion from microglia. Cell Rep. 2022, 39, 110791. [Google Scholar] [CrossRef]
  61. Rubartelli, A.; Sitia, R. Secretion of Mammalian Proteins that Lack a Signal Sequence. In Unusual Secretory Pathways: From Bacteria to Man; Molecular Biology Intelligence Unit Book Series; Springer: Berlin/Heidelberg, Germany, 1997; pp. 87–114. [Google Scholar] [CrossRef]
  62. Sarkar, S.; Rokad, D.; Malovic, E.; Luo, J.; Harischandra, D.S.; Jin, H.; Anantharam, V.; Huang, X.; Lewis, M.; Kanthasamy, A.; et al. Manganese activates NLRP3 inflammasome signaling and propagates exosomal release of ASC in microglial cells. Sci. Signal. 2019, 12, eaat9900. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Qu, Y.; Ramachandra, L.; Mohr, S.; Franchi, L.; Harding, C.V.; Nunez, G.; Dubyak, G.R. P2X7 receptor-stimulated secretion of MHC class II-containing exosomes requires the ASC/NLRP3 inflammasome but is independent of caspase-1. J. Immunol. 2009, 182, 5052–5062. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Ramachandra, L.; Qu, Y.; Wang, Y.; Lewis, C.J.; Cobb, B.A.; Takatsu, K.; Boom, W.H.; Dubyak, G.R.; Harding, C.V. Mycobacterium tuberculosis Synergizes with ATP To Induce Release of Microvesicles and Exosomes Containing Major Histocompatibility Complex Class II Molecules Capable of Antigen Presentation. Infect. Immun. 2010, 78, 5116. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Barrera-Avalos, C.; Briceño, P.; Valdés, D.; Imarai, M.; Leiva-Salcedo, E.; Rojo, L.E.; Milla, L.A.; Huidobro-Toro, J.P.; Robles-Planells, C.; Escobar, A.; et al. P2X7 receptor is essential for cross-dressing of bone marrow-derived dendritic cells. iScience 2021, 24, 103520. [Google Scholar] [CrossRef]
  66. Zeng, F.; Morelli, A.E. Extracellular vesicle-mediated MHC cross-dressing in immune homeostasis, transplantation, infectious diseases, and cancer. Semin. Immunopathol. 2018, 40, 477–490. [Google Scholar] [CrossRef]
  67. Verderio, C.; Muzio, L.; Turola, E.; Bergami, A.; Novellino, L.; Ruffini, F.; Riganti, L.; Corradini, I.; Francolini, M.; Garzetti, L.; et al. Myeloid microvesicles are a marker and therapeutic target for neuroinflammation. Ann. Neurol. 2012, 72, 610–624. [Google Scholar] [CrossRef] [Green Version]
  68. Alarcón-Vila, C.; Baroja-Mazo, A.; de Torre-Minguela, C.; Martínez, C.M.; Martínez-García, J.J.; Martínez-Banaclocha, H.; García-Palenciano, C.; Pelegrin, P. CD14 release induced by P2X7 receptor restricts inflammation and increases survival during sepsis. eLife 2020, 9, e60849. [Google Scholar] [CrossRef]
  69. Takenouchi, T.; Tsukimoto, M.; Iwamaru, Y.; Sugama, S.; Sekiyama, K.; Sato, M.; Kojima, S.; Hashimoto, M.; Kitani, H. Extracellular ATP induces unconventional release of glyceraldehyde-3-phosphate dehydrogenase from microglial cells. Immunol. Lett. 2015, 167, 116–124. [Google Scholar] [CrossRef] [Green Version]
  70. Wilson, H.L.; Francis, S.E.; Dower, S.K.; Crossman, D.C. Secretion of intracellular IL-1 receptor antagonist (type 1) is dependent on P2X7 receptor activation. J. Immunol. 2004, 173, 1202–1208. [Google Scholar] [CrossRef] [Green Version]
  71. Barberà-Cremades, M.; Gómez, A.I.; Baroja-Mazo, A.; Martínez-Alarcón, L.; Martínez, C.M.; de Torre-Minguela, C.; Pelegrín, P. P2X7 Receptor Induces Tumor Necrosis Factor-α Converting Enzyme Activation and Release to Boost TNF-α Production. Front. Immunol. 2017, 8, 862. [Google Scholar] [CrossRef] [Green Version]
  72. Moore, S.F.; MacKenzie, A.B. Murine macrophage P2X7 receptors support rapid prothrombotic responses. Cell. Signal. 2007, 19, 855–866. [Google Scholar] [CrossRef] [PubMed]
