Next Article in Journal
Experimental Infection of Mid-Gestation Pregnant Female and Intact Male Sheep with Zika Virus
Next Article in Special Issue
Structural and Functional Characterization of the Secondary Mutation N126K Selected by Various HIV-1 Fusion Inhibitors
Previous Article in Journal
Immunization of Cats against Fel d 1 Results in Reduced Allergic Symptoms of Owners
Previous Article in Special Issue
Quantitative FRET-FLIM-BlaM to Assess the Extent of HIV-1 Fusion in Live Cells
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Purinergic Receptors: Elucidating the Role of these Immune Mediators in HIV-1 Fusion

Division of Infectious Diseases, Department of Medicine, Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
*
Author to whom correspondence should be addressed.
Viruses 2020, 12(3), 290; https://doi.org/10.3390/v12030290
Submission received: 24 January 2020 / Revised: 4 March 2020 / Accepted: 5 March 2020 / Published: 7 March 2020
(This article belongs to the Special Issue Mechanisms of Viral Fusion and Applications in Antivirals)

Abstract

:
Purinergic receptors are inflammatory mediators activated by extracellular nucleotides released by dying or injured cells. Several studies have described an important role for these receptors in HIV-1 entry, particularly regarding their activity on HIV-1 viral membrane fusion. Several reports identify purinergic receptor antagonists that inhibit HIV-1 membrane fusion; these drugs are suspected to act through antagonizing Env-chemokine receptor interactions. They also appear to abrogate activity of downstream mediators that potentiate activation of the NLRP3 inflammasome pathway. Here we review the literature on purinergic receptors, the drugs that inhibit their function, and the evidence implicating these receptors in HIV-1 entry.

1. Introduction

1.1. HIV and Inflammation

HIV-1 infection is a major cause of morbidity and mortality worldwide, with 37 million individuals living with the disease and 2 million new infections occurring annually [1]. Improved antiretroviral therapy (ART) has significantly increased the life expectancy of people with HIV-1 (PWH). Still, PWH experience chronic inflammation that leads to co-morbidities including cancer, bone disease, and cardiovascular disease [2,3,4,5,6,7,8,9,10,11,12]. The source of this chronic inflammation is multifactorial in nature and includes cell-mediated effects, overproduction of soluble cytokines, coinfections, and immune dysregulation. Increased cell senescence is a primary component of rapid aging and is characterized by CD8+ T lymphocyte accumulation and activation [12,13] and secretion of inflammatory mediators that induce chronic low-level inflammation and heightened cell turnover [12,14,15,16]. Desai, et al., characterized multiple progressive sequelae as contributors to HIV-induced accelerated aging: HIV-1 replication in activated immune cells, GI mucosal epithelium depletion with loss of Th17 cells, microbial translocation, thymic dysfunction with loss of naïve T lymphocytes and regulatory CD4+ T lymphocytes, and clonal expansion of activated immune cells. This clonal expansion results in the loss of CD28 on T lymphocytes and expedited telomere shortening, ultimately producing nonfunctional, senescent T lymphocytes [10,13,17,18,19,20]. Early infection can also result in fibrosis and dysfunction of lymphoid organs and co-infection with pathogens such as cytomegalovirus (CMV) [21]. Immune exhaustion contributes to inflammation, as HIV-induced PD-1 overexpression on CD4+ and CD8+ T lymphocytes is not fully normalized by ART [16]. The viral reservoir marks an important mechanism by which inflammation can persist; in lymphoid tissue, HIV-1 productive infection in a small percentage of permissive cells can result in abortive infection of bystander cells (>95% CD4+ T lymphocyte population). Bystander cells can undergo pyroptosis, a caspase-1-mediated programmed cell death. Pyroptosis could re-activate more latently-infected cells and attract more uninfected cells to die and sustain chronic inflammation [22,23,24,25,26,27,28]. Finally, soluble biomarkers including d-dimer, hsCRP and IL-6, soluble CD14, and sTNFR II have been associated with comorbidities in large patient cohorts [7,11,29,30,31,32,33,34,35]. These elevated inflammatory biomarkers occur despite ART and do not improve even with treatment intensification [36,37].

1.2. Purinergic Receptors

Purinergic receptors have been recognized since the 1970s as mediating an important mechanism of immune function and cell signaling [38,39]. They are widely expressed on mammalian cells. These receptors can detect extracellular nucleotides to activate intracellular signaling events. The receptors are divided into three categories: P1 adenosine receptors, P2X heterotrimeric ATP-gated cation channels, and P2Y G-protein coupled receptors [40,41]. There is a rich literature on the role of the P2X receptor as an important regulator of inflammation and other physiological signaling events [42,43,44,45,46]. The P2X receptors are ATP-gated cation channels. An ATP or other nucleotide agonist binds to the extracellular region of the P2X receptor, inducing a conformational change that opens the receptor central pore, facilitating cation flux [40,47]. Some receptor subtypes have larger pore openings that enable large molecules to pass through [48,49,50]. P2X receptors can undergo rapid desensitization with prolonged ATP exposure, resulting in closure of the channel [51,52]. The subunits of functional P2X receptors can assemble into either homotrimers or heterotrimers; each subunit contains two transmembrane domains, a large extracellular loop with 10 conserved cysteine residues and glycosylation sites, and intracellular N and C termini with consensus phosphorylation sites [41,43,53,54,55]. The P2X7 receptor subtype (P2RX7) is the best characterized in the innate immune response and its role has been well described in inflammatory cytokine signaling, antigen presentation, and lymphocyte proliferation and differentiation [56,57,58]. P2RX7 is activated by sub-millimolar concentrations of extracellular ATP. Physiologically, this ATP is transiently released in the bloodstream in response to acute cell death or injury [40]. P2RX7 has a large pore when assembled as a homotrimer; sustained P2RX7 activation can result in pore opening, enabling passage of molecules up to 900 Da that can induce cell death [43,59]. P2X1 receptors (P2RX1) are expressed at low levels on myeloid cells and lesser so on lymphocytes; fewer reports, therefore, have characterized this receptor with respect to inflammation [60,61,62,63,64,65,66].
P2Y receptors have broad functions across a range of physiological systems including blood clotting, hormonal secretion, vascular responses, cell migration, wound healing, and immune responses [38,39,46,49,67,68,69]. The P2Y receptor subtypes are G protein-coupled receptors that consist of seven transmembrane domains with an extracellular N-terminus and an intracellular C-terminus [67,70,71]. P2Y receptor activation results in G-protein dissociation of α and βγ subunits that induce downstream effector signaling. The P2Y class of receptors consists of a large diversity of sequences that impact different pharmacologic functions [72]. There are eight monomer subtypes. Of these, four (P2Y1, P2Y2, P2Y4, and P2Y6) activate phospholipase C via Gq, three (P2Y12, P2Y13, and P2Y14) inhibit adenylyl cyclase and activate GIRK-family K+ channels through Gi, and one (P2Y11) triggers intracellular Ca2+ and in cAMP levels through both Gq and Gs.

