Skip to main content
Purinergic Signalling logoLink to Purinergic Signalling
. 2009 Feb 3;5(2):163–173. doi: 10.1007/s11302-009-9132-8

P2X7 receptors regulate multiple types of membrane trafficking responses and non-classical secretion pathways

Yan Qu 2, George R Dubyak 1,
PMCID: PMC2686822  PMID: 19189228

Abstract

Activation of the P2X7 receptor (P2X7R) triggers a remarkably diverse array of membrane trafficking responses in leukocytes and epithelial cells. These responses result in altered profiles of cell surface lipid and protein composition that can modulate the direct interactions of P2X7R-expressing cells with other cell types in the circulation, in blood vessels, at epithelial barriers, or within sites of immune and inflammatory activation. Additionally, these responses can result in the release of bioactive proteins, lipids, and large membrane complexes into extracellular compartments for remote communication between P2X7R-expressing cells and other cells that amplify or modulate inflammation, immunity, and responses to tissue damages. This review will discuss P2X7R-mediated effects on membrane composition and trafficking in the plasma membrane (PM) and intracellular organelles, as well as actions of P2X7R in controlling various modes of non-classical secretion. It will review P2X7R regulation of: (1) phosphatidylserine distribution in the PM outer leaflet; (2) shedding of PM surface proteins; (3) release of PM-derived microvesicles or microparticles; (4) PM blebbing; (5) cell–cell fusion resulting in formation of multinucleate cells; (6) phagosome maturation and fusion with lysosomes; (7) permeability of endosomes with internalized pathogen-associated molecular patterns; (8) permeability/integrity of mitochondria; (9) exocytosis of secretory lysosomes; and (10) release of exosomes from multivesicular bodies.

Keywords: P2X7 receptor, Plasma membrane, Non-classical secretion, Membrane trafficking

Introduction

Although rapid changes in ionic fluxes and ion homeostasis are the best characterized cellular responses to activation of the P2X7 receptor (P2X7R), this adenosine triphosphate (ATP)-gated ion channel also triggers a remarkably diverse array of membrane trafficking responses in immune and inflammatory effector cells. These responses result in altered profiles of cell surface lipid and protein composition that can modulate the direct interactions of P2X7R-expressing leukocytes with other cell types in the circulation, the vasculature, or sites of immune and inflammatory activation. Additionally, these responses can result in the release of bioactive proteins, lipids, and large membrane complexes into extracellular compartments for longer range and remote communication between P2X7R-expressing leukocytes and other cells that amplify or modulate innate and acquired immunity. As an organizational framework, this review will first discuss P2X7R-mediated regulation of membrane composition and trafficking at the level of the plasma membrane, and will then survey known or likely actions of P2X7R in controlling the composition or trafficking of intracellular membranes.

P2X7R-mediated regulation of plasma membrane composition and trafficking

P2X7R regulation of phosphatidylserine distribution in the plasma membrane

Phosphatidylserine (PS) transfer to the extracellular leaflet of the plasma membrane (PM) is a well-characterized signal that facilitates: (1) the clearance of apoptotic/senescent cells or apoptotic bodies by professional or non-professional phagocytes; and (2) the activation of platelet-mediated hemostatic responses [13]. The outer leaflet of eukaryotic plasma membrane bilayers predominantly contains sphingomyelin and phosphatidylcholine while aminophospholipids, such as PS and phosphatidylethanolamine, are enriched in the inner leaflet. Phospholipid distribution within these outer and inner leaflets of the PM is tightly regulated by specialized enzymatic mechanisms that include “flippases”, “floppases”, and “scramblases” [4, 5]. Loss of membrane phospholipid asymmetry, particularly PS flip from the inner to the outer leaflets, has been associated with many pathologic conditions. Although the mechanism underlying loss of PS asymmetry remains only partially understood, current models propose that two ATP-dependent transporters—“flippase” and “floppase”—actively maintain inward or outward aminophospholipid transport while a Ca2+-dependent, but ATP-independent, scramblase mediates the bidirectional movement of phospholipids between the bilayer leaflets [6].

Activation of P2X7R, which induces a rapid and sustained Ca2+ influx, has been reported to stimulate PS translocation (as measured by fluorescein-conjugated annexin-V binding to the plasma membrane) in HEK293 cells expressing the P2X7R [7]. While prolonged activation of P2X7R results in irreversible PS flip and cell death within 6 h, brief pulses of P2X7R stimulation (≤10 min) trigger reversible PS flip that precedes microvesicle shedding and is independent of apoptotic cell death. Although—as noted above—PS translocation is considered a key indicator of apoptotic cell death, these observations regarding reversible PS flip during brief P2X7R activation suggest that cell surface PS redistribution can be part of normal physiological responses to developmental cues or cell stress. Indeed, this notion of physiological PS exposure is supported by an increasing number of examples. Transient expression of PS on the surface is essential for skeletal muscle cell differentiation and mediates myotube formation [8]. Likewise, exposure of sperm cells to bicarbonate results in profound architectural changes in the sperm cell plasma membrane including a PS flip that appears to be an important and early physiological event during sperm capacitation [9]. Neutrophils stimulated with chemotactic peptides, such as formylated Met-Leu-Phe (fMLP), exhibit transient cell surface PS expression [10]. Macrophages per se undergo membrane PS flip during their recognition and phagocytosis of dead cells [1114]. Similarly, rapid changes in cell surface PS triggered by the natively expressed P2X7R in various leukocyte types have been associated with physiological signal transduction. Elliott et al. have described a P2X7R-stimulated PS asymmetry in non-apoptotic murine T lymphocytes that is associated with altered expression of adhesion molecules, negative regulation of the CD45 transmembrane tyrosine phosphatase, and suppressed activity of P-glycoprotein [15]. Moreover, a high level of PS in the inner leaflet of the T-cell plasma membrane markedly inhibits channel activity of one naturally occurring allelic variant (Leu-451) of the murine P2X7R, but not the Pro-451 variant of this receptor. The mechanism(s) underlying P2X7R-induced changes in PS asymmetry remain incompletely defined but likely involve indirect effects of altered ionic flux on floppase/scramblase activation or, perhaps, direct protein–protein interaction between these enzymes and the P2X7R itself.

Another potential physiological role for the P2X7R-induced PS exposure observed in non-apoptotic T lymphocytes is modulation of T-cell migration, particularly extravasation and homing to inflammatory loci. This possibility is supported by several observations: (1) PS translocation has been correlated with increased cellular adhesion to endothelia [16]; and (2) activation of P2X7R triggers shedding of CD62L [15], a major homing receptor [17], whose shedding occurs immediately before extravasation at inflammatory loci [18]. Moreover, as with PS exposure, shedding of CD62L from T cells occurs within seconds of P2X7R activation. MDR1 inhibitors that block PS translocation also completely prevent P2X7R-induced CD62L shedding [15]. It is possible that the increased membrane fluidity resulting from P2X7R activation facilitates the marked changes in cell shape required for the diapedesis of leukocytes between the packed endothelial cells that comprise these barriers.

P2X7R regulation of plasma membrane protein shedding

As noted above, P2X7R activation in murine T cells triggers a rapid loss of cell surface CD62L (l-selectin), which is correlated with the increased levels of cell surface PS. This latter finding reiterates the initial observations by Wiley and colleagues who demonstrated an extracellular ATP-mediated loss of CD62L from human leukemic B cells [19, 20]. Subsequent studies of blood leukocytes from normal C57BL/6 mice versus P2X7R knockout mice verified that the ATP-induced loss of CD62L observed in murine T cells, B cells, and monocytes was absolutely dependent on P2X7R rather than other P2 receptor subtypes [21]. Because CD62L is an intrinsic membrane protein with a single membrane-spanning domain, a reduction in cell surface CD62L can reflect: (1) protease-mediated shedding of the large CD62L ecto-domain; (2) internalization of CD62L via endocytosis; or (3) release of intact C62L protein within membrane vesicles that evaginate and scission away from the plasma membrane (see below). The ability of matrix metalloprotease (MMP) inhibitors to markedly suppress P2X7R-dependent loss of surface CD62L from human B cells indicated that bona fide shedding of the ecto-domain is the predominant mechanism. P2X7R activation in human B cells and monocyte-derived dendritic cells has also been shown to elicit the shedding of CD23, an intrinsic plasma membrane protein that functions as a low-affinity receptor for IgE with links to chronic inflammatory diseases [22, 23]. The ATP-induced loss of surface anti-CD23 epitopes was correlated with the extracellular accumulation of soluble CD23 protein which was suppressed in the presence of MMP inhibitors. CD27, a 55-kDa member of the TNF receptor superfamily, is another cell surface protein that is rapidly shed from murine T lymphocytes by a MMP-mediated mechanism in response to P2X7R activation [24]. The ligand for CD27 is CD70, yet another intrinsic plasma membrane protein expressed on activated dendritic cells or B cells. CD27–CD70 interactions occur in the context of antigen presentation to effector or memory T cells.

Thus, each of the three membrane proteins known to be shed during P2X7R stimulation is involved in a temporally or spatially distinct phase of the immune response: homing of leukocytes to immune/inflammatory loci (CD62L); direct interactions between antigen-presenting and antigen-responsive subsets of leukocytes (CD27); and activation of leukocytes by soluble immunoregulatory molecules that accumulate during particular immunological states (CD23). This suggests that local P2X7R signaling can fine-tune the duration or intensity of these diverse phases of innate or acquired immunity. It is likely that other leukocyte surface proteins can be shed via MMP-mediated pathways that are entrained by transient P2X7R stimulation. Major unresolved issues include the mechanism(s) by P2X7R activation is coupled to MMP activation and identification of the particular MMP subtypes that are targeted by this ATP-gated ion channel.

P2X7R regulation of plasma membrane microvesicle release

The ability of P2X7R activation to rapidly modulate the composition of the cell surface proteome extends beyond the MMP-mediated shedding of specific membrane proteins described above. Stimulus-induced shedding of plasma membrane-derived microvesicles—which bear distinctive complements of membrane markers—has been described in both hematopoietic cell types (platelets, DCs, and neutrophils) and non-hematopoietic cell types (epithelial cells, cancer cells, and neurons) [2529]. The size of these released vesicles ranges between 100 nm and 1 μm in diameter which distinguishes them from the 1–4-μm apoptotic bodies containing fragmented DNA derived from damaged cells, and from the 30–80-nm exosomes (discussed below) derived from the intraluminal vesicles of endosomal multivesicular bodies (MVB). Depending on their cellular origin and the stimulus which triggers their release, microvesicles can differ with regard to intravesicular contents, membrane lipid and protein compositions, biological functions, and nomenclature. Small membrane vesicles released into the circulation from activated platelets are described in the literature as microparticles (MP); these have major roles in blood coagulation by providing a large surface area for the assembly of clotting factors that bind to the exposed phosphatidylserine of the released MPs [30, 31]. Activated human neutrophils release “ectosomes” that exhibit anti-inflammatory activity and mediate inhibition of DC maturation from cultured blood monocytes [26, 27, 32]. Particles secreted from the basolateral membranes of various epithelial cells during tissue development are called “argosomes” due to their involvement in the transfer of developmental morphogens between cells and the maintenance of morphogen gradients [33].

Of particular relevance to this review, stimulation of P2X7R in human DC [34], murine microglial lines [35], and THP-1 human monocytes [36] triggers rapid release of plasma membrane-derived microvesicles (MV) which have been associated with the non-classical secretion of IL-1β and IL-18 in response to extracellular ATP. This ATP-induced microvesicle shedding is correlated with decreased plasma membrane capacitance [36] and is markedly inhibited by the removal of extracellular Ca2+ [28] or the pretreatment of cells with P2X7R antagonists such as oxidized ATP or KN-62 [34]. The shed microvesicles can contain: (1) soluble proteins including unprocessed proIL-1β, caspase-1 processed mature IL-1β, and the caspase-1 itself; (2) intrinsic membrane proteins including MHC-II, P2X7R itself, P2Y2 receptors, CD63, CD39, and LAMP-1 [35, 3740]; and (3) an increased level of surface active phospholipids such as PS [36]. Therefore, released MV have been proposed, or might be considered, as: (1) effective mechanisms for extracellular delivery of leaderless proteins such as bioactive cytokines (IL-1β and IL-18), the so-called danger molecules such as the high mobility group box 1 protein [41, 42], and normal intracellular thiol-reducing agents [41, 42]; or (2) a novel route for the long range intercellular transfer of membrane proteins and lipids (MHC-II, P2X7R, P2Y2, CD39, PS).

The molecular basis for P2X7R-dependent microvesicle shedding remains poorly characterized. This shedding may reflect direct effects of altered ion homeostasis, particularly increased cytosolic Ca2+ on both proteases that regulate association of the plasma membrane with the submembrane cytoskeleton and on phospholipases that induce localized changes in lipid composition and fluidity with consequent perturbation of bilayer lipid packing and geometry. It is also possible that danger signals other than extracellular ATP can act synergistically with gated P2X7R to modulate the rate, extent, and composition of microvesicle release. For example, lipopolysaccharide (LPS) per se has been shown to induce a slow microvesicle release from human monocytes [43]. However, the rapid ATP-induced release of P2X7R-positive microvesicle membranes from murine macrophages can occur independently of LPS priming and before IL-1β maturation and secretion [40].

P2X7R regulation of plasma membrane blebbing

Membrane blebbing, which can be related to, but also distinguished from, microvesicle shedding, represents a phenomenon wherein sections of plasma membrane reversibly protrude and retract at the cell surface. A variety of stimuli, including the activation of P2X7R, have been shown to trigger membrane blebbing. However, neither the intracellular mechanisms that regulate bleb formation nor the physiological or pathological roles of these blebs are entirely understood. This dynamic and reversible blebbing triggered by P2X7R may be involved in directed migration of leukocytes through extracellular matrices to sites of infection or tissue damage, and/or the membrane trafficking responses that culminate in scission of the extended blebs away from the plasma membrane surface to produce the released microvesicles/microparticles discussed in the previous section. Studies with different cell types have indicated that P2X7R-stimulated membrane blebbing appears to involve several common signaling pathways [44, 45]. ATP-induced membrane blebbing was found to be concurrent with actin reorganization in murine macrophages [46] and could be dissociated from the non-classical release of mature IL-1β that is also stimulated in parallel by P2X7R in such macrophages [47]. Disruption of actin filaments by cytochalasin D, which inhibits actin polymerization and depolymerization, diminished the P2X7R-induced blebbing without affecting the receptor’s ionotropic function. Activation of Rho and p38 MAPKs occurred concurrently with P2X7R-induced membrane blebbing [46]. Likewise, pharmacological inhibitors of p38, Rho, and Rho kinases all reduced P2X7R-stimulated actin reorganization as well as the blebbing [44, 46, 47]. Deletion of the C terminus of P2X7R completely abrogated its interaction with epithelia membrane proteins (EMPs)/tetraspanins in engineered HEK293 cells. This was correlated with a marked attenuation of blebbing suggesting that P2X7R–EMP interaction is crucial for induction of rapid blebbing [45]. A recent analysis by Dixon and colleagues of P2X7R-dependent membrane blebbing in osteoblasts found that extracellular ATP activated phospholipases D (PLD) and A2 leading to production and release of lysophosphatidic acid (LPA) [48]. Antagonism or desensitization of G-protein-coupled LPA receptors in these osteoblasts suppressed blebbing in response to either LPA or P2X7R agonists but did not affect the ionotropic actions of P2X7R. LPA triggered a rapid Rho kinase-dependent membrane blebbing in osteoblasts from wild-type or P2X7R knockout mice while ATP induced blebbing only in the wild-type osteoblasts. Thus, LPA functions as an autocrine mediator downstream of P2X7R to induce signaling cascades that control osteoblast membrane blebbing in response to ATP. These authors speculate that this autocrine loop may be involved in P2X7R-stimulated osteogenesis during skeletal development and mechanotransduction. Although previous studies have verified the ability of P2X7R to stimulate PLD and PLA2 activity via both Ca2+-dependent and Ca2+-independent signaling cascades in several cell types, the study by the Dixon group provides a novel link of these latter P2X7R-initiated responses to the dynamic regulation of the actin cytoskeleton which is critical for many integrative leukocyte functions, such as transient cell–cell contact during homing or antigen presentation, direct cell–cell fusion during granuloma formation, and phagocytosis of pathogens or host debris during infection or inflammation. Notably, activation of P2X7R has also been linked to several of these responses including membrane fusion events such as formation of multinucleated giant cells and the killing of intracellular mycobacteria [49, 50].

P2X7R regulation of cell–cell fusion and formation of multinucleate giant cells

Formation of multinucleated giant cells (MGC) has been used as a hallmark of chronic inflammatory reactions found in foreign body reactions, sterile inflammation, and infective granulomas [51]. Additionally, the fusion of monocyte-related progenitors into the multinucleate osteoclasts is a significant example of physiological cell–cell fusion that is critical for normal bone homeostasis. Several lines of evidence implicate a role for P2X7R in MGC formation in cells of hematopoietic origin including macrophages, monocytes, and osteoclasts, wherein P2X7R is highly expressed [49, 52, 53]. MGCs within granuloma are short-lived cells which begin to die several hours after fusion, and this apoptotic death is followed by cytoplasmic fragmentation. The ability of cells grown to confluence in culture to spontaneously form MGCs has been correlated with the expression level of P2X7R [49]. This is consistent with observations in human macrophages wherein the fusion triggered by Con A and interferon-γ can be inhibited by the P2X7R antagonist, oxidized ATP. However, expression of P2X7R per se is not obligatory for this process because mock-transfected HEK293 cells and osteoclasts derived from P2X7R-deficient mice still retain the capacity to form MGC, albeit to a lesser degree [5356]. Notably, the C-terminal domain of P2X7R appears to be required for MGC formation based on the fusion indices of HEK293 cells engineered to express full-length versus C-terminally truncated P2X7R. This raises the issue as to which candidate proteins might interact with P2X7R to regulate this dramatic change of membrane fluidity and cytoskeletal rearrangements. It is unclear whether P2X7R-mediated cell–cell fusion is an outcome or a cause of pathologic cell–cell interactions in disease states. Likewise, more studies are needed to assess whether cell–cell fusion shares common or parallel signaling pathways observed for other P2X7R-dependent membrane trafficking responses such as PS flip, microvesicle shedding, and membrane blebbing.

P2X7R-mediated regulation of intracellular membrane traffic and organelle function

P2X7R regulation of phagosome maturation and microbial killing

Several pathogenic microbes, including mycobacteria and Chlamydia, are able to survive in host macrophages by subverting the normal bacteriocidal processes that sequentially involve microbe internalization within phagosomes, maturation of these phagosomes, and efficient fusion of the mature phagosomes with lysosomes to result in bacterial killing and clearance. P2X7R activation has been shown to promote the killing of both mycobacteria [50, 57, 58] and Chlamydia [59, 60] in two ways: enhanced maturation of the microbe-containing phagosomes and enhanced killing of the microbe-infected host cells. The first mechanism involves a P2X7R-mediated activation of phospholipase D that facilitates maturation and fusion of the mycobacteria/chlamydia-containing phagosomes with lysosomes [50]. As noted previously, P2X7R-dependent activation of phospholipase D and A2 has been described in several cell types including macrophages [48, 58, 6163]. Although this P2X7R→PLD pathway clearly contributes to the phagosomal maturation and microbiocidal effects stimulated by extracellular ATP, additional purinergic receptors are involved given the differential microbiocidal actions of ATP versus BzATP [58]. Moreover, a P2X7R-independent but ATP-dependent killing of intracellular Mycobacterium tuberculosis is supported by studies of mycobacterial killing in macrophages from P2X7R knockout mice [64]. The particular contributions of, and possible crosstalk between, the P2X7R-dependent and P2X7R-independent components of ATP-dependent microbial killing are important areas for additional investigation. Recent studies by Kusner and colleagues [6567] have demonstrated that—in addition to PLD—there are critical signaling roles for increased Ca2+, calmodulin-dependent protein kinases, and sphingosine kinase, in the maturation of microbe-loaded phagosomes. Given the ability of various P2Y family receptors to regulate Ca2+ and lipid signaling, it seems likely that P2X7 and various P2Y receptors act cooperatively to mediate the actions of extracellular ATP on killing of internalized mycobacteria or chlamydia. Such an interaction between P2X7R and another P2 receptor subtype(s) could explain the disparate associations of particular P2X7R polymorphisms with altered progression of tubercular phenotypes within different subgroups of human populations or mouse models [64, 6875].

The second pathway by which P2X7R activation can attenuate mycobacterial survival is through direct killing of the infected macrophages [57, 76]. This acts to reduce the intracellular niches wherein these microbial pathogens survive and eventually proliferate. Previous studies have has demonstrated that M. tuberculosis can inhibit apoptosis of infected macrophages by preventing TNF-α-mediated autocrine signaling pathways [77]. However, treatment of such infected cells with ATP or other P2X7R agonists can overcome this blockade in cell death signaling. It remains to be established whether this P2X7R-induced killing of mycobacteria-infected macrophages reflects apoptosis, necrosis, pyroptosis, or a combination of these mechanistically distinct cell death pathways.

P2X7R-induced transfer of endosomal contents to the cytosolic compartment

One of the best characterized physiological responses to P2X7R stimulation is the maturation and secretion of the proinflammatory cytokine IL-1β [78]. This requires the activation of the cytokine processing enzyme, caspase-1. ATP and other host-derived small molecules, such as uric acid crystals [79], have been identified as “danger signals” [80] that act to amplify certain phases of the innate immune or inflammatory responses entrained by pathogen-derived molecular patterns (PAMPs) such as flagellin, bacterial and viral RNA, CpG-DNA, and muramyl dipeptide [8184]. These are best characterized as agonistic ligands for the various Toll-like receptors (TLR) that are intrinsic membrane proteins localized to the plasma membrane or endosomal membranes. PAMP binding to TLRs triggers NFκB- and MAPK-based signaling cascades that culminate in transcriptional activation of many proinflammatory genes, including the precursor form of IL-1β. However, certain PAMPs also act as direct ligands or indirect regulators for the soluble, cytosolic nucleotide oligomerization domain (NOD) family receptors. Some NOD proteins, including NALP1, NALP3/cyropyrin, and IPAF, function as central adapter molecules for the assembly of the cytosolic “inflammasome” protein complexes that facilitate the rapid proteolytic activation of caspase-1 required for maturation of IL-1β. The efficient assembly of certain caspase-1 inflammasomes, particularly those involving NALP1 and NALP3, requires both coordinated delivery of the cognate PAMPs or PAMP metabolites into the cytosol and presence of additional endogenous signals generated in response to the host danger molecules, such as extracellular ATP or uric acid. A rapid decrease in cytosolic K+ concentration appears to be one such endogenous signal [8587].

Recent reports from several labs suggest that P2X7R may be involved in both the delivery of PAMPs to the cytosol and the generation of the endogenous signal(s) required for rapid inflammasome assembly. Pelegrin and Surprenant [88, 89] first demonstrated that pannexin-1, a gap junction-like protein, can functionally interact with the P2X7R to elicit the secondary changes in plasma membrane permeabilization (as measured by organic dye influx) that typify P2X7R activation. Significantly, treatment of macrophages with pannexin-1 inhibitors also strongly attenuated the P2X7R-induced activation of caspase-1 and IL-1β maturation/secretion. Subsequent studies by Nunez and colleagues [90, 91] indicated that this role for pannexin-1 involves a delivery of PAMPs (LPS or muramyl dipeptide) from endosomal compartments into the cytosol to initiate assembly of Nalp3/cryopyrin inflammasome. These authors propose that extracellular PAMPs are first internalized within recycling endosomes but—in the absence of P2X7R activation—are only slowly released from this compartment into the cytosol. ATP stimulation of P2X7R triggers conformational changes in the pannexin-1 pools of both the plasma membrane and endosomal membranes with consequent efflux of PAMPs from the endosomal lumen into the cytosol. Consistent with this scenario, inhibition of endosomal maturation by chloroquine completely abrogated ATP-induced caspase-1 activation in macrophages.

P2X7R-induced changes in mitochondrial permeability and function

A growing body of data indicates that P2X7R activation not only regulates permeability of the plasma membrane but also the permeability/integrity of intracellular membrane compartments. In addition to endosomes, Adinolfi et al. have shown that overexpression of P2X7R—even in the absence of maximal activation by exogenous ATP—in HEK293 cells hyperpolarizes mitochondrial membrane potential, increases basal intramitochondrial Ca2+, increases total cellular ATP content, and confers an ability to grow in the absence of serum [92]. In contrast, maximal activation of these overexpressed P2X7R by millimolar extracellular ATP triggered a rapid depolarization of the mitochondria which was associated with a very large increase in mitochondrial Ca2+ accumulation, mitochondrial fragmentation, and cell death. These studies indicated that low-level autocrine activation of the overexpressed P2X7R by released endogenous ATP was responsible for the increased mitochondrial potential and growth advantage, while the massive influx of extracellular Ca2+ induced by maximally activated receptors triggered the collapse of mitochondrial integrity and reduced viability. Similar studies by Mackenzie et al. [7] noted that the massive collapse of mitochondrial ion homeostasis (leading to cell death) induced by sustained P2X7R activation was dependent on increased Rho-dependent kinase activity but was independent of Ca2+. Analysis of natively expressed P2X7R in rat submandibular gland cells revealed that ATP-induced mitochondrial depolarization was additionally dependent on the influx of extracellular Na+ that accompanies P2X7R channel gating [93]. Although P2X7R has traditionally been associated with the early phases of inflammation, cytotoxicity, and proapoptotic induction, the additional ability of low-level P2X7R activation to increase mitochondrial energy metabolism and actually confer growth advantage may be germane to recently identified roles for altered P2X7 expression/function in several cancers [9497]. The apparent “double-edged sword” role of P2X7R in the regulation of mitochondrial integrity and function may underlie its seemingly paradoxical actions as a trigger of cell death in some contexts but an inducer of cell survival and growth in other settings.

P2X7R-induced exocytosis of secretory lysosomes

As noted in the previous section, activation of P2X7R can trigger the rapid release or secretion of several proinflammatory cytokines compartmentalized within microvesicles that originate from evaginated blebs of plasma membrane which scission away from the cell body. However, multiple lines of data indicate that P2X7R can also stimulate the release of various intracellular macromolecules via the more classical exocytotic fusion of intracellular storage granules or organelles with the plasma membrane. Indeed, the studies of Gomperts and colleagues [98101] on the ability of extracellular ATP to trigger degranulation of mast cells were among the earliest examples of “P2z” (now P2X7) receptor action on secretory cell types. Similarly, activation of the P2X7R in rat submandibular gland cells elicits a rapid release of granules containing proteases such as kallikrein [102]. Another example of P2X7R-stimulated exocytosis is the fusion of the so-called secretory lysosomes with the plasma membrane triggered by extracellular ATP which occurs concurrently with the release of cytokine IL-1β and its converting enzyme caspase-1 in murine macrophages [40] and human monocytes [39]. Secretory lysosomes represent a specialized subtype of lysosomes that predominantly are expressed in hematopoietic cells [103]. These include the histamine- and serotonin-containing dense granules of mast cells, the perforin- and granzyme-containing granules of cytolytic T lymphocytes, and the major basic protein granules of eosinophils. Additionally, macrophages and dendritic cells can release lysosomes that contain many of the proteases (e.g., cathepsins) and other degradative enzymes that are classically considered as lysosomal marker proteins. Thus, the exocytotic mobilization of these various secretory lysosomes/granules results in the export of critical mediators of diverse immune and inflammatory responses. As for other secretory granules that are released via regulated exocytosis, the fusion of the various secretory lysosomes with the plasma membrane is regulated by characteristic t- and v-SNARE proteins and Ca2+-dependent synaptotagmins [104106].

Overlap between the P2X7R-triggered signaling pathways that regulate lysosome exocytosis and those required for IL-1β secretion has suggested that these lysosomes may comprise a mechanism for non-classical IL-1β secretion [107]. In human monocytes, both events can be blocked by inhibitors that target Ca2+-dependent and Ca2+-independent phospholipase A2 and phosphatidylcholine-specific phospholipase C [37]. Perturbation of microtubule-directed movements of secretory lysosomes also attenuates the ATP-triggered export of IL-1β [39, 107]. Moreover, studies performed in P2X7R knockout murine macrophages suggest that IP3-dependent Ca2+ mobilization and protein kinase C activation mediated by G-protein-coupled P2Ys are not sufficient for either secretory lysosome exocytosis or IL-1β release [40]. This latter report also indicated that both the P2X7R-dependent mobilization of secretory lysosomes and the secretion of IL-1β are strongly potentiated by the LPS–NFκB signaling pathway and requires de novo protein synthesis. Despite these multiple correlations between IL-1β release and exocytosis of secretory lysosomes, removal of extracellular Ca2+ completely abrogates ATP-induced lysosome exocytosis from murine macrophages without affecting IL-1β secretion, which unequivocally dissociates lysosome exocytosis from the non-classical IL-1β secretion machinery regulated by P2X7R [40]. Additionally, immunocytochemical analyses of intact murine macrophages indicate no significant redistribution of cytosolic IL-1β into secretory lysosomes during P2X7R activation [108]. The wide range of proteases, phospholipases, and heat shock proteins contained within the secretory lysosomes [109, 110] of macrophages and other immune effector cells may be critical for additional regulatory roles of P2X7R in extracellular matrix remodeling and release of other bioactive mediators.

P2X7R-induced release of multivesicular body (MVB)-derived exosomes

In addition to plasma membrane-derived microvesicles, immune and inflammatory cells can release another pool of membrane-delimited vesicles termed exosomes [111, 112]. These are derived from the intraluminal vesicles (ILV) contained within the MVB generated by invagination of the limit membranes of recycling endosomes. These ILVs are often enriched in plasma membrane proteins, such as ligand-occupied receptors, which are internalized on the endosomes. While many MVBs fuse with lysosomes to deliver the ILVs and their component proteins for degradation/down-regulation, some MVBs can be redirected for fusion with the plasma membranes to release the ILVs into extracellular compartment as exosomes. Significantly, exosomes released from macrophages, dendritic cells, or B lymphocytes contain intrinsic membrane proteins, such as the type II major histocompatibility complex (MHCII) that play critical roles in immune recognition and antigen presentation [113, 114]. When released from leukocytes that have internalized and processed microbial or other foreign proteins, exosomes will contain MHCII loaded with antigenic peptide (pMHCII exosomes) [115]. A growing literature indicates that these released pMHCII exosomes can activate remote T lymphocytes either by direct antigen presentation from the pMHCII displayed on the exosome surface, or an indirect pathway wherein the pMHCII exosomes are taken up by remote naive dendritic cells for DC-mediated antigen presentation [116118]. In addition to this model of “remote” antigen presentation, released exosomes may be used for other novel types of intercellular communication including the cell-to-cell transfer of: (1) intact mRNAs and microRNAs [119]; and (2) the direct transfer of oncogenic growth factor receptors from tumor cells to remote non-transformed cells [120].

We have recently reported observations that support an involvement of exosomes as yet another non-classical pathway for the P2X7R-stimulated secretion of IL-1β [40]. These data suggest that both plasma membrane-derived microvesicles and MVB-derived exosomes can be used to “encapsulate” small volumes of cytosol containing caspase-1 inflammasome complexes and their procytokine substrates. In other studies (Qu et al., unpublished data), we have found that P2X7R activation in murine macrophages and dendritic cells triggers the rapid extracellular release of two pools of membranes containing MHCII. In macrophages primed with both interferon-γ and LPS or DC primed with LPS, extracellular ATP stimulated the export of ∼15% of the total cellular MHCII pool within 90 min. The released MHCII was present within two morphologically and biochemically distinct populations of membrane vesicles: (1) plasma membrane-derived microvesicles (100–500 nm diameter) that sediment at 10,000×g and also contain P2X7R protein, actin, and the LAMP1 lysosomal membrane protein; and (2) multivesicular body-derived exosomes (30–80 nm diameter) that sediment at 100,000×g and lack the P2X7R, actin, and LAMP1 markers. Significantly, both pools of released, purified MHCII membranes were capable of binding antigenic peptide and activating T-cell-receptor-dependent IL-2 production in antigen-specific T-cell hybridoma cells. These observations link the well-characterized actions of the P2X7 receptor on innate immune responses within highly localized regions of microbial infection to a possible non-juxtacrine intercellular communication pathway for delivery of microbial antigen to T cells, with consequent long-range engagement of the adaptive immune response.

Footnotes

This work was supported by NIH grants R01-GM36387 and P01-HLHL18708 (G.R.D.).

References

  • 1.Fadok VA, Bratton DL, Rose DM, Pearson A, Ezekewitz RA, Henson PM (2000) A receptor for phosphatidylserine-specific clearance of apoptotic cells. Nature 405:85–90 [DOI] [PubMed]
  • 2.Bevers EM, Comfurius P, Zwaal RF (1983) Changes in membrane phospholipid distribution during platelet activation. Biochim Biophys Acta 736:57–66 [DOI] [PubMed]
  • 3.Comfurius P, Zwaal RF (1977) The enzymatic synthesis of phosphatidylserine and purification by CM-cellulose column chromatography. Biochim Biophys Acta 488:36–42 [DOI] [PubMed]
  • 4.Bevers EM, Comfurius P, Dekkers DW, Zwaal RF (1999) Lipid translocation across the plasma membrane of mammalian cells. Biochim Biophys Acta 1439:317–330 [DOI] [PubMed]
  • 5.Balasubramanian K, Schroit AJ (2003) Aminophospholipid asymmetry: a matter of life and death. Annu Rev Physiol 65:701–734 [DOI] [PubMed]
  • 6.Zwaal RF, Schroit AJ (1997) Pathophysiologic implications of membrane phospholipid asymmetry in blood cells. Blood 89:1121–1132 [PubMed]
  • 7.Mackenzie AB, Young MT, Adinolfi E, Surprenant A (2005) Pseudoapoptosis induced by brief activation of ATP-gated P2X7 receptors. J Biol Chem 280:33968–33976 [DOI] [PubMed]
  • 8.van den Eijnde SM, van den Hoff MJ, Reutelingsperger CP, van Heerde WL, Henfling ME, Vermeij-Keers C, Schutte B, Borgers M, Ramaekers FC (2001) Transient expression of phosphatidylserine at cell–cell contact areas is required for myotube formation. J Cell Sci 114:3631–3642 [DOI] [PubMed]
  • 9.de Vries KJ, Wiedmer T, Sims PJ, Gadella BM (2003) Caspase-independent exposure of aminophospholipids and tyrosine phosphorylation in bicarbonate responsive human sperm cells. Biol Reprod 68:2122–2134 [DOI] [PubMed]
  • 10.Frasch SC, Henson PM, Nagaosa K, Fessler MB, Borregaard N, Bratton DL (2004) Phospholipid flip–flop and phospholipid scramblase 1 (PLSCR1) co-localize to uropod rafts in formylated Met-Leu-Phe-stimulated neutrophils. J Biol Chem 279:17625–17633 [DOI] [PubMed]
  • 11.Gardai SJ, McPhillips KA, Frasch SC, Janssen WJ, Starefeldt A, Murphy-Ullrich JE, Bratton DL, Oldenborg PA, Michalak M, Henson PM (2005) Cell-surface calreticulin initiates clearance of viable or apoptotic cells through trans-activation of LRP on the phagocyte. Cell 123:321–334 [DOI] [PubMed]
  • 12.Fadok VA, Bratton DL, Frasch SC, Warner ML, Henson PM (1998) The role of phosphatidylserine in recognition of apoptotic cells by phagocytes. Cell Death Differ 5:551–562 [DOI] [PubMed]
  • 13.Marguet D, Luciani MF, Moynault A, Williamson P, Chimini G (1999) Engulfment of apoptotic cells involves the redistribution of membrane phosphatidylserine on phagocyte and prey. Nat Cell Biol 1:454–456 [DOI] [PubMed]
  • 14.Hamon Y, Broccardo C, Chambenoit O, Luciani MF, Toti F, Chaslin S, Freyssinet JM, Devaux PF, McNeish J, Marguet D, Chimini G (2000) ABC1 promotes engulfment of apoptotic cells and transbilayer redistribution of phosphatidylserine. Nat Cell Biol 2:399–406 [DOI] [PubMed]
  • 15.Elliott JI, Surprenant A, Marelli-Berg FM, Cooper JC, Cassady-Cain RL, Wooding C, Linton K, Alexander DR, Higgins CF (2005) Membrane phosphatidylserine distribution as a non-apoptotic signalling mechanism in lymphocytes. Nat Cell Biol 7:808–816 [DOI] [PubMed]
  • 16.Manodori AB, Barabino GA, Lubin BH, Kuypers FA (2000) Adherence of phosphatidylserine-exposing erythrocytes to endothelial matrix thrombospondin. Blood 95:1293–1300 [PubMed]
  • 17.Galkina E, Tanousis K, Preece G, Tolaini M, Kioussis D, Florey O, Haskard DO, Tedder TF, Ager A (2003) l-selectin shedding does not regulate constitutive T cell trafficking but controls the migration pathways of antigen-activated T lymphocytes. J Exp Med 198:1323–1335 [DOI] [PMC free article] [PubMed]
  • 18.Kishimoto TK, Jutila MA, Berg EL, Butcher EC (1989) Neutrophil Mac-1 and MEL-14 adhesion proteins inversely regulated by chemotactic factors. Science 245:1238–1241 [DOI] [PubMed]
  • 19.Jamieson GP, Snook MB, Thurlow PJ, Wiley JS (1996) Extracellular ATP causes of loss of l-selectin from human lymphocytes via occupancy of P2Z purinocepters. J Cell Physiol 166:637–642 [DOI] [PubMed]
  • 20.Gu B, Bendall LJ, Wiley JS (1998) Adenosine triphosphate-induced shedding of CD23 and l-selectin (CD62L) from lymphocytes is mediated by the same receptor but different metalloproteases. Blood 92:946–951 [PubMed]
  • 21.Labasi JM, Petrushova N, Donovan C, McCurdy S, Lira P, Payette MM, Brissette W, Wicks JR, Audoly L, Gabel CA (2002) Absence of the P2X7 receptor alters leukocyte function and attenuates an inflammatory response. J Immunol 168:6436–6445 [DOI] [PubMed]
  • 22.Sluyter R, Wiley JS (2002) Extracellular adenosine 5′-triphosphate induces a loss of CD23 from human dendritic cells via activation of P2X7 receptors. Int Immunol 14:1415–1421 [DOI] [PubMed]
  • 23.Chen JR, Gu BJ, Dao LP, Bradley CJ, Mulligan SP, Wiley JS (1999) Transendothelial migration of lymphocytes in chronic lymphocytic leukaemia is impaired and involved down-regulation of both l-selectin and CD23. Br J Haematol 105:181–189 [PubMed]
  • 24.Moon H, Na HY, Chong KH, Kim TJ (2006) P2X7 receptor-dependent ATP-induced shedding of CD27 in mouse lymphocytes. Immunol Lett 102:98–105 [DOI] [PubMed]
  • 25.VanWijk MJ, VanBavel E, Sturk A, Nieuwland R (2003) Microparticles in cardiovascular diseases. Cardiovasc Res 59:277–287 [DOI] [PubMed]
  • 26.Hess C, Sadallah S, Hefti A, Landmann R, Schifferli JA (1999) Ectosomes released by human neutrophils are specialized functional units. J Immunol 163:4564–4573 [PubMed]
  • 27.Eken C, Gasser O, Zenhaeusern G, Oehri I, Hess C, Schifferli JA (2008) Polymorphonuclear neutrophil-derived ectosomes interfere with the maturation of monocyte-derived dendritic cells. J Immunol 180:817–824 [DOI] [PubMed]
  • 28.Barry OP, Pratico D, Savani RC, FitzGerald GA (1998) Modulation of monocyte–endothelial cell interactions by platelet microparticles. J Clin Invest 102:136–144 [DOI] [PMC free article] [PubMed]
  • 29.Heijnen HF, Schiel AE, Fijnheer R, Geuze HJ, Sixma JJ (1999) Activated platelets release two types of membrane vesicles: microvesicles by surface shedding and exosomes derived from exocytosis of multivesicular bodies and alpha-granules. Blood 94:3791–3799 [PubMed]
  • 30.George JN, Thoi LL, McManus LM, Reimann TA (1982) Isolation of human platelet membrane microparticles from plasma and serum. Blood 60:834–840 [PubMed]
  • 31.Zwaal RF, Comfurius P, Bevers EM (2004) Scott syndrome, a bleeding disorder caused by defective scrambling of membrane phospholipids. Biochim Biophys Acta 1636:119–128 [DOI] [PubMed]
  • 32.Gasser O, Schifferli JA (2004) Activated polymorphonuclear neutrophils disseminate anti-inflammatory microparticles by ectocytosis. Blood 104:2543–2548 [DOI] [PubMed]
  • 33.Greco V, Hannus M, Eaton S (2001) Argosomes: a potential vehicle for the spread of morphogens through epithelia. Cell 106:633–645 [DOI] [PubMed]
  • 34.Pizzirani C, Ferrari D, Chiozzi P, Adinolfi E, Sandona D, Savaglio E, Di Virgilio F (2007) Stimulation of P2 receptors causes release of IL-1beta-loaded microvesicles from human dendritic cells. Blood 109:3856–3864 [DOI] [PubMed]
  • 35.Bianco F, Pravettoni E, Colombo A, Schenk U, Moller T, Matteoli M, Verderio C (2005) Astrocyte-derived ATP induces vesicle shedding and IL-1{beta} release from microglia. J Immunol 174:7268–7277 [DOI] [PubMed]
  • 36.MacKenzie A, Wilson HL, Kiss-Toth E, Dower SK, North RA, Surprenant A (2001) Rapid secretion of interleukin-1beta by microvesicle shedding. Immunity 15:825–835 [DOI] [PubMed]
  • 37.Andrei C, Margiocco P, Poggi A, Lotti LV, Torrisi MR, Rubartelli A (2004) Phospholipases C and A2 control lysosome-mediated IL-1 beta secretion: implications for inflammatory processes. Proc Natl Acad Sci U S A 101:9745–9750 [DOI] [PMC free article] [PubMed]
  • 38.Gudipaty L, Munetz J, Verhoef PA, Dubyak GR (2003) Essential role for Ca2+ in regulation of IL-1beta secretion by P2X7 nucleotide receptor in monocytes, macrophages, and HEK-293 cells. Am J Physiol Cell Physiol 285:C286–C99 [DOI] [PubMed]
  • 39.Carta S, Tassi S, Semino C, Fossati G, Mascagni P, Dinarello CA, Rubartelli A (2006) Histone deacetylase inhibitors prevent exocytosis of interleukin-1beta-containing secretory lysosomes: role of microtubules. Blood 108:1618–1626 [DOI] [PMC free article] [PubMed]
  • 40.Qu Y, Franchi L, Nunez G, Dubyak GR (2007) Nonclassical IL-1 beta secretion stimulated by P2X7 receptors is dependent on inflammasome activation and correlated with exosome release in murine macrophages. J Immunol 179:1913–1925 [DOI] [PubMed]
  • 41.Gardella S, Andrei C, Ferrera D, Lotti LV, Torrisi MR, Bianchi ME, Rubartelli A (2002) The nuclear protein HMGB1 is secreted by monocytes via a non-classical, vesicle-mediated secretory pathway. EMBO Rep 3:995–1001 [DOI] [PMC free article] [PubMed]
  • 42.Angelini G, Gardella S, Ardy M, Ciriolo MR, Filomeni G, Di Trapani G, Clarke F, Sitia R, Rubartelli A (2002) Antigen-presenting dendritic cells provide the reducing extracellular microenvironment required for T lymphocyte activation. Proc Natl Acad Sci U S A 99:1491–1496 [DOI] [PMC free article] [PubMed]
  • 43.Robinson RA, Worfolk L, Tracy PB (1992) Endotoxin enhances the expression of monocyte prothrombinase activity. Blood 79:406–416 [PubMed]
  • 44.Morelli A, Chiozzi P, Chiesa A, Ferrari D, Sanz JM, Falzoni S, Pinton P, Rizzuto R, Olson MF, Di Virgilio F (2003) Extracellular ATP causes ROCK I-dependent bleb formation in P2X7-transfected HEK293 cells. Mol Biol Cell 14:2655–2664 [DOI] [PMC free article] [PubMed]
  • 45.Wilson HL, Wilson SA, Surprenant A, North RA (2002) Epithelial membrane proteins induce membrane blebbing and interact with the P2X7 receptor C terminus. J Biol Chem 277:34017–34023 [DOI] [PubMed]
  • 46.Pfeiffer ZA, Aga M, Prabhu U, Watters JJ, Hall DJ, Bertics PJ (2004) The nucleotide receptor P2X7 mediates actin reorganization and membrane blebbing in RAW 264.7 macrophages via p38 MAP kinase and Rho. J Leukoc Biol 75:1173–1182 [DOI] [PubMed]
  • 47.Verhoef PA, Estacion M, Schilling W, Dubyak GR (2003) P2X7 receptor-dependent blebbing and the activation of Rho-effector kinases, caspases, and IL-1 beta release. J Immunol 170:5728–5738 [DOI] [PubMed]
  • 48.Panupinthu N, Zhao L, Possmayer F, Ke HZ, Sims SM, Dixon SJ (2007) P2X7 nucleotide receptors mediate blebbing in osteoblasts through a pathway involving lysophosphatidic acid. J Biol Chem 282:3403–3412 [DOI] [PubMed]
  • 49.Chiozzi P, Sanz JM, Ferrari D, Falzoni S, Aleotti A, Buell GN, Collo G, Di Virgilio F (1997) Spontaneous cell fusion in macrophage cultures expressing high levels of the P2Z/P2X7 receptor. J Cell Biol 138:697–706 [DOI] [PMC free article] [PubMed]
  • 50.Fairbairn IP, Stober CB, Kumararatne DS, Lammas DA (2001) ATP-mediated killing of intracellular mycobacteria by macrophages is a P2X(7)-dependent process inducing bacterial death by phagosome–lysosome fusion. J Immunol 167:3300–3307 [DOI] [PubMed]
  • 51.Adams DO (1976) The granulomatous inflammatory response. A review. Am J Pathol 84:164–192 [PMC free article] [PubMed]
  • 52.Falzoni S, Chiozzi P, Ferrari D, Buell G, Di Virgilio F (2000) P2X(7) receptor and polykarion formation. Mol Biol Cell 11:3169–3176 [DOI] [PMC free article] [PubMed]
  • 53.Lemaire I, Falzoni S, Leduc N, Zhang B, Pellegatti P, Adinolfi E, Chiozzi P, Di Virgilio F (2006) Involvement of the purinergic P2X7 receptor in the formation of multinucleated giant cells. J Immunol 177:7257–7265 [DOI] [PubMed]
  • 54.Ke HZ, Qi H, Weidema AF, Zhang Q, Panupinthu N, Crawford DT, Grasser WA, Paralkar VM, Li M, Audoly LP, Gabel CA, Jee WS, Dixon SJ, Sims SM, Thompson DD (2003) Deletion of the P2X7 nucleotide receptor reveals its regulatory roles in bone formation and resorption. Mol Endocrinol 17:1356–1367 [DOI] [PubMed]
  • 55.Gartland A, Buckley KA, Hipskind RA, Perry MJ, Tobias JH, Buell G, Chessell I, Bowler WB, Gallagher JA (2003) Multinucleated osteoclast formation in vivo and in vitro by P2X7 receptor-deficient mice. Crit Rev Eukaryot Gene Expr 13:243–253 [DOI] [PubMed]
  • 56.Gartland A, Buckley KA, Bowler WB, Gallagher JA (2003) Blockade of the pore-forming P2X7 receptor inhibits formation of multinucleated human osteoclasts in vitro. Calcif Tissue Int 73:361–369 [DOI] [PubMed]
  • 57.Lammas DA, Stober C, Harvey CJ, Kendrick N, Panchalingam S, Kumararatne DS (1997) ATP-induced killing of mycobacteria by human macrophages is mediated by purinergic P2Z(P2X7) receptors. Immunity 7:433–444 [DOI] [PubMed]
  • 58.Kusner DJ, Adams J (2000) ATP-induced killing of virulent Mycobacterium tuberculosis within human macrophages requires phospholipase D. J Immunol 164:379–388 [DOI] [PubMed]
  • 59.Coutinho-Silva R, Stahl L, Raymond MN, Jungas T, Verbeke P, Burnstock G, Darville T, Ojcius DM (2003) Inhibition of chlamydial infectious activity due to P2X7R-dependent phospholipase D activation. Immunity 19:403–412 [DOI] [PubMed]
  • 60.Darville T, Welter-Stahl L, Cruz C, Sater AA, Andrews CW Jr, Ojcius DM (2007) Effect of the purinergic receptor P2X7 on Chlamydia infection in cervical epithelial cells and vaginally infected mice. J Immunol 179:3707–3714 [DOI] [PubMed]
  • 61.Humphreys BD, Dubyak GR (1996) Induction of the P2z/P2X7 nucleotide receptor and associated phospholipase D activity by lipopolysaccharide and IFN-gamma in the human THP-1 monocytic cell line. J Immunol 157:5627–5637 [PubMed]
  • 62.Pochet S, Gomez-Munoz A, Marino A, Dehaye JP (2003) Regulation of phospholipase D by P2X7 receptors in submandibular ductal cells. Cell Signal 15:927–935 [DOI] [PubMed]
  • 63.Fernando KC, Gargett CE, Wiley JS (1999) Activation of the P2Z/P2X7 receptor in human lymphocytes produces a delayed permeability lesion: involvement of phospholipase D. Arch Biochem Biophys 362:197–202 [DOI] [PubMed]
  • 64.Sikora A, Liu J, Brosnan C, Buell G, Chessel I, Bloom BR (1999) Cutting edge: purinergic signaling regulates radical-mediated bacterial killing mechanisms in macrophages through a P2X7-independent mechanism. J Immunol 163:558–561 [PubMed]
  • 65.Malik ZA, Thompson CR, Hashimi S, Porter B, Iyer SS, Kusner DJ (2003) Cutting edge: Mycobacterium tuberculosis blocks Ca2+ signaling and phagosome maturation in human macrophages via specific inhibition of sphingosine kinase. J Immunol 170:2811–2815 [DOI] [PubMed]
  • 66.Thompson CR, Iyer SS, Melrose N, VanOosten R, Johnson K, Pitson SM, Obeid LM, Kusner DJ (2005) Sphingosine kinase 1 (SK1) is recruited to nascent phagosomes in human macrophages: inhibition of SK1 translocation by Mycobacterium tuberculosis. J Immunol 174:3551–3561 [DOI] [PubMed]
  • 67.Connolly SF, Kusner DJ (2007) The regulation of dendritic cell function by calcium-signaling and its inhibition by microbial pathogens. Immunol Res 39:115–127 [DOI] [PubMed]
  • 68.Mancino G, Placido R, Di Virgilio F (2001) P2X7 receptors and apoptosis in tuberculosis infection. J Biol Regul Homeost Agents 15:286–293 [PubMed]
  • 69.Li CM, Campbell SJ, Kumararatne DS, Bellamy R, Ruwende C, McAdam KP, Hill AV, Lammas DA (2002) Association of a polymorphism in the P2X7 gene with tuberculosis in a Gambian population. J Infect Dis 186:1458–1462 [DOI] [PubMed]
  • 70.Myers AJ, Eilertson B, Fulton SA, Flynn JL, Canaday DH (2005) The purinergic P2X7 receptor is not required for control of pulmonary Mycobacterium tuberculosis infection. Infect Immun 73:3192–3195 [DOI] [PMC free article] [PubMed]
  • 71.Fernando SL, Saunders BM, Sluyter R, Skarratt KK, Goldberg H, Marks GB, Wiley JS, Britton WJ (2007) A polymorphism in the P2X7 gene increases susceptibility to extrapulmonary tuberculosis. Am J Respir Crit Care Med 175:360–366 [DOI] [PubMed]
  • 72.Franco-Martinez S, Nino-Moreno P, Bernal-Silva S, Baranda L, Rocha-Meza M, Portales-Cervantes L, Layseca-Espinosa E, Gonzalez-Amaro R, Portales-Perez D (2006) Expression and function of the purinergic receptor P2X7 in patients with pulmonary tuberculosis. Clin Exp Immunol 146:253–261 [DOI] [PMC free article] [PubMed]
  • 73.Britton WJ, Fernando SL, Saunders BM, Sluyter R, Wiley JS (2007) The genetic control of susceptibility to Mycobacterium tuberculosis. Novartis Found Symp 281:79–89 discussion 89–92, 208–9 [DOI] [PubMed]
  • 74.Nino-Moreno P, Portales-Perez D, Hernandez-Castro B, Portales-Cervantes L, Flores-Meraz V, Baranda L, Gomez-Gomez A, Acuna-Alonzo V, Granados J, Gonzalez-Amaro R (2007) P2X7 and NRAMP1/SLC11 A1 gene polymorphisms in Mexican mestizo patients with pulmonary tuberculosis. Clin Exp Immunol 148:469–477 [DOI] [PMC free article] [PubMed]
  • 75.Mokrousov I, Sapozhnikova N, Narvskaya O (2008) Mycobacterium tuberculosis co-existence with humans: making an imprint on the macrophage P2X7 receptor gene? J Med Microbiol 57:581–584 [DOI] [PubMed]
  • 76.Lu H, Shen C, Brunham RC (2000) Chlamydia trachomatis infection of epithelial cells induces the activation of caspase-1 and release of mature IL-18. J Immunol 165:1463–1469 [DOI] [PubMed]
  • 77.Balcewicz-Sablinska MK, Keane J, Kornfeld H, Remold HG (1998) Pathogenic Mycobacterium tuberculosis evades apoptosis of host macrophages by release of TNF-R2, resulting in inactivation of TNF-alpha. J Immunol 161:2636–2641 [PubMed]
  • 78.Di Virgilio F (2007) Liaisons dangereuses: P2X(7) and the inflammasome. Trends Pharmacol Sci 28:465–472 [DOI] [PubMed]
  • 79.Martinon F, Petrilli V, Mayor A, Tardivel A, Tschopp J (2006) Gout-associated uric acid crystals activate the NALP3 inflammasome. Nature 440:237–241 [DOI] [PubMed]
  • 80.Martinon F (2008) Detection of immune danger signals by NALP3. J Leukoc Biol 83:507–511 [DOI] [PubMed]
  • 81.Mariathasan S, Monack DM (2007) Inflammasome adaptors and sensors: intracellular regulators of infection and inflammation. Nat Rev Immunol 7:31–40 [DOI] [PubMed]
  • 82.Martinon F (2007) Orchestration of pathogen recognition by inflammasome diversity: variations on a common theme. Eur J Immunol 37:3003–3006 [DOI] [PubMed]
  • 83.McDermott MF, Tschopp J (2007) From inflammasomes to fevers, crystals and hypertension: how basic research explains inflammatory diseases. Trends Mol Med 13:381–388 [DOI] [PubMed]
  • 84.Ye Z, Ting JP (2008) NLR, the nucleotide-binding domain leucine-rich repeat containing gene family. Curr Opin Immunol 20:3–9 [DOI] [PubMed]
  • 85.Franchi L, Kanneganti TD, Dubyak GR, Nunez G (2007) Differential requirement of P2X7 receptor and intracellular K+ for caspase-1 activation induced by intracellular and extracellular bacteria. J Biol Chem 282:18810–18818 [DOI] [PubMed]
  • 86.Mariathasan S, Weiss DS, Newton K, McBride J, O’Rourke K, Roose-Girma M, Lee WP, Weinrauch Y, Monack DM, Dixit VM (2006) Cryopyrin activates the inflammasome in response to toxins and ATP. Nature 440:228–232 [DOI] [PubMed]
  • 87.Kahlenberg JM, Dubyak GR (2004) Mechanisms of caspase-1 activation by P2X7 receptor-mediated K+ release. Am J Physiol Cell Physiol 286:C1100–C1108 [DOI] [PubMed]
  • 88.Pelegrin P, Surprenant A (2006) Pannexin-1 mediates large pore formation and interleukin-1beta release by the ATP-gated P2X7 receptor. EMBO J 25:5071–5082 [DOI] [PMC free article] [PubMed]