  73. Gabrielli, M.; Prada, I.; Joshi, P.; Falcicchia, C.; D’Arrigo, G.; Rutigliano, G.; Battocchio, E.; Zenatelli, R.; Tozzi, F.; Radeghieri, A.; et al. Microglial large extracellular vesicles propagate early synaptic dysfunction in Alzheimer’s disease. Brain 2022, 145, 2849–2868. [Google Scholar] [CrossRef] [PubMed]
  74. Gouwens, L.K.; Ismail, M.S.; Rogers, V.A.; Zeller, N.T.; Garrad, E.C.; Amtashar, F.S.; Makoni, N.J.; Osborn, D.C.; Nichols, M.R. Aβ42 Protofibrils Interact with and Are Trafficked through Microglial-Derived Microvesicles. ACS Chem. Neurosci. 2018, 9, 1416–1425. [Google Scholar] [CrossRef] [PubMed]
  75. Joshi, P.; Turola, E.; Ruiz, A.; Bergami, A.; Libera, D.D.; Benussi, L.; Giussani, P.; Magnani, G.; Comi, G.; Legname, G.; et al. Microglia convert aggregated amyloid-β into neurotoxic forms through the shedding of microvesicles. Cell Death Differ. 2014, 21, 582–593. [Google Scholar] [CrossRef] [Green Version]
  76. Asai, H.; Ikezu, S.; Tsunoda, S.; Medalla, M.; Luebke, J.; Haydar, T.; Wolozin, B.; Butovsky, O.; Kügler, S.; Ikezu, T. Depletion of microglia and inhibition of exosome synthesis halt tau propagation. Nat. Neurosci. 2015, 18, 1584–1593. [Google Scholar] [CrossRef] [Green Version]
  77. Ruan, Z.; Delpech, J.C.; Venkatesan Kalavai, S.; Van Enoo, A.A.; Hu, J.; Ikezu, S.; Ikezu, T. P2RX7 inhibitor suppresses exosome secretion and disease phenotype in P301S tau transgenic mice. Mol. Neurodegener. 2020, 15, 47. [Google Scholar] [CrossRef]
  78. Crotti, A.; Sait, H.R.; McAvoy, K.M.; Estrada, K.; Ergun, A.; Szak, S.; Marsh, G.; Jandreski, L.; Peterson, M.; Reynolds, T.L.; et al. BIN1 favors the spreading of Tau via extracellular vesicles. Sci. Rep. 2019, 9, 9477. [Google Scholar] [CrossRef] [Green Version]
  79. Fan, R.Z.; Guo, M.; Luo, S.; Cui, M.; Tieu, K. Exosome release and neuropathology induced by α-synuclein: New insights into protective mechanisms of Drp1 inhibition. Acta Neuropathol. Commun. 2019, 7, 184. [Google Scholar] [CrossRef]
  80. Guo, M.; Wang, J.; Zhao, Y.; Feng, Y.; Han, S.; Dong, Q.; Cui, M.; Tieu, K. Microglial exosomes facilitate α-synuclein transmission in Parkinson’s disease. Brain 2020, 143, 1476–1497. [Google Scholar] [CrossRef]
  81. Arend, W.P. The balance between IL-1 and IL-1Ra in disease. Cytokine Growth Factor Rev. 2002, 13, 323–340. [Google Scholar] [CrossRef]
  82. D’Arrigo, G.; Gabrielli, M.; Scaroni, F.; Swuec, P.; Amin, L.; Pegoraro, A.; Adinolfi, E.; Di Virgilio, F.; Cojoc, D.; Legname, G.; et al. Astrocytes-derived extracellular vesicles in motion at the neuron surface: Involvement of the prion protein. J. Extracell. Vesicles 2021, 10, e12114. [Google Scholar] [CrossRef] [PubMed]
  83. Gu, B.J.; Rathsam, C.; Stokes, L.; McGeachie, A.B.; Wiley, J.S. Extracellular ATP dissociates nonmuscle myosin from P2X7 complex: This dissociation regulates P2X7 pore formation. Am. J. Physiol.-Cell Physiol. 2009, 297, 430–439. [Google Scholar] [CrossRef] [Green Version]
  84. Gabrielli, M.; Tozzi, F.; Verderio, C.; Origlia, N. Emerging Roles of Extracellular Vesicles in Alzheimer’s Disease: Focus on Synaptic Dysfunction and Vesicle–Neuron Interaction. Cells 2023, 12, 63. [Google Scholar] [CrossRef] [PubMed]
  85. Abdullah, M.; Ruan, Z.; Bueser, K.R.; Ikezu, S.; Ikezu, T. The systemic disruption of P2rx7 alleviates tau pathology in P301S tau mice via inhibition of extracellular vesicle release. Alzheimer’s Dement. 2022, 18, e064257. [Google Scholar] [CrossRef]