1.3. Purinergic Receptors and HIV-1

Purinergic signaling has been demonstrated to be key to the immune response in several infectious processes [73,74,75], including bacterial [76,77,78,79,80], protozoal [81,82,83,84], and viral infections including respiratory viral infections [85,86], hepatitides [63,87,88,89], cytomegalovirus [90], and HIV-1 [91,92,93,94,95,96]. Interestingly, purinergic receptors have been implicated in both immunoprotective and immunopathologic roles in these infection [75,88,97,98]. There is a growing body of evidence that implicates purinergic receptors and both selective and non-selective antagonists of these receptors in HIV-1 infection [99,100,101,102,103,104,105]. Our laboratory and others have explored the role of purinergic receptors in HIV-1 infection and have reported that purinergic receptor antagonists can block HIV-1 infection and reduce inflammatory cytokine production associated with infection [96,106,107]. More specific probing appears to implicate drugs that target purinergic receptors in the inhibition of HIV-1 viral membrane fusion. This will be the subject of the following section.
The first studies that suggested a role for purinergic receptors in HIV-1 pathogenesis took place in the 1980s when suramin was proposed as an anti-retroviral agent and was given to 98 patients with AIDS following in vitro observations of suramin inhibiting reverse transcriptase activity [108]. Suramin was quickly abandoned as a treatment, however, this was due to drug toxicity that resulted in numerous patient fatalities. The fatalities took place in individuals with a high burden of disease and, therefore, suramin’s impact on HIV-1 replication was impossible to assess. Recent investigations focus on the roles of purinergic receptors in HIV-1 pathogenesis and the activities of compounds distinct from suramin.
Since that time, there have been a number of studies that have uncovered roles for purinergic receptors in HIV-1 pathogenesis [91,92,93,94,95,96]. Adenosine receptors have been implicated based on data suggesting an association between CD39 expression and AIDS progression [109]. CD39, an ectoenzyme, generates adenosine that induces purinergic adenosine receptor signaling. The authors observed that inhibition of regulatory T lymphocytes was reduced by CD39 downregulation, resulting in associated elevated levels of T-cell A2A adenosine receptor in HIV-1 infected patients. Additionally, the authors noted that a CD39 polymorphism was associated with reduced CD39 expression and a delay in onset of AIDS.
Sorrell, et al., probed the role of extracellular ATP signaling and observed that treatment with a non-selective P2X antagonist modulated opiate neurotoxicity through HIV-1 Tat [110]. Tovar, et al., observed that HIV-infected macrophages released ATP that reduced dendritic spine density through glutamate receptor downmodulation that was dependent on purinergic receptor signaling [111]. More recently, growing literature has supported the notion of purinergic receptors and their antagonists directly impacting HIV-1 viral replication. Séror, et al., demonstrated that HIV-1 infection induces ATP release in human lymphocytes [95] and that the selective depletion of P2Y2 diminished the HIV-induced inflammatory response. Immunofluorescence indicated that P2Y2 and the ATP channel Pannexin-1 appeared to accumulate at the virological synapse, where an infected donor cell can mediate efficient cell-to-cell transfer to an uninfected cell [112,113].
Hazleton, et al., demonstrated that purinergic receptors play a key role in HIV-1 replication in macrophages through selective pharmacologic inhibition of P2RX1, P2RX7, and P2RY1 [93]. Giroud, et al. described a role for P2RX1 in blocking HIV-1 binding to the chemokine receptors CCR5 and CXCR4 [106]. Swartz, et al., demonstrated that non-selective antagonists of P2X receptors can inhibit HIV-1 infection of CD4+ lymphocytes through both cell-to-cell and cell-free mechanisms [96]. Orellana et al., reported that both R5- and X4-tropic HIV-1 infection activates the pannexin-1 ATP channel; this enabled HIV-1 internalization in CD4+ T cells [94]. Graziano, et al., demonstrated a P2RX7-dependent phenomenon whereby exposure to extracellular ATP induced rapid release of HIV-1 particles in human monocyte-derived [114]. Numerous studies describe the role of purinergic receptors and their antagonists at various stages of the viral life cycle and downstream impacts on pathogenesis. In the following section, we will review the literature as it pertains to the role of purinergic receptors and their antagonists on HIV-1 fusion.

1.4. Purinergic Receptors and HIV-1 Fusion

While a broad range of purinergic receptor antagonists have been demonstrated to inhibit HIV-1 infection [99,100,101,102,103,104,105], only P2RX1-specific compounds have been implicated in HIV-1 membrane fusion. Marin, et al. developed and employed a high-throughput BlaM-Vpr assay that identifies virus-cell membrane fusion via cleavage of a cytoplasmic β-lactamase-fused reporter substrate subsequent to membrane fusion of a β-lactamase-fused virus [115]. A screen of 1280 compounds identified the P2 × 1-antagonizing compounds NF279 and NF449 as inhibitors of virus-cell fusion in the TZM-bl cell line. NF449 and NF279 were further reported as fusion inhibitors in primary monocyte-derived macrophage (MDM), CEM-A, Jurkat, and NP2 cells as measured by the BlaM-Vpr assay [93,106]. Our laboratory developed a novel HIV-1 Cre-lox assay to measure cell-to-cell mediated virus-membrane fusion [96,116,117]. In this assay, transduced Jurkat cells express a reporter cassette containing a lox-P flanked dsRed reporter followed by a Cre-inducible GFP site. Transfected donor Jurkat cells produce HIV-1 that is derived from NL4-3 and packages the Cre Recombinase enzyme (HIV-1 Gag-iCre); HIV-1 Gag-iCre viral fusion with target Jurkat floxRG donor cells releases Cre into the target cell cytoplasm, thereby inducing a red-to-green switch in fluorescence [116,117]. Using this assay, we identified the P2RX1-selective compounds NF279, PPNDS, and NF023 as potent inhibitors of cell-to-cell mediated virus-membrane fusion. The broad-spectrum P2XR antagonist PPADS exhibited partial inhibition of virus-membrane fusion [96].
P2RX7 and P2YR antagonists have been shown to inhibit HIV-1 infection at potencies similar to those of P2RX1 antagonists [93,95,96,107,117,118], but these compounds do not appear to impact virus-cell membrane fusion. The P2RX7 antagonist A740003, which inhibits HIV-1 infection [93], fails to inhibit virus-membrane fusion as measured by BlaM-Vpr assays [93,107,115]. The same assay demonstrated that the broad-spectrum P2 receptor inhibitor PPADS likewise has no effect on virus-membrane fusion, and the Jurkat floxRG assay detected minimal inhibition of fusion inhibition resultant of PPADS treatment [96,115].
Select antagonists of the P2Y class of purinergic receptors have similarly inhibited HIV-1 productive infection in various assays [85,95,105], but there have been no reports of these compounds directly affecting virus-cell fusion. Swartz, et al., using the Jurkat floxRG assay, conducted a screen with a library of 71 purinergic antagonists and found no evidence of fusion inhibition induced by any of the P2YR antagonists [96].
The P2Y11R antagonist NF340, an inhibitor of HIV-1 infection, was assessed as a fusion inhibitor via BlaM-Vpr assays. The compound demonstrated minimal activity as an inhibitor of HXB2, BaL26, and R3A Env-expressing HIV-1 entry in CEM-A or TZM-bl cells, and this minimal effect was seen only at high concentrations [106,115]. The BlaM-Vpr assay was further used to study the P2RY1 antagonist MRS2500 and the P2RY2 antagonist PSB 1114; neither MRS2500 nor PSB 1114 inhibited virus-cell fusion in TZM-bl cells [115].
Giroud et al., employed a temperature-arrested stage (TAS) fusion assay in which prolonged virus-cell incubation at a reduced temperature minimizes the binding efficiency of the Env-coreceptor complex but maintains relative permissivity to CD4-Env binding [106]. The resultant high ratio of Env-CD4 complexes to Env-coreceptor complexes renders subsequent raised-temperature-induced viral fusion more sensitive to inhibition via coreceptor antagonists than to inhibition via CD4 antagonists. When assessed as a TAS fusion inhibitor, NF279 exhibited inhibitory potency that mirrors coreceptor antagonist activity; this potency is distinct from those of CD4 antagonists. A TAS drug-substitution assay revealed that unlike a CD4 antagonist, NF279 fails to inhibit viral fusion to TZM-bl cells at the CD4 binding step. NF279 instead was shown to be interchangeable with coreceptor antagonists in inhibiting fusion. Complicating the hypothesis that NF279 functions as a fusion inhibitor downstream of CD4 binding, employment of this assay with primary MDMs revealed a partial antagonizing activity of NF279 against the Env-CD4 complex in these cells.
A calcium influx assay in TZM-bl cells was used to elucidate the relationship between NF279 and CCR5/CXCR4. Following stimulation with a coreceptor agonist or gp120, NF279 inhibited subsequent cellular calcium influx characteristic of virus-coreceptor binding in a manner similar to that of coreceptor antagonists. This observation, however, was also inconsistent across cell types as NF279 only partially inhibited CXCR4-mediated calcium influx in CEM-A cells. NF279 was also found to inhibit calcium influx induced by a CXCR3/CXCR7 agonist in these cells, highlighting the possibility that NF279 nonspecifically binds to chemokine receptors.
The activity of NF279 was further compared to those of coreceptor antagonists via a BlaM-Vpr assay that assessed BaL26 viral fusion with TZM-bl and JGR.H11 cells expressing a CCR5 mutant (G163R) with reduced binding efficiency for gp120. NF279 as well as the dual CCR5 and CC2b antagonist TAK-779 inhibited virus-cell fusion in both of the CCR5 mutant cell lines; NF279 exhibited ~3-fold increased inhibitory potency against the mutant cell lines relative to wild-type cells. This observation was distinct from CCR5 antagonists Schering C and AD101, both of which failed to inhibit virus-cell fusion in the CCR5 mutant cells. The increased sensitivity of the CCR5 mutants to NF279-mediated fusion inhibition suggests possible competition between NF279 and CCR5 for gp120 binding.
Furthermore, the P2RX1-selective compounds NF279 and NF449 have been shown to inhibit HIV-1 cellular entry with greater activity against X4-tropic virus (HIV-1 HXB2) than with R5- or dual-tropic virus (HIV-1 BaL26 and HIV-1 R3A, respectively) [106,115]. The observation that viral tropism modulates the inhibitory potency of NF449 and NF279 against viral fusion indicates that Env and/or coreceptor mediate purinergic antagonist-dependent fusion inhibition.
Our laboratory additionally demonstrated that NF449 and NF279-mediated fusion inhibition relies on intact gp41 and displays varied activity against diverse clades. The inhibitory activity of NF279 and NF449 appeared to interfere specifically with Env. This was observed as NF279 and NF449 inhibited viral membrane fusion and interfered with PG9 antibody neutralization. PG9 targets the V1V2 region of HIV-1 gp120 [118]. These observations suggested that NF279 and NF449 might act on the V1V2 region, either directly or indirectly.

2. Discussion

Despite the efficacy of ART in suppressing HIV-1 replication and maintaining CD4+ T lymphocyte counts, PWH experiences numerous comorbidities resultant of HIV-induced chronic inflammation. These inflammatory comorbidities are a prominent concern in HIV-1 treatment as they are an impediment to quality-of-life among PWH and are the leading cause of early mortality among ART-treated PWH.
The immunomodulatory purinergic receptors have been implicated in the immune response against multiple pathogens, and numerous antagonists of these receptors have been shown to abrogate HIV-1 infection in vitro at varying potencies. Select studies have reported reduced expression and/or function of purinergic signaling pathway components as being associated with relatively better outcomes for PWH, but these data are limited and their scope minimal. In assessing the hypothesized clinical applications of P2XR antagonists, our laboratory demonstrated P2XR antagonist-mediated inhibition of both HIV-1 infection and HIV-induced inflammatory cytokine production. These data suggest that P2XR antagonists may represent a novel class of dual-target therapeutics.
Regarding the mechanism of these drugs, the fact that the immunomodulatory purinergic receptors are expressed extracellularly indicates that abrogation of HIV-induced inflammation occurs before HIV-1 cellular entry. Comprehensive screens including multiple classes of purinergic receptor antagonists have identified P2RX1 antagonists, specifically, as inhibitors of cell-virus fusion. P2RX1 antagonists have been demonstrated to inhibit virus-cell fusion in both primary cells and multiple cell lines. This antagonism has been demonstrated through multiple methodologies including the described BlaM-Vpr, Jurkat floxRG, and temperature-arrested state (TAS) fusion assays. Antagonists of the P2RX7 and P2YR receptors, in contrast, were reported as exhibiting minimal or absent activity in inhibiting virus-cell fusion.
Although fusion antagonism appears specific to P2RX1-specific antagonists, these suramin-derived compounds lose receptor subtype specificity at the micromolar concentrations used in these studies [106,107]. Some antagonists have previously been shown to inhibit post-fusion targets, such as the original observation of anti-reverse transcriptase activity by suramin. The documented nonspecific binding of these compounds to multiple diverse targets, paired with the observation that fusion inhibition is only seen with P2RX1 antagonists, renders it unlikely that the inhibition of HIV-1 fusion and infection by these compounds is exclusively resultant of purinergic receptor antagonism.
Because of the low binding specificity of these antagonists at the concentrations required to inhibit HIV-1 infection, it is difficult to assess the precise mechanism by which they inhibit. Numerous studies have, however, identified Env-coreceptor interactions as a likely target of P2RX1 antagonist-mediated fusion inhibition. NF279 has greater inhibitory potency against X4-tropic virus relative to R5-tropic viruses, suggesting that inhibitory activity is dependent on Env and/or coreceptor structure. A TAS assay employed by Giroud, et al. demonstrated that NF279 inhibited fusion at a stage following virus-CD4 attachment and that the kinetics of this inhibition resembled those of coreceptor antagonists [106]. NF279 was additionally shown to inhibit coreceptor agonist-induced calcium influx in a manner similar to coreceptor antagonists.
These observations, however, were inconsistent across cell lines: NF279 was found to only partially inhibit CXCR4-associated calcium influx in CEM-A cells; in this study, NF279 also exhibited inhibitory activity against other chemokine receptor agonists. NF279 was further shown to partially inhibit CD4 attachment in CEM-A cells. These incongruous findings could represent nonspecific binding of NF279 to chemokine receptors, but they may also be resultant of NF279 antagonism of non-coreceptor ion channels such as the various purinergic receptors.
Further probing the mechanism by which the P2RX1 antagonists may inhibit coreceptor engagement, Giroud et al. reported that NF279 exhibits increased inhibitory potency when virus-coreceptor affinity is reduced by a CCR5 mutation, suggesting competition between NF279 and Env for coreceptor binding [106]. Similarly, our group has shown that NF449 inhibits PG9 antibody binding to the V1V2 region of gp120, indicating drug-coreceptor binding competition. Further implicating gp120 in NF449-mediated fusion inhibition, we have reported that NF449 inhibitory activity is dependent on intact gp41. Fusion inhibition is not, however, dependent on gp41 itself, suggesting that gp41-induced structural changes in the conformation of gp120 interfere with NF449-mediated fusion inhibition. Our group has proposed a mechanism in which NF449 either directly binds to the V1V2 region of gp120, blocking coreceptor engagement, or instead induces a conformational change in the V1V2 loop that inhibits coreceptor binding.
Interestingly, NF279 has been widely reported as a more potent inhibitor of HIV-1 infection and membrane fusion than NF449, but NF449 was demonstrated to more potently inhibit PG9-Env binding. This may represent differential binding affinity of the two antagonists to Env and/or additional inhibitory off-target effects of NF279, such as the coreceptor binding suggested by Giroud et al. [106].
Taken as a whole, we suspect that purinergic receptors themselves are not primary mediators of HIV-1 membrane fusion. P2X antagonism via pharmacotherapy, however, likely plays a role in the abrogation of the HIV-induced inflammatory response. It is possible, therefore, that drug-Env binding is the primary mediator of fusion inhibition, while concurrent inhibition of P2X receptors abrogates the HIV-induced cellular inflammatory response. Ultimately, studies investigating the mechanism by which the P2RX1 antagonists inhibit virus-cell fusion and virus-induced inflammatory response are inconclusive due to the non-specificity of these compounds, varied activity among different antagonists, and inconsistent results between cell types.
Based on the collective literature documenting P2RX1 antagonist activity in inhibiting virus-cell fusion, further investigation into the activities and mechanisms of these antagonists is needed. This line of research is particularly relevant to elucidating HIV-1 pathogenesis, as the mechanism by which HIV-1 induces chronic inflammation and its consequential comorbidities remains undefined. Identifying the target of these drugs may provide needed insight into the identification of novel therapeutics that dually mitigate both HIV-1 infection and its resultant chronic inflammation. Developing such dual-target drugs is of critical importance in the treatment of PWH as effective modulation of the HIV-induced immune response is required to enhance the quality-of-life and normalize the lifespan of PWH.

Funding

This research was funded by T. Swartz from NIH, NIAID K08-AI120806 and by the Schneider-Lesser Foundation. The funders had no role in the design of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript, or in the decision to publish the results.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Yoshimura, K. Current status of HIV/AIDS in the ART era. J. Infect. Chemother. 2017, 23, 12–16. [Google Scholar] [CrossRef] [PubMed]
  2. Spitsin, S.; Tebas, P.; Barrett, J.S.; Pappa, V.; Kim, D.; Taylor, D.; Evans, D.L.; Douglas, S.D. Antiinflammatory effects of aprepitant coadministration with cART regimen containing ritonavir in HIV-infected adults. Jci Insight 2017, 2. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Schouten, J.; Wit, F.W.; Stolte, I.G.; Kootstra, N.A.; van der Valk, M.; Geerlings, S.E.; Prins, M.; Reiss, P.; Group, A.G.C.S. Cross-sectional comparison of the prevalence of age-associated comorbidities and their risk factors between HIV-infected and uninfected individuals: The AGEhIV cohort study. Clin. Infect. Dis. 2014, 59, 1787–1797. [Google Scholar] [CrossRef] [PubMed]
  4. O'Brien, M.P.; Hunt, P.W.; Kitch, D.W.; Klingman, K.; Stein, J.H.; Funderburg, N.T.; Berger, J.S.; Tebas, P.; Clagett, B.; Moisi, D.; et al. A Randomized Placebo Controlled Trial of Aspirin Effects on Immune Activation in Chronically Human Immunodeficiency Virus-Infected Adults on Virologically Suppressive Antiretroviral Therapy. Open Forum Infect. Dis. 2017, 4, ofw278. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Marchetti, G.; Tincati, C.; Silvestri, G. Microbial translocation in the pathogenesis of HIV infection and AIDS. Clin. Microbiol Rev. 2013, 26, 2–18. [Google Scholar] [CrossRef] [Green Version]
  6. Kearns, A.; Gordon, J.; Burdo, T.H.; Qin, X. HIV-1-Associated Atherosclerosis: Unraveling the Missing Link. J. Am. Coll Cardiol. 2017, 69, 3084–3098. [Google Scholar] [CrossRef]
  7. Kaplan-Lewis, E.; Aberg, J.A.; Lee, M. Aging with HIV in the ART era. Semin. Diagn Pathol. 2017, 34, 384–397. [Google Scholar] [CrossRef]
  8. Guaraldi, G.; Orlando, G.; Zona, S.; Menozzi, M.; Carli, F.; Garlassi, E.; Berti, A.; Rossi, E.; Roverato, A.; Palella, F. Premature age-related comorbidities among HIV-infected persons compared with the general population. Clin. Infect. Dis. 2011, 53, 1120–1126. [Google Scholar] [CrossRef] [Green Version]
  9. Freiberg, M.S.; Chang, C.C.; Kuller, L.H.; Skanderson, M.; Lowy, E.; Kraemer, K.L.; Butt, A.A.; Bidwell Goetz, M.; Leaf, D.; Oursler, K.A.; et al. HIV infection and the risk of acute myocardial infarction. Jama Intern. Med. 2013, 173, 614–622. [Google Scholar] [CrossRef] [PubMed]
  10. Desai, S.; Landay, A. Early immune senescence in HIV disease. Curr. Hiv Aids Rep. 2010, 7, 4–10. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  11. Aberg, J.A. Aging, inflammation, and HIV infection. Top. Antivir. Med. 2012, 20, 101–105. [Google Scholar] [PubMed]
  12. Deeks, S.G. HIV infection, inflammation, immunosenescence, and aging. Annu. Rev. Med. 2011, 62, 141–155. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Hunt, P.W.; Martin, J.N.; Sinclair, E.; Bredt, B.; Hagos, E.; Lampiris, H.; Deeks, S.G. T cell activation is associated with lower CD4+ T cell gains in human immunodeficiency virus-infected patients with sustained viral suppression during antiretroviral therapy. J. Infect. Dis. 2003, 187, 1534–1543. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Lederman, M.M.; Calabrese, L.; Funderburg, N.T.; Clagett, B.; Medvik, K.; Bonilla, H.; Gripshover, B.; Salata, R.A.; Taege, A.; Lisgaris, M.; et al. Immunologic failure despite suppressive antiretroviral therapy is related to activation and turnover of memory CD4 cells. J. Infect. Dis. 2011, 204, 1217–1226. [Google Scholar] [CrossRef] [Green Version]
  15. Massanella, M.; Negredo, E.; Perez-Alvarez, N.; Puig, J.; Ruiz-Hernandez, R.; Bofill, M.; Clotet, B.; Blanco, J. CD4 T-cell hyperactivation and susceptibility to cell death determine poor CD4 T-cell recovery during suppressive HAART. AIDS 2010, 24, 959–968. [Google Scholar] [CrossRef]
  16. Massanella, M.; Fromentin, R.; Chomont, N. Residual inflammation and viral reservoirs: Alliance against an HIV cure. Curr. Opin. Hiv Aids 2016, 11, 234–241. [Google Scholar] [CrossRef] [Green Version]
  17. Hunt, P.W.; Sinclair, E.; Rodriguez, B.; Shive, C.; Clagett, B.; Funderburg, N.; Robinson, J.; Huang, Y.; Epling, L.; Martin, J.N.; et al. Gut epithelial barrier dysfunction and innate immune activation predict mortality in treated HIV infection. J. Infect. Dis. 2014, 210, 1228–1238. [Google Scholar] [CrossRef] [Green Version]
  18. Brenchley, J.M.; Schacker, T.W.; Ruff, L.E.; Price, D.A.; Taylor, J.H.; Beilman, G.J.; Nguyen, P.L.; Khoruts, A.; Larson, M.; Haase, A.T.; et al. CD4+ T cell depletion during all stages of HIV disease occurs predominantly in the gastrointestinal tract. J. Exp. Med. 2004, 200, 749–759. [Google Scholar] [CrossRef] [Green Version]
  19. Brenchley, J.M.; Price, D.A.; Douek, D.C. HIV disease: Fallout from a mucosal catastrophe? Nat. Immunol. 2006, 7, 235–239. [Google Scholar] [CrossRef]
  20. Mehandru, S.; Poles, M.A.; Tenner-Racz, K.; Horowitz, A.; Hurley, A.; Hogan, C.; Boden, D.; Racz, P.; Markowitz, M. Primary HIV-1 infection is associated with preferential depletion of CD4+ T lymphocytes from effector sites in the gastrointestinal tract. J. Exp. Med. 2004, 200, 761–770. [Google Scholar] [CrossRef] [Green Version]
  21. Klatt, N.R.; Chomont, N.; Douek, D.C.; Deeks, S.G. Immune activation and HIV persistence: Implications for curative approaches to HIV infection. Immunol. Rev. 2013, 254, 326–342. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Doitsh, G.; Cavrois, M.; Lassen, K.G.; Zepeda, O.; Yang, Z.; Santiago, M.L.; Hebbeler, A.M.; Greene, W.C. Abortive HIV infection mediates CD4 T cell depletion and inflammation in human lymphoid tissue. Cell 2010, 143, 789–801. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Doitsh, G.; Galloway, N.L.; Geng, X.; Yang, Z.; Monroe, K.M.; Zepeda, O.; Hunt, P.W.; Hatano, H.; Sowinski, S.; Mu√±oz-Arias, I.; et al. Cell death by pyroptosis drives CD4 T-cell depletion in HIV-1 infection. Nature 2014, 505, 509–514. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Doitsh, G.; Greene, W.C. Dissecting How CD4 T Cells Are Lost During HIV Infection. Cell Host Microbe 2016, 19, 280–291. [Google Scholar] [CrossRef] [Green Version]
  25. Doitsh, G.; Galloway, N.L.; Geng, X.; Yang, Z.; Monroe, K.M.; Zepeda, O.; Hunt, P.W.; Hatano, H.; Sowinski, S.; Muñoz-Arias, I.; et al. Corrigendum: Cell death by pyroptosis drives CD4 T-cell depletion in HIV-1 infection. Nature 2017, 544, 124. [Google Scholar] [CrossRef]
  26. Galloway, N.L.; Doitsh, G.; Monroe, K.M.; Yang, Z.; Munoz-Arias, I.; Levy, D.N.; Greene, W.C. Cell-to-Cell Transmission of HIV-1 Is Required to Trigger Pyroptotic Death of Lymphoid-Tissue-Derived CD4 T Cells. Cell Rep. 2015, 12, 1555–1563. [Google Scholar] [CrossRef] [Green Version]
  27. Monroe, K.M.; Yang, Z.; Johnson, J.R.; Geng, X.; Doitsh, G.; Krogan, N.J.; Greene, W.C. IFI16 DNA sensor is required for death of lymphoid CD4 T cells abortively infected with HIV. Science 2014, 343, 428–432. [Google Scholar] [CrossRef] [Green Version]
  28. Mu√±oz-Arias, I.; Doitsh, G.; Yang, Z.; Sowinski, S.; Ruelas, D.; Greene, W.C. Blood-Derived CD4 T Cells Naturally Resist Pyroptosis during Abortive HIV-1 Infection. Cell Host Microbe 2015, 18, 463–470. [Google Scholar] [CrossRef] [Green Version]
  29. El-Sadr, W.M.; Lundgren, J.; Neaton, J.D.; Gordin, F.; Abrams, D.; Arduino, R.C.; Babiker, A.; Burman, W.; Clumeck, N.; Cohen, C.J.; et al. CD4+ count-guided interruption of antiretroviral treatment. N. Engl. J. Med. 2006, 355, 2283–2296. [Google Scholar] [CrossRef] [Green Version]
  30. Neuhaus, J.; Jacobs, D.R., Jr.; Baker, J.V.; Calmy, A.; Duprez, D.; La Rosa, A.; Kuller, L.H.; Pett, S.L.; Ristola, M.; Ross, M.J.; et al. Markers of inflammation, coagulation, and renal function are elevated in adults with HIV infection. J. Infect. Dis. 2010, 201, 1788–1795. [Google Scholar] [CrossRef]
  31. Bild, D.E.; Bluemke, D.A.; Burke, G.L.; Detrano, R.; Diez Roux, A.V.; Folsom, A.R.; Greenland, P.; Jacob, D.R.; Kronmal, R.; Liu, K.; et al. Multi-Ethnic Study of Atherosclerosis: Objectives and design. Am. J. Epidemiol. 2002, 156, 871–881. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Tan, H.Y.; Yong, Y.K.; Shankar, E.M.; Paukovics, G.; Ellegard, R.; Larsson, M.; Kamarulzaman, A.; French, M.A.; Crowe, S.M. Aberrant Inflammasome Activation Characterizes Tuberculosis-Associated Immune Reconstitution Inflammatory Syndrome. J. Immunol. 2016, 196, 4052–4063. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Tenorio, A.R.; Zheng, Y.; Bosch, R.J.; Krishnan, S.; Rodriguez, B.; Hunt, P.W.; Plants, J.; Seth, A.; Wilson, C.C.; Deeks, S.G.; et al. Soluble markers of inflammation and coagulation but not T-cell activation predict non-AIDS-defining morbid events during suppressive antiretroviral treatment. J. Infect. Dis. 2014, 210, 1248–1259. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Cutter, G.R.; Burke, G.L.; Dyer, A.R.; Friedman, G.D.; Hilner, J.E.; Hughes, G.H.; Hulley, S.B.; Jacobs, D.R.; Liu, K.; Manolio, T.A. Cardiovascular risk factors in young adults. The CARDIA baseline monograph. Control. Clin. Trials 1991, 12, 1S–77S. [Google Scholar] [CrossRef]
  35. Leng, S.X.; Margolick, J.B. Understanding frailty, aging, and inflammation in HIV infection. Curr. Hiv/Aids Rep. 2015, 12, 25–32. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Dinoso, J.B.; Kim, S.Y.; Wiegand, A.M.; Palmer, S.E.; Gange, S.J.; Cranmer, L.; O'Shea, A.; Callender, M.; Spivak, A.; Brennan, T.; et al. Treatment intensification does not reduce residual HIV-1 viremia in patients on highly active antiretroviral therapy. Proc. Natl. Acad. Sci. USA 2009, 106, 9403–9408. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  37. Hatano, H.; Hayes, T.L.; Dahl, V.; Sinclair, E.; Lee, T.H.; Hoh, R.; Lampiris, H.; Hunt, P.W.; Palmer, S.; McCune, J.M.; et al. A randomized, controlled trial of raltegravir intensification in antiretroviral-treated, HIV-infected patients with a suboptimal CD4+ T cell response. J. Infect. Dis. 2011, 203, 960–968. [Google Scholar] [CrossRef]
  38. Burnstock, G. Purinergic signalling: Its unpopular beginning, its acceptance and its exciting future. Bioessays: News Rev. Mol. Cell. Dev. Biol. 2012, 34, 218–225. [Google Scholar] [CrossRef]
  39. Burnstock, G. Purinergic signalling: Pathophysiology and therapeutic potential. Keio J. Med. 2013, 62, 63–73. [Google Scholar] [CrossRef] [Green Version]
  40. North, R.A. Molecular physiology of P2X receptors. Physiol. Rev. 2002, 82, 1013–1067. [Google Scholar] [CrossRef]
  41. Burnstock, G.; Knight, G.E. Cellular distribution and functions of P2 receptor subtypes in different systems. Int. Rev. Cytol. 2004, 240, 31–304. [Google Scholar] [CrossRef] [PubMed]
  42. Dubyak, G.R. Signal transduction by P2-purinergic receptors for extracellular ATP. Am. J. Respir. Cell Mol. Biol. 1991, 4, 295–300. [Google Scholar] [CrossRef] [PubMed]
  43. Surprenant, A.; North, R.A. Signaling at purinergic P2X receptors. Annu. Rev. Physiol. 2009, 71, 333–359. [Google Scholar] [CrossRef] [Green Version]
  44. Khakh, B.S.; North, R.A. P2X receptors as cell-surface ATP sensors in health and disease. Nature 2006, 442, 527–532. [Google Scholar] [CrossRef] [PubMed]
  45. Burnstock, G.; Kennedy, C. P2X receptors in health and disease. Adv. Pharmacol. 2011, 61, 333–372. [Google Scholar] [CrossRef] [PubMed]
  46. Burnstock, G. Purinergic signalling and disorders of the central nervous system. Nat. Rev. Drug Discov. 2008, 7, 575–590. [Google Scholar] [CrossRef] [PubMed]
  47. Khakh, B.S. Molecular physiology of P2X receptors and ATP signalling at synapses. Nat. Rev. Neurosci. 2001, 2, 165–174. [Google Scholar] [CrossRef]
  48. Khakh, B.S.; Bao, X.R.; Labarca, C.; Lester, H.A. Neuronal P2X transmitter-gated cation channels change their ion selectivity in seconds. Nat. Neurosci. 1999, 2, 322–330. [Google Scholar] [CrossRef]
  49. Khakh, B.S.; Lester, H.A. Dynamic selectivity filters in ion channels. Neuron 1999, 23, 653–658. [Google Scholar] [CrossRef] [Green Version]
  50. Virginio, C.; MacKenzie, A.; Rassendren, F.A.; North, R.A.; Surprenant, A. Pore dilation of neuronal P2X receptor channels. Nat. Neurosci. 1999, 2, 315–321. [Google Scholar] [CrossRef]
  51. Jarvis, M.F.; Khakh, B.S. ATP-gated P2X cation-channels. Neuropharmacology 2009, 56, 208–215. [Google Scholar] [CrossRef] [PubMed]
  52. Rettinger, J.; Schmalzing, G. Activation and desensitization of the recombinant P2X1 receptor at nanomolar ATP concentrations. J. Gen. Physiol. 2003, 121, 451–461. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Coddou, C.; Yan, Z.; Obsil, T.; Huidobro-Toro, J.P.; Stojilkovic, S.S. Activation and regulation of purinergic P2X receptor channels. Pharmacol. Rev. 2011, 63, 641–683. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Erb, L.; Liao, Z.; Seye, C.I.; Weisman, G.A. P2 receptors: Intracellular signaling. Pflug. Arch. Eur. J. Physiol. 2006, 452, 552–562. [Google Scholar] [CrossRef] [PubMed]
  55. Nicke, A.; Baumert, H.G.; Rettinger, J.; Eichele, A.; Lambrecht, G.; Mutschler, E.; Schmalzing, G. P2X1 and P2X3 receptors form stable trimers: A novel structural motif of ligand-gated ion channels. Embo J. 1998, 17, 3016–3028. [Google Scholar] [CrossRef] [PubMed]
  56. 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]
  57. Schenk, U.; Frascoli, M.; Proietti, M.; Geffers, R.; Traggiai, E.; Buer, J.; Ricordi, C.; Westendorf, A.M.; Grassi, F. ATP inhibits the generation and function of regulatory T cells through the activation of purinergic P2X receptors. Sci. Signal. 2011, 4, ra12. [Google Scholar] [CrossRef]
  58. Junger, W.G. Immune cell regulation by autocrine purinergic signalling. Nat. Rev. Immunol. 2011, 11, 201–212. [Google Scholar] [CrossRef] [Green Version]
  59. Browne, L.E.; Compan, V.; Bragg, L.; North, R.A. P2X7 receptor channels allow direct permeation of nanometer-sized dyes. J. Neurosci. Off. J. Soc. Neurosci. 2013, 33, 3557–3566. [Google Scholar] [CrossRef] [Green Version]
  60. De Ita, M.; Vargas, M.H.; Carbajal, V.; Ortiz-Quintero, B.; López-López, C.; Miranda-Morales, M.; Barajas-López, C.; Montaño, L.M. ATP releases ATP or other nucleotides from human peripheral blood leukocytes through purinergic P2 receptors. Life Sci. 2016, 145, 85–92. [Google Scholar] [CrossRef]
  61. Berchtold, S.; Ogilvie, A.L.; Bogdan, C.; Mühl-Zürbes, P.; Ogilvie, A.; Schuler, G.; Steinkasserer, A. Human monocyte derived dendritic cells express functional P2X and P2Y receptors as well as ecto-nucleotidases. Febs Lett. 1999, 458, 424–428. [Google Scholar] [CrossRef] [Green Version]
  62. Clifford, E.E.; Parker, K.; Humphreys, B.D.; Kertesy, S.B.; Dubyak, G.R. The P2X1 receptor, an adenosine triphosphate-gated cation channel, is expressed in human platelets but not in human blood leukocytes. Blood 1998, 91, 3172–3181. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Ashraf, W.; Manzoor, S.; Ashraf, J.; Ahmed, Q.L.; Khalid, M.; Tariq, M.; Imran, M.; Aziz, H. Transcript analysis of P2X receptors in PBMCs of chronic HCV patients: An insight into antiviral treatment response and HCV-induced pathogenesis. Viral Immunol. 2013, 26, 343–350. [Google Scholar] [CrossRef] [PubMed]
  64. Sluyter, R.; Barden, J.A.; Wiley, J.S. Detection of P2X purinergic receptors on human B lymphocytes. Cell Tissue Res. 2001, 304, 231–236. [Google Scholar] [CrossRef] [Green Version]
  65. Lecut, C.; Frederix, K.; Johnson, D.M.; Deroanne, C.; Thiry, M.; Faccinetto, C.; Marée, R.; Evans, R.J.; Volders, P.G.; Bours, V.; et al. P2X1 ion channels promote neutrophil chemotaxis through Rho kinase activation. J. Immunol. 2009, 183, 2801–2809. [Google Scholar] [CrossRef] [Green Version]
  66. López-López, C.; Jaramillo-Polanco, J.; Portales-Pérez, D.P.; Gómez-Coronado, K.S.; Rodríguez-Meléndez, J.G.; Cortés-García, J.D.; Espinosa-Luna, R.; Montaño, L.M.; Barajas-López, C. Two P2X1 receptor transcripts able to form functional channels are present in most human monocytes. Eur. J. Pharm. 2016, 793, 82–88. [Google Scholar] [CrossRef]
  67. von Kugelgen, I.; Harden, T.K. Molecular pharmacology, physiology, and structure of the P2Y receptors. Adv. Pharmacol. 2011, 61, 373–415. [Google Scholar] [CrossRef]
  68. Wang, Z.X.; Nakayama, T.; Sato, N.; Izumi, Y.; Kasamaki, Y.; Ohta, M.; Soma, M.; Aoi, N.; Matsumoto, K.; Ozawa, Y.; et al. Association of the purinergic receptor P2Y, G-protein coupled, 2 (P2RY2) gene with myocardial infarction in Japanese men. Circ. J. Off. J. Jpn. Circ. Soc. 2009, 73, 2322–2329. [Google Scholar] [CrossRef] [Green Version]
  69. Burnstock, G. Purinergic signaling and vascular cell proliferation and death. Arterioscler. Thromb. Vasc. Biol. 2002, 22, 364–373. [Google Scholar] [CrossRef]
  70. Boarder, M.R.; Weisman, G.A.; Turner, J.T.; Wilkinson, G.F. G protein-coupled P2 purinoceptors: From molecular biology to functional responses. Trends Pharmacol. Sci. 1995, 16, 133–139. [Google Scholar] [CrossRef]
  71. Barnard, E.A. The transmitter-gated channels: A range of receptor types and structures. Trends Pharmacol. Sci. 1996, 17, 305–309. [Google Scholar] [CrossRef]
  72. von Kugelgen, I. Pharmacological profiles of cloned mammalian P2Y-receptor subtypes. Pharmacol. Ther. 2006, 110, 415–432. [Google Scholar] [CrossRef] [PubMed]
  73. Miller, C.M.; Boulter, N.R.; Fuller, S.J.; Zakrzewski, A.M.; Lees, M.P.; Saunders, B.M.; Wiley, J.S.; Smith, N.C. The role of the P2X(7) receptor in infectious diseases. PLoS Pathog. 2011, 7, e1002212. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Morandini, A.C.; Savio, L.E.; Coutinho-Silva, R. The role of P2X7 receptor in infectious inflammatory diseases and the influence of ectonucleotidases. Biomed. J. 2014, 37, 169–177. [Google Scholar] [CrossRef]
  75. Savio, L.E.B.; de Andrade Mello, P.; da Silva, C.G.; Coutinho-Silva, R. The P2X7 Receptor in Inflammatory Diseases: Angel or Demon? Front. Pharm. 2018, 9, 52. [Google Scholar] [CrossRef] [Green Version]
  76. 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. Expression and function of the purinergic receptor P2X7 in patients with pulmonary tuberculosis. Clin. Exp. Immunol. 2006, 146, 253–261. [Google Scholar] [CrossRef]
  77. Wu, G.; Zhao, M.; Gu, X.; Yao, Y.; Liu, H.; Song, Y. The effect of P2X7 receptor 1513 polymorphism on susceptibility to tuberculosis: A meta-analysis. Infect. Genet. Evol. J. Mol. Epidemiol. Evol. Genet. Infect. Dis. 2014, 24, 82–91. [Google Scholar] [CrossRef]
  78. Xiao, J.; Sun, L.; Yan, H.; Jiao, W.; Miao, Q.; Feng, W.; Wu, X.; Gu, Y.; Jiao, A.; Guo, Y.; et al. Metaanalysis of P2X7 gene polymorphisms and tuberculosis susceptibility. Fems Immunol. Med. Microbiol. 2010, 60, 165–170. [Google Scholar] [CrossRef] [Green Version]
  79. Yi, L.; Cheng, D.; Shi, H.; Huo, X.; Zhang, K.; Zhen, G. A meta-analysis of P2X7 gene-762T/C polymorphism and pulmonary tuberculosis susceptibility. PLoS ONE 2014, 9, e96359. [Google Scholar] [CrossRef] [Green Version]
  80. Darville, T.; Welter-Stahl, L.; Cruz, C.; Sater, A.A.; Andrews, C.W., Jr.; Ojcius, D.M. Effect of the purinergic receptor P2X7 on Chlamydia infection in cervical epithelial cells and vaginally infected mice. J. Immunol. 2007, 179, 3707–3714. [Google Scholar] [CrossRef] [Green Version]
  81. Marques-da-Silva, C.; Chaves, M.M.; Chaves, S.P.; Figliuolo, V.R.; Meyer-Fernandes, J.R.; Corte-Real, S.; Lameu, C.; Ulrich, H.; Ojcius, D.M.; Rossi-Bergmann, B.; et al. Infection with Leishmania amazonensis upregulates purinergic receptor expression and induces host-cell susceptibility to UTP-mediated apoptosis. Cell. Microbiol. 2011, 13, 1410–1428. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  82. Marques-da-Silva, C.; Chaves, M.M.; Rodrigues, J.C.; Corte-Real, S.; Coutinho-Silva, R.; Persechini, P.M. Differential modulation of ATP-induced P2X7-associated permeabilities to cations and anions of macrophages by infection with Leishmania amazonensis. PLoS ONE 2011, 6, e25356. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Lees, M.P.; Fuller, S.J.; McLeod, R.; Boulter, N.R.; Miller, C.M.; Zakrzewski, A.M.; Mui, E.J.; Witola, W.H.; Coyne, J.J.; Hargrave, A.C.; et al. P2X7 receptor-mediated killing of an intracellular parasite, Toxoplasma gondii, by human and murine macrophages. J. Immunol. 2010, 184, 7040–7046. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Miller, C.M.; Zakrzewski, A.M.; Ikin, R.J.; Boulter, N.R.; Katrib, M.; Lees, M.P.; Fuller, S.J.; Wiley, J.S.; Smith, N.C. Dysregulation of the inflammatory response to the parasite, Toxoplasma gondii, in P2X7 receptor-deficient mice. Int. J. Parasitol. 2011, 41, 301–308. [Google Scholar] [CrossRef] [PubMed]
  85. Paoletti, A.; Raza, S.Q.; Voisin, L.; Law, F.; Pipoli da Fonseca, J.; Caillet, M.; Kroemer, G.; Perfettini, J.L. Multifaceted roles of purinergic receptors in viral infection. Microbes Infect. Inst. Pasteur 2012, 14, 1278–1283. [Google Scholar] [CrossRef] [PubMed]
  86. Lee, B.H.; Hwang, D.M.; Palaniyar, N.; Grinstein, S.; Philpott, D.J.; Hu, J. Activation of P2X(7) receptor by ATP plays an important role in regulating inflammatory responses during acute viral infection. PLoS ONE 2012, 7, e35812. [Google Scholar] [CrossRef] [Green Version]
  87. Lamas Longarela, O.; Schmidt, T.T.; Schoneweis, K.; Romeo, R.; Wedemeyer, H.; Urban, S.; Schulze, A. Proteoglycans act as cellular hepatitis delta virus attachment receptors. PLoS ONE 2013, 8, e58340. [Google Scholar] [CrossRef] [Green Version]
  88. Taylor, J.M.; Han, Z. Purinergic receptor functionality is necessary for infection of human hepatocytes by hepatitis delta virus and hepatitis B virus. PLoS ONE 2010, 5, e15784. [Google Scholar] [CrossRef] [Green Version]
  89. Manzoor, S.; Idrees, M.; Ashraf, J.; Mehmood, A.; Butt, S.; Fatima, K.; Akbar, H.; Rehaman, I.U.; Qadri, I. Identification of ionotrophic purinergic receptors in Huh-7 cells and their response towards structural proteins of HCV genotype 3a. Virol. J. 2011, 8, 431. [Google Scholar] [CrossRef] [Green Version]
  90. Zandberg, M.; van Son, W.J.; Harmsen, M.C.; Bakker, W.W. Infection of human endothelium in vitro by cytomegalovirus causes enhanced expression of purinergic receptors: A potential virus escape mechanism? Transplantation 2007, 84, 1343–1347. [Google Scholar] [CrossRef]
  91. Pacheco, P.A.; Faria, R.X.; Ferreira, L.G.; Paixao, I.C. Putative roles of purinergic signaling in human immunodeficiency virus-1 infection. Biol. Direct 2014, 9, 21. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  92. Barat, C.; Gilbert, C.; Imbeault, M.; Tremblay, M.J. Extracellular ATP reduces HIV-1 transfer from immature dendritic cells to CD4+ T lymphocytes. Retrovirology 2008, 5, 30. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Hazleton, J.E.; Berman, J.W.; Eugenin, E.A. Purinergic receptors are required for HIV-1 infection of primary human macrophages. J. Immunol. 2012, 188, 4488–4495. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Orellana, J.A.; Velasquez, S.; Williams, D.W.; Saez, J.C.; Berman, J.W.; Eugenin, E.A. Pannexin1 hemichannels are critical for HIV infection of human primary CD4+ T lymphocytes. J. Leukoc. Biol. 2013, 94, 399–407. [Google Scholar] [CrossRef] [Green Version]
  95. Seror, C.; Melki, M.T.; Subra, F.; Raza, S.Q.; Bras, M.; Saidi, H.; Nardacci, R.; Voisin, L.; Paoletti, A.; Law, F.; et al. Extracellular ATP acts on P2Y2 purinergic receptors to facilitate HIV-1 infection. J. Exp. Med. 2011, 208, 1823–1834. [Google Scholar] [CrossRef]
  96. Swartz, T.H.; Esposito, A.M.; Durham, N.D.; Hartmann, B.M.; Chen, B.K. P2X-selective purinergic antagonists are strong inhibitors of HIV-1 fusion during both cell-to-cell and cell-free infection. J. Virol. 2014, 88, 11504–11515. [Google Scholar] [CrossRef] [Green Version]
  97. Rosli, S.; Kirby, F.J.; Lawlor, K.E.; Rainczuk, K.; Drummond, G.R.; Mansell, A.; Tate, M.D. Repurposing drugs targeting the P2X7 receptor to limit hyperinflammation and disease during influenza virus infection. Br. J. Pharm. 2019. [Google Scholar] [CrossRef] [Green Version]
  98. Leyva-Grado, V.H.; Ermler, M.E.; Schotsaert, M.; Gonzalez, M.G.; Gillespie, V.; Lim, J.K.; García-Sastre, A. Contribution of the Purinergic Receptor P2X7 to Development of Lung Immunopathology during Influenza Virus Infection. MBio 2017, 8. [Google Scholar] [CrossRef] [Green Version]
  99. Belete, H.A.; Hubmayr, R.D.; Wang, S.; Singh, R.D. The role of purinergic signaling on deformation induced injury and repair responses of alveolar epithelial cells. PLoS ONE 2011, 6, e27469. [Google Scholar] [CrossRef] [Green Version]
  100. Busillo, J.M.; Azzam, K.M.; Cidlowski, J.A. Glucocorticoids sensitize the innate immune system through regulation of the NLRP3 inflammasome. J. Biol. Chem. 2011, 286, 38703–38713. [Google Scholar] [CrossRef] [Green Version]
  101. Deli, T.; Csernoch, L. Extracellular ATP and cancer: An overview with special reference to P2 purinergic receptors. Pathol Oncol. Res. 2008, 14, 219–231. [Google Scholar] [CrossRef] [PubMed]
  102. Franchi, L.; Kanneganti, T.D.; Dubyak, G.R.; Nunez, G. Differential requirement of P2X7 receptor and intracellular K+ for caspase-1 activation induced by intracellular and extracellular bacteria. J. Biol. Chem. 2007, 282, 18810–18818. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. McIlvain, H.B.; Ma, L.; Ludwig, B.; Manners, M.T.; Martone, R.L.; Dunlop, J.; Kaftan, E.J.; Kennedy, J.D.; Whiteside, G.T. Purinergic receptor-mediated morphological changes in microglia are transient and independent from inflammatory cytokine release. Eur. J. Pharm. 2010, 643, 202–210. [Google Scholar] [CrossRef] [PubMed]
  104. Pillai, S.; Bikle, D.D. Adenosine triphosphate stimulates phosphoinositide metabolism, mobilizes intracellular calcium, and inhibits terminal differentiation of human epidermal keratinocytes. J. Clin. Investig. 1992, 90, 42–51. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  105. Paoletti, A.; Allouch, A.; Caillet, M.; Saïdi, H.; Subra, F.; Nardacci, R.; Wu, Q.; Muradova, Z.; Voisin, L.; Raza, S.Q.; et al. HIV-1 Envelope Overcomes NLRP3-Mediated Inhibition of F-Actin Polymerization for Viral Entry. Cell Rep. 2019, 28, 3381–3394.e3387. [Google Scholar] [CrossRef] [Green Version]
  106. Giroud, C.; Marin, M.; Hammonds, J.; Spearman, P.; Melikyan, G.B. P2X1 Receptor Antagonists Inhibit HIV-1 Fusion by Blocking Virus-Coreceptor Interactions. J. Virol. 2015, 89, 9368–9382. [Google Scholar] [CrossRef] [Green Version]
  107. Soare, A.Y.; Durham, N.D.; Gopal, R.; Tweel, B.; Hoffman, K.W.; Brown, J.A.; O'Brien, M.; Bhardwaj, N.; Lim, J.K.; Chen, B.K.; et al. P2X Antagonists Inhibit HIV-1 Productive Infection and Inflammatory Cytokines Interleukin-10 (IL-10) and IL-1β in a Human Tonsil Explant Model. J. Virol. 2019, 93, e01186-18. [Google Scholar] [CrossRef] [Green Version]
  108. De Clercq, E. Suramin in the treatment of AIDS: Mechanism of action. Antivir. Res. 1987, 7, 1–10. [Google Scholar] [CrossRef]
  109. Nikolova, M.; Carriere, M.; Jenabian, M.A.; Limou, S.; Younas, M.; Kok, A.; Hue, S.; Seddiki, N.; Hulin, A.; Delaneau, O.; et al. CD39/adenosine pathway is involved in AIDS progression. PLoS Pathog. 2011, 7, e1002110. [Google Scholar] [CrossRef] [Green Version]
  110. Sorrell, M.E.; Hauser, K.F. Ligand-Gated Purinergic Receptors Regulate HIV-1 Tat and Morphine Related Neurotoxicity in Primary Mouse Striatal Neuron-Glia Co-Cultures. J. Neuroimmune Pharmacol. Off. J. Soc. Neuroimmune Pharmacol. 2014, 9, 233–244. [Google Scholar] [CrossRef] [Green Version]
  111. Tovar, Y.R.L.B.; Kolson, D.L.; Bandaru, V.V.; Drewes, J.L.; Graham, D.R.; Haughey, N.J. Adenosine triphosphate released from HIV-infected macrophages regulates glutamatergic tone and dendritic spine density on neurons. J. Neuroimmune Pharmacol. Off. J. Soc. Neuroimmune Pharmacol. 2013, 8, 998–1009. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  112. Jolly, C.; Kashefi, K.; Hollinshead, M.; Sattentau, Q.J. HIV-1 cell to cell transfer across an Env-induced, actin-dependent synapse. J. Exp. Med. 2004, 199, 283–293. [Google Scholar] [CrossRef] [PubMed]
  113. Hubner, W.; Chen, P.; Del Portillo, A.; Liu, Y.; Gordon, R.E.; Chen, B.K. Sequence of human immunodeficiency virus type 1 (HIV-1) Gag localization and oligomerization monitored with live confocal imaging of a replication-competent, fluorescently tagged HIV-1. J. Virol 2007, 81, 12596–12607. [Google Scholar] [CrossRef] [Green Version]
  114. Graziano, F.; Desdouits, M.; Garzetti, L.; Podini, P.; Alfano, M.; Rubartelli, A.; Furlan, R.; Benaroch, P.; Poli, G. Extracellular ATP induces the rapid release of HIV-1 from virus containing compartments of human macrophages. Proc. Natl. Acad. Sci. USA 2015, 112, E3265–E3273. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  115. Marin, M.; Du, Y.; Giroud, C.; Kim, J.H.; Qui, M.; Fu, H.; Melikyan, G.B. High-Throughput HIV-Cell Fusion Assay for Discovery of Virus Entry Inhibitors. Assay Drug Dev. Technol. 2015, 13, 155–166. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  116. Esposito, A.M.; Soare, A.Y.; Patel, F.; Satija, N.; Chen, B.K.; Swartz, T.H. A High-throughput Cre-Lox Activated Viral Membrane Fusion Assay to Identify Inhibitors of HIV-1 Viral Membrane Fusion. J. Vis. Exp. 2018. [Google Scholar] [CrossRef]
  117. Esposito, A.M.; Cheung, P.; Swartz, T.H.; Li, H.; Tsibane, T.; Durham, N.D.; Basler, C.F.; Felsenfeld, D.P.; Chen, B.K. A high throughput Cre-lox activated viral membrane fusion assay identifies pharmacological inhibitors of HIV entry. Virology 2016, 490, 6–16. [Google Scholar] [CrossRef]
  118. Soare, A.Y.; Malik, H.S.; Durham, N.D.; Freeman, T.L.; Alvarez, R.; Patel, F.; Satija, N.; Upadhyay, C.; Hioe, C.E.; Chen, B.K.; et al. P2X1 selective antagonists block HIV-1 infection through inhibition of envelope conformation-dependent fusion. J. Virol. 2019. [Google Scholar] [CrossRef]

Share and Cite

MDPI and ACS Style

Freeman, T.L.; Swartz, T.H. Purinergic Receptors: Elucidating the Role of these Immune Mediators in HIV-1 Fusion. Viruses 2020, 12, 290. https://doi.org/10.3390/v12030290

AMA Style

Freeman TL, Swartz TH. Purinergic Receptors: Elucidating the Role of these Immune Mediators in HIV-1 Fusion. Viruses. 2020; 12(3):290. https://doi.org/10.3390/v12030290

Chicago/Turabian Style

Freeman, Tracey L., and Talia H. Swartz. 2020. "Purinergic Receptors: Elucidating the Role of these Immune Mediators in HIV-1 Fusion" Viruses 12, no. 3: 290. https://doi.org/10.3390/v12030290

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