  • 89.Pelegrin P, Surprenant A (2007) Pannexin-1 couples to maitotoxin- and nigericin-induced interleukin-1beta release through a dye uptake-independent pathway. J Biol Chem 282:2386–2394 [DOI] [PubMed]
  • 90.Marina-Garcia N, Franchi L, Kim YG, Miller D, McDonald C, Boons GJ, Nunez G (2008) Pannexin-1-mediated intracellular delivery of muramyl dipeptide induces caspase-1 activation via Cryopyrin/NLRP3 independently of Nod2. J Immunol 180:4050–4057 [DOI] [PubMed]
  • 91.Kanneganti TD, Lamkanfi M, Kim YG, Chen G, Park JH, Franchi L, Vandenabeele P, Nunez G (2007) Pannexin-1-mediated recognition of bacterial molecules activates the cryopyrin inflammasome independent of Toll-like receptor signaling. Immunity 26:433–443 [DOI] [PubMed]
  • 92.Adinolfi E, Callegari MG, Ferrari D, Bolognesi C, Minelli M, Wieckowski MR, Pinton P, Rizzuto R, Di Virgilio F (2005) Basal activation of the P2X7 ATP receptor elevates mitochondrial calcium and potential, increases cellular ATP levels, and promotes serum-independent growth. Mol Biol Cell 16(7):3260–3272 [DOI] [PMC free article] [PubMed]
  • 93.Garcia-Marcos M, Fontanils U, Aguirre A, Pochet S, Dehaye JP, Marino A (2005) Role of sodium in mitochondrial membrane depolarization induced by P2X7 receptor activation in submandibular glands. FEBS Lett 579:5407–5413 [DOI] [PubMed]
  • 94.Adinolfi E, Melchiorri L, Falzoni S, Chiozzi P, Morelli A, Tieghi A, Cuneo A, Castoldi G, Di Virgilio F, Baricordi OR (2002) P2X7 receptor expression in evolutive and indolent forms of chronic B lymphocytic leukemia. Blood 99:706–708 [DOI] [PubMed]
  • 95.Greig AV, Linge C, Healy V, Lim P, Clayton E, Rustin MH, McGrouther DA, Burnstock G (2003) Expression of purinergic receptors in non-melanoma skin cancers and their functional roles in A431 cells. J Invest Dermatol 121:315–327 [DOI] [PubMed]
  • 96.Slater M, Scolyer RA, Gidley-Baird A, Thompson JF, Barden JA (2003) Increased expression of apoptotic markers in melanoma. Melanoma Res 13:137–145 [DOI] [PubMed]
  • 97.Slater M, Danieletto S, Gidley-Baird A, Teh LC, Barden JA (2004) Early prostate cancer detected using expression of non-functional cytolytic P2X7 receptors. Histopathology 44:206–215 [DOI] [PubMed]
  • 98.Cockcroft S, Gomperts BD (1979) ATP induces nucleotide permeability in rat mast cells. Nature 279:541–542 [DOI] [PubMed]
  • 99.Cockcroft S, Gomperts BD (1979) Activation and inhibition of calcium-dependent histamine secretion by ATP ions applied to rat mast cells. J Physiol 296:229–243 [DOI] [PMC free article] [PubMed]
  • 100.Cockcroft S, Gomperts BD (1980) The ATP4- receptor of rat mast cells. Biochem J 188:789–798 [DOI] [PMC free article] [PubMed]
  • 101.Tatham PE, Cusack NJ, Gomperts BD (1988) Characterisation of the ATP4- receptor that mediates permeabilisation of rat mast cells. Eur J Pharmacol 147:13–21 [DOI] [PubMed]
  • 102.Alzola E, Perez-Etxebarria A, Kabre E, Fogarty DJ, Metioui M, Chaib N, Macarulla JM, Matute C, Dehaye JP, Marino A (1998) Activation by P2X7 agonists of two phospholipases A2 (PLA2) in ductal cells of rat submandibular gland. Coupling of the calcium-independent PLA2 with kallikrein secretion. J Biol Chem 273:30208–30217 [DOI] [PubMed]
  • 103.Blott EJ, Griffiths GM (2002) Secretory lysosomes. Nat Rev Mol Cell Biol 3:122–131 [DOI] [PubMed]
  • 104.Andrews NW (2000) Regulated secretion of conventional lysosomes. Trends Cell Biol 10:316–321 [DOI] [PubMed]
  • 105.Holt OJ, Gallo F, Griffiths GM (2006) Regulating secretory lysosomes. J Biochem (Tokyo) 140:7–12 [DOI] [PubMed]
  • 106.Fowler KT, Andrews NW, Huleatt JW (2007) Expression and function of synaptotagmin VII in CTLs. J Immunol 178:1498–1504 [DOI] [PMC free article] [PubMed]
  • 107.Andrei C, Dazzi C, Lotti L, Torrisi MR, Chimini G, Rubartelli A (1999) The secretory route of the leaderless protein interleukin 1beta involves exocytosis of endolysosome-related vesicles. Mol Biol Cell 10:1463–1475 [DOI] [PMC free article] [PubMed]
  • 108.Brough D, Rothwell NJ (2007) Caspase-1-dependent processing of pro-interleukin-1beta is cytosolic and precedes cell death. J Cell Sci 120:772–781 [DOI] [PubMed]
  • 109.Clark R, Griffiths GM (2003) Lytic granules, secretory lysosomes and disease. Curr Opin Immunol 15:516–521 [DOI] [PubMed]
  • 110.Griffiths G (2002) What’s special about secretory lysosomes? Semin Cell Dev Biol 13:279–284 [DOI] [PubMed]
  • 111.Thery C, Zitvogel L, Amigorena S (2002) Exosomes: composition, biogenesis and function. Nat Rev Immunol 2:569–579 [DOI] [PubMed]
  • 112.Johnstone RM (2006) Exosomes biological significance: a concise review. Blood Cells Mol Dis 36:315–321 [DOI] [PubMed]
  • 113.Raposo G, Nijman HW, Stoorvogel W, Liejendekker R, Harding CV, Melief CJ, Geuze HJ (1996) B lymphocytes secrete antigen-presenting vesicles. J Exp Med 183:1161–1172 [DOI] [PMC free article] [PubMed]
  • 114.Thery C, Boussac M, Veron P, Ricciardi-Castagnoli P, Raposo G, Garin J, Amigorena S (2001) Proteomic analysis of dendritic cell-derived exosomes: a secreted subcellular compartment distinct from apoptotic vesicles. J Immunol 166:7309–7318 [DOI] [PubMed]
  • 115.Muntasell A, Berger AC, Roche PA (2007) T cell-induced secretion of MHC class II-peptide complexes on B cell exosomes. Embo J 26:4263–4272 [DOI] [PMC free article] [PubMed]
  • 116.Thery C, Duban L, Segura E, Veron P, Lantz O, Amigorena S (2002) Indirect activation of naive CD4+ T cells by dendritic cell-derived exosomes. Nat Immunol 3:1156–1162 [DOI] [PubMed]
  • 117.Quah BJ, O’Neill HC (2005) The immunogenicity of dendritic cell-derived exosomes. Blood Cells Mol Dis 35:94–110 [DOI] [PubMed]
  • 118.Li XB, Zhang ZR, Schluesener HJ, Xu SQ (2006) Role of exosomes in immune regulation. J Cell Mol Med 10:364–375 [DOI] [PMC free article] [PubMed]
  • 119.Valadi H, Ekstrom K, Bossios A, Sjostrand M, Lee JJ, Lotvall JO (2007) Exosome-mediated transfer of mRNAs and microRNAs is a novel mechanism of genetic exchange between cells. Nat Cell Biol 9:654–659 [DOI] [PubMed]
  • 120.Sanderson MP, Keller S, Alonso A, Riedle S, Dempsey PJ, Altevogt P (2008) Generation of novel, secreted epidermal growth factor receptor (EGFR/ErbB1) isoforms via metalloprotease-dependent ectodomain shedding and exosome secretion. J Cell Biochem 103:1783–1797 [DOI] [PMC free article] [PubMed]

Articles from Purinergic Signalling are provided here courtesy of Springer

RESOURCES