  86. Francistiová, L.; Bianchi, C.; Di Lauro, C.; Sebastián-Serrano, Á.; de Diego-García, L.; Kobolák, J.; Dinnyés, A.; Díaz-Hernández, M. The Role of P2X7 Receptor in Alzheimer’s Disease. Front. Mol. Neurosci. 2020, 13, 94. [Google Scholar] [CrossRef] [PubMed]
  87. Carvalho, K.; Martin, E.; Ces, A.; Sarrazin, N.; Lagouge-Roussey, P.; Nous, C.; Boucherit, L.; Youssef, I.; Prigent, A.; Faivre, E.; et al. P2X7-deficiency improves plasticity and cognitive abilities in a mouse model of Tauopathy. Prog. Neurobiol. 2021, 206, 102139. [Google Scholar] [CrossRef] [PubMed]
  88. Di Lauro, C.; Bianchi, C.; Sebastián-Serrano, Á.; Soria-Tobar, L.; Alvarez-Castelao, B.; Nicke, A.; Díaz-Hernández, M. P2X7 receptor blockade reduces tau induced toxicity, therapeutic implications in tauopathies. Prog. Neurobiol. 2022, 208, 102173. [Google Scholar] [CrossRef]
  89. Martin, E.; Amar, M.; Dalle, C.; Youssef, I.; Boucher, C.; Le Duigou, C.; Brückner, M.; Prigent, A.; Sazdovitch, V.; Halle, A.; et al. New role of P2X7 receptor in an Alzheimer’s disease mouse model. Mol. Psychiatry 2019, 24, 108–125. [Google Scholar] [CrossRef] [Green Version]
  90. Illes, P.; Rubini, P.; Huang, L.; Tang, Y. The P2X7 receptor: A new therapeutic target in Alzheimer’s disease. Expert Opin. Ther. Targets 2019, 23, 165–176. [Google Scholar] [CrossRef]
  91. Carmo, M.R.S.; Menezes, A.P.F.; Nunes, A.C.L.; Pliássova, A.; Rolo, A.P.; Palmeira, C.M.; Cunha, R.A.; Canas, P.M.; Andrade, G.M. The P2X7 receptor antagonist Brilliant Blue G attenuates contralateral rotations in a rat model of Parkinsonism through a combined control of synaptotoxicity, neurotoxicity and gliosis. Neuropharmacology 2014, 81, 142–152. [Google Scholar] [CrossRef]
  92. Ferrazoli, E.G.; de Souza, H.D.N.; Nascimento, I.C.; Oliveira-Giacomelli, Á.; Schwindt, T.T.; Britto, L.R.; Ulrich, H. Brilliant Blue G, But Not Fenofibrate, Treatment Reverts Hemiparkinsonian Behavior and Restores Dopamine Levels in an Animal Model of Parkinson’s Disease. Cell Transplant. 2017, 26, 669–677. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Oliveira-Giacomelli, Á.; Albino, C.M.; de Souza, H.D.N.; Corrêa-Velloso, J.; de Jesus Santos, A.P.; Baranova, J.; Ulrich, H. P2Y6 and P2X7 Receptor Antagonism Exerts Neuroprotective/ Neuroregenerative Effects in an Animal Model of Parkinson’s Disease. Front. Cell. Neurosci. 2019, 13, 476. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Xia, Y.; Zhang, G.; Han, C.; Ma, K.; Guo, X.; Wan, F.; Kou, L.; Yin, S.; Liu, L.; Huang, J.; et al. Microglia as modulators of exosomal alpha-synuclein transmission. Cell Death Dis. 2019, 10, 174. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Figure 1. Schematic representation of EV release upon ATP-induced P2X7R activation. Upon ATP stimulation, P2X7R activation induces the recruitment of an Src kinase at the C-terminus of the receptor and the activation of ROCK and p38 MAP kinase triggering cytoskeletal reorganization (i.e., MLC and LIMK) and the translocation of A-SMase to the outer leaflet of the plasma membrane. A-SMase hydrolyzes sphingomyelin to ceramide, facilitating blebs’ formation and large EV shedding (route 1). P2X7R activation and the consequent efflux of K+ also promotes the release of small EVs, generated in the endosomal compartment as ILVs, by inducing NLRP3/ASC/procaspase-1 inflammasome assembling. Inflammasome activation regulates the membrane trafficking pathways that control MVB fusion to the plasma membrane via the cleavage of RILP (route 2). Image created with BioRender.com (accessed on 15 March 2023).
Figure 1. Schematic representation of EV release upon ATP-induced P2X7R activation. Upon ATP stimulation, P2X7R activation induces the recruitment of an Src kinase at the C-terminus of the receptor and the activation of ROCK and p38 MAP kinase triggering cytoskeletal reorganization (i.e., MLC and LIMK) and the translocation of A-SMase to the outer leaflet of the plasma membrane. A-SMase hydrolyzes sphingomyelin to ceramide, facilitating blebs’ formation and large EV shedding (route 1). P2X7R activation and the consequent efflux of K+ also promotes the release of small EVs, generated in the endosomal compartment as ILVs, by inducing NLRP3/ASC/procaspase-1 inflammasome assembling. Inflammasome activation regulates the membrane trafficking pathways that control MVB fusion to the plasma membrane via the cleavage of RILP (route 2). Image created with BioRender.com (accessed on 15 March 2023).
Ijms 24 09805 g001
Figure 2. Schematic representation of cytokine processing in EVs upon ATP-induced P2X7R activation. P2X7R activation, by inducing NLRP3 inflammasome assembling, triggers pro-caspase-1 cleavage, which generates active caspase-1 that, in turn, drives the enzymatic activation of the leaderless pro-inflammatory cytokines IL-1β and IL-18. On the other hand, ATP induces TNF packaging into EVs as a transmembrane pro-TNF isoform. Image created with BioRender.com (accessed on 15 March 2023).
Figure 2. Schematic representation of cytokine processing in EVs upon ATP-induced P2X7R activation. P2X7R activation, by inducing NLRP3 inflammasome assembling, triggers pro-caspase-1 cleavage, which generates active caspase-1 that, in turn, drives the enzymatic activation of the leaderless pro-inflammatory cytokines IL-1β and IL-18. On the other hand, ATP induces TNF packaging into EVs as a transmembrane pro-TNF isoform. Image created with BioRender.com (accessed on 15 March 2023).
Ijms 24 09805 g002
Table 1. Proteins released as part of EV cargo upon P2X7R activation and their phato/physiological impact.
Table 1. Proteins released as part of EV cargo upon P2X7R activation and their phato/physiological impact.
EV CargoEV TypeEV Cellular SourceInvolved Patho/Physiological ProcessesRefs.
Fibulin 1, Comp, Plasminogen and the matricellular proteins thrombospondin 1 and 4, Vinculin, and Fermt3Small and large EVsRat microgliaAdhesion/extracellular matrix organization[56]
Cathepsin D and C, Lamp1, Vcp, and CD68Small and large EVsRat microgliaAutophagy-lysosomal pathway[56]
Capzb, Cap1, and ARP2 actin-related proteinSmall and large EVsRat microgliaCytoskeleton organization[56]
MHC-IISmall EVsMurine macrophages and dendritic cellsDissemination and presentation of
foreign antigens
[63]
Large EVsHuman dendritic cells[45]
Small and large EVsMurine macrophages[64]
Gpi, Ldha, Mdh2, Tranketolase, Glutamate dehydrogenase 1, Acacb, and othersSmall and large EVsRat microgliaEnergy metabolism[56]
CD14EVsMurine macrophagesInflammation[68]
GAPDHSmall and large EVsMurine microglia[69]
IL-18Large EVsHuman macrophages[6]
IL1βLarge EVsHuman monocytes
Rat microglia
Human dendritic cells
[48]
[46]
[45]
Small EVsMurine macrophages[57]
IL-RaLarge EVsMurine macrophages[70]
Inflammasome componentsLarge EVsRat microglia
Human dendritic cells
[46]
[45]
Small EVsMurine macrophages
Murine microglia
[57]
[62]
TACESmall EVs Mouse macrophages[71]
TFLarge EVsHuman dendritic cells
Murine macrophages
[44]
[72]
TNFLarge EVsMurine macrophage[7]
Large EVsMurine microglia
Rat microglia
Neurodegeneration[73,74]
[75]
Tau protein Small EVsMurine microglia[76,77]
Small EVsMurine microglia[78]
α-synucleinSmall EVsMurine microglia[79,80]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Golia, M.T.; Gabrielli, M.; Verderio, C. P2X7 Receptor and Extracellular Vesicle Release. Int. J. Mol. Sci. 2023, 24, 9805. https://doi.org/10.3390/ijms24129805

AMA Style

Golia MT, Gabrielli M, Verderio C. P2X7 Receptor and Extracellular Vesicle Release. International Journal of Molecular Sciences. 2023; 24(12):9805. https://doi.org/10.3390/ijms24129805

Chicago/Turabian Style

Golia, Maria Teresa, Martina Gabrielli, and Claudia Verderio. 2023. "P2X7 Receptor and Extracellular Vesicle Release" International Journal of Molecular Sciences 24, no. 12: 9805. https://doi.org/10.3390/ijms24129805

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop