Skip to main content

MINI REVIEW article

Front. Pharmacol., 14 March 2018
Sec. Experimental Pharmacology and Drug Discovery
This article is part of the Research Topic Purinergic Pharmacology, Volume I View all 62 articles

Purinergic Ligands as Potential Therapeutic Tools for the Treatment of Inflammation-Related Intestinal Diseases

  • 1Medicinal Chemistry Unit, School of Pharmacy, University of Camerino, Camerino, Italy
  • 2Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy

Inflammation-related intestinal diseases are a set of various conditions presenting an overactive enteric immune system. A continuous overproduction of pro-inflammatory cytokines and a decreased production of anti-inflammatory modulators are generally observed, while morpho-functional alterations of the enteric nervous system lead to intestinal secretory and motor dysfunctions. The factors at the basis of these conditions are still to be totally identified and current therapeutic strategies are aimed only at achieving and maintaining remission states, by using therapeutic tools like aminosalicylates, corticosteroids, immunomodulators, biological drugs (i.e., monoclonal antibodies), and eventually surgery. Recent reports described a key role of purinergic mediators (i.e., adenosine and its nucleotides ATP and ADP) in the regulation of the activity of immune cells and enteric nervous system, showing also that alterations of the purinergic signaling are linked to pathological conditions of the intestinal tract. These data prompted to a series of investigations to test the therapeutic potential for inflammation-related intestinal conditions of compounds able to restore or modulate an altered purinergic signaling within the gut. This review provides an overview on these investigations, describing the results of preclinical and/or clinical evaluation of compounds able to stimulate or inhibit specific P2 (i.e., P2X7) or P1 (i.e., A2A or A3) receptor signaling and to modify the adenosine levels through the modulation of enzymes activity (i.e., Adenosine Deaminase) or nucleoside transporters. Recent developments in the field are also reported and the most promising purine-based therapeutic strategies for the treatment of inflammation-related gastrointestinal disorders are schematically summarized.

Introduction

Inflammatory bowel diseases (IBDs) comprise Crohn’s disease and ulcerative colitis and are conditions presenting an overactive intestinal immune system. The exact etiology of these diseases is still unclear but may be related to genetic predisposition or environmental factors and is characterized by an inappropriate immune response taking to morpho-functional alterations of the host’s enteric nervous system and intestinal secretory and motor dysfunctions. A loss of balance between the production of pro-inflammatory cytokines and anti-inflammatory mediators has been observed. Current therapeutic strategies are based on anti-inflammatory agents and targeted to achieve and maintain the remission state.

It is well established that during inflammation ATP is extracellularly released, a process involving pannexins or connexins and promoted by various stimuli (Eltzschig et al., 2012; Idzko et al., 2014). Extracellular ATP (eATP) is then degraded to adenosine by the ectonucleotidases CD39 and CD73 (Allard et al., 2017). While eATP generally plays a pro-inflammatory role through the activation of P2 (P2X and P2Y) purinergic receptors, the ATP degradation to adenosine usually represents a stop-signal for the inflammation process, with adenosine playing as anti-inflammatory agent through the activation of its P1 receptor targets. Adenosine is then removed from the extracellular environment by nucleoside transporters and/or metabolic enzymes.

Over the years increasing evidences pointed out a critical involvement of the purinergic system in the pathophysiology of IBDs, thus spurring the research toward the evaluation of the potential therapeutic benefits in terms of anti-inflammatory activity, arising by pharmacological targeting of purinergic pathways (Hasko and Cronstein, 2004; Hasko and Pacher, 2008; Hasko et al., 2008; Burnstock, 2011, 2014; Burnstock et al., 2017). Furthermore, the involvement of ATP in the enteric motor dysfunctions associated with bowel inflammation is a hot topic deserving further investigations.

P2 Purinergic Receptors

P2X Purinergic Receptors

P2X receptors are ligand-gated ion channels activated by eATP and permeable to Na+, K+, and Ca2+ (North and Jarvis, 2013; North, 2016). Seven P2X subtypes are known that may assemble as homo- or heterotrimers. Upon prolonged stimulation, some subtypes like the P2X7R undergo a rearrangement with the formation of a pore permeable to large molecules.

P2XR modulators are of great interest for several potential therapeutic applications, like treatment of pain, cough, cancer, and inflammation-related diseases (Burnstock and Kennedy, 2011; Syed and Kennedy, 2012; Muller, 2015). P2XR agonists are ATP derivatives obtained by modification of the purine base (i.e., 2-meSATP), the ribose ring (i.e., BzATP), or the polyphosphate chain (like the stable analogs αβ-meATP, βγ-meATP, and ATPγS) (Coddou et al., 2011; Dal Ben et al., 2015; Lambertucci et al., 2015).

P2XR antagonists are generally negatively charged molecules like TNP-ATP (Virginio et al., 1998) and analogs (Dal Ben et al., 2017), the irreversible inhibitor oxidized ATP (o-ATP) (Murgia et al., 1993), the P2X3R antagonist A-317491, and the polyanion suramin and its derivatives. Further classes of P2XR inhibitors are uncharged molecules based on heterocyclic scaffolds and behaving as non-competitive (allosteric) antagonists (Muller, 2015). A relevant number of structural classes of compounds were developed as P2X7R inhibitors (Park and Kim, 2017) given the key role of this receptor in pain and inflammation-related conditions (Arulkumaran et al., 2011; Gulbransen et al., 2012; De Marchi et al., 2016; Burnstock and Knight, 2017; Di Virgilio et al., 2017). P2X7R-targeting compounds have been developed also as radiolabeled molecules to be used as pharmacological tools or markers (Donnelly-Roberts et al., 2009; Lord et al., 2015; Fantoni et al., 2017; Territo et al., 2017; Jin et al., 2018).

The potent, selective, and orally bioavailable P2X7R antagonist AZD9056 was studied in phase-two clinical trials for the treatment of rheumatoid arthritis (RA) and chronic obstructive pulmonary disease (COPD) showing to be well tolerated (2005-004110-32_Clinical_Trial_Results, 2005; Keystone et al., 2012). The efficacy and safety of AZD9056 was also clinically assessed in the management of patients affected by moderate/severe Crohn’s disease. Although the lack in change of inflammatory parameters, this study demonstrated that AZD9056 has the potential to improve symptoms, in particular abdominal pain, in patients with IBDs (Eser et al., 2015). Other P2X7R antagonists (CE-224,535 and GSK1482160) were studied in clinical trials for RA and inflammatory pain conditions or showed (JNJ47965567) ability to enter the CNS.

In a rat model of trinitrobenzene sulfonic acid (TNBS) colitis, the administration of the P2X7R inhibitor A740003 determined a reduction of T-cell and macrophage infiltration in the lamina propria, followed by a reduction in tissue TNF and IL-1β concentrations, with a consequent amelioration of inflammation severity (Marques et al., 2014). In parallel, Neves et al. (2014) reported that mice lacking P2X7Rs and subjected to TNBS or dextran sulfate sodium (DSS) treatment failed to develop intestinal inflammation or other symptoms associated with colitis, thus indicating a protective role resulting from P2X7R blockade. In the same study, by exploring the expression of this receptor subtype in colonic mucosa of IBD patients, the authors observed high P2X7R levels in inflamed epithelium and lamina propria, where it colocalizes more with dendritic cells and macrophages, leading to hypothesize a role of P2X7R signaling in the pathogenesis of IBDs. Furthermore, Cesaro et al. (2010) pointed out the pivotal role of P2X7R in the complex cross-talk occurring between intestinal epithelial cells and immune cells. The pharmacological stimulation of P2X7R in a human colonic epithelial cell monolayer induced caspase-1 activation and IL-1β release, pro-inflammatory mediators critically involved in the recruitment of polymorphonuclear leukocytes within the intestinal mucosa in the presence of inflammation. Subsequently, several preclinical studies performed in animal models of colitis revealed a significant role of P2X7R in the pathophysiology of intestinal inflammation (Marques et al., 2014; Neves et al., 2014; Wan et al., 2016). Increasing efforts have been made to investigate the involvement of purinergic pathways in the pathophysiology of enteric motor dysfunction typically observed in the presence of intestinal inflammation, although the available data are fragmentary (Antonioli et al., 2013). Recently, Antonioli et al. (2014b) provided evidence about a marked increase in P2X7R immunostaining, and an enhanced modulating action of these receptors on colonic neuromotility in a rat model of DNBS-induced colitis.

Beside the importance of the P2X7R in the gastrointestinal diseases described in literature, recent papers suggest that even other P2X subtypes could play a relevant role the gastrointestinal pathophysiology (Paulino et al., 2011; Weng et al., 2015; Guo et al., 2016). Antagonists of the P2X3R showed promising activity in alleviating inflammatory and neuropathic pain in preclinical studies. A-317491 reduced visceral hypersensitivity in an experimental model of colitis, suggesting P2X3R as target for the treatment of inflammation-related abdominal pain syndromes (Deiteren et al., 2015).

P2Y Purinergic Receptors

P2Y receptors are G protein-coupled receptors of which eight subtypes (P2Y1, P2Y2, P2Y4, P2Y6, and P2Y11-14) are currently known. The endogenous agonists of these receptors are ATP, ADP, UTP, UDP, and UDP-glucose, with each P2Y subtype presenting peculiar pharmacological properties and preference for di- or triphosphate nucleotides (Jacobson and Muller, 2016). P2YRs are widely expressed in the body and involved in several biological functions. Beside the well-known inhibition of platelet aggregation, for which P2Y12R modulators like Clopidogrel, Prasugrel, Cangrelor, and Ticagrelor are approved for human use, P2YRs play important roles in neurotransmission and modulation of immune system (Le Duc et al., 2017).

P2YR ligands were developed by modification of endogenous ligands at the base (i.e., 2-thioUTP or 2-MeSATP), the sugar (i.e., MRS2365), or the phosphate chain (i.e., PSB1114) (von Kugelgen, 2006; Jacobson et al., 2012; Conroy et al., 2016; Jacobson and Muller, 2016). Dinucleoside derivatives are also ligands of the P2YRs. Diquafosol (approved in Japan for the treatment of Dry Eye disorder) and Denufosol (clinically evaluated for bronchial indication in cystic fibrosis) are representative compounds of this category and are endowed with dual P2Y2/P2Y4 receptor agonist profile. Suramin-based compounds are also P2YR modulators. Yet, several P2YR ligands belong to other structural classes not necessarily containing negatively charged functions (Conroy et al., 2016).

Considering the role of the P2Y2, P2Y6, and P2Y12 receptors in the inflammation-related conditions, compounds acting on these P2Y subtypes have been evaluated in such frames. P2Y12R antagonists present interesting potential to prevent a chronic inflammation promoted by this protein and to modulate the inflammatory pain (Thomas and Storey, 2015; Beko et al., 2017). Focusing on the bowel-related conditions, Grbic et al. (2008), investigating the role of ATP receptors in the pathogenesis of intestinal inflammation, reported that the pro-inflammatory cytokines TNF-α or IFN-γ determined an increased expression of P2Y2 and P2Y6 receptors in the colonic mucosa of mice with DSS colitis (Grbic et al., 2008; Degagne et al., 2013). Noteworthy, the pharmacological activation of P2Y2R via 2-thioUTP in a mouse model of DSS-induced colitis reduced the disease activity index and histological score values as well as a decrease in bacterial translocation to the spleen when compared with untreated mice, thus highlighting a protective role of P2Y2R in sustaining the remission phase in this experimental model of colitis (Degagne et al., 2013). By contrast, stimulation of the P2Y6R led to the activation of calcium-independent PKCδ upstream of ERK1/2, followed by the stimulation of c-fos phosphorylation and the recruitment of c-fos/c-jun dimers at level of the AP-1 motif located within the core promoter region of IL-8 gene, thus determining an increase of IL-8 release (Grbic et al., 2012). Despite these encouraging results, further investigations are needed to evaluate the putative beneficial effect of P2Y2 and P2Y6 receptor ligands in counteracting intestinal inflammation.

Considering the enteric motor dysfunction associated to intestinal inflammation, interesting findings were provided about the involvement of P2Y1R in the regulation of colonic neuromuscular activity in a model of TNBS-induced colitis in guinea-pig (Strong et al., 2010). Inflamed colonic specimens displayed a marked decrease in the fecal pellet output and a significant reduction of inhibitory junction potential (IJP). Of note, the pharmacological analysis of the determinant of IJP revealed that the purinergic component, mediated by P2Y1R, was impaired, despite immunohistochemical assays did not display significant alterations of nerve fiber density in circular muscle strips from animals with colitis (Strong et al., 2010).

P1 Adenosine Receptors

Adenosine Receptors (P1 receptors or ARs) are G protein-coupled receptors known as four subtypes (A1AR, A2AAR, A2BAR, and A3AR). Like the other Purinergic Receptors, ARs are widely expressed in the body and regulate many physiological functions. The endogenous ligand adenosine has a short half-life as it is internalized by nucleoside transporters and/or quickly modified to inosine by Adenosine Deaminase (ADA) or to AMP by Adenosine Kinase (ADK).

Medicinal chemistry efforts were aimed at developing compounds presenting higher metabolic stability and improved potency and selectivity compared to the endogenous ligand (Cristalli and Volpini, 2003; Jacobson and Gao, 2006; Wilson and Mustafa, 2009; Ciruela, 2011). A key modification of adenosine is the introduction of a N-alkyl-carboxamido function in the 4′-position to obtain NECA (N-ethyl) or MECA (N-methyl) derivatives, with an improved potency at all the ARs. Typical A1AR and A3AR agonists contain bulky groups in the N6-position combined with modifications in the 2-position (Kim et al., 1994; Kiesman et al., 2009; Volpini et al., 2009). Reference A3AR agonists are IB-MECA and Cl-IB-MECA, in clinical trials for inflammation-related conditions (RA and psoriasis) (Borea et al., 2015; Jacobson et al., 2017). A2AAR agonists are generally NECA derivatives presenting complex chains in the 2-position (i.e., CGS21680, ATL-146e, and ATL-313 where the 4′-carboxamido group is further modified). High A2AAR affinity and selectivity was obtained by inserting a bulky arylalkyl function in the N6-position to obtain UK-432097 (de Lera Ruiz et al., 2014).

About the molecules able to block the AR function, AR antagonists are generally divided in non-xanthine- and xanthine-based derivatives. Non-xanthine AR antagonists are based on a large variety of scaffolds (generally heterocycles). Reference compounds for pharmacological studies at ARs may be found within this heterogeneous group, some of which were developed also as water-soluble molecules, pro-drugs, and radiolabeled compounds. Xanthine-based AR antagonists contain the A2AAR inhibitor Istradefylline that was approved to market in Japan as antiparkinsonian tool (Jacobson and Muller, 2016).

At present, most of available studies investigating the role of AR signaling in several experimental models of colitis showed remarkable beneficial effects upon pharmacological modulation of A2AAR (Odashima et al., 2005; Cavalcante et al., 2006; Naganuma et al., 2006; Rahimian et al., 2010; Antonioli et al., 2011; Pallio et al., 2016). The A2AAR agonists ATL-146e or ATL-313 significantly reduced mucosal inflammation of colon, with a marked decrease in pro-inflammatory cytokine levels and in leukocyte infiltration and an increase of levels of anti-inflammatory cytokines (Naganuma et al., 2006; Odashima et al., 2006). Recently Pallio et al. (2016) demonstrated the beneficial effects arising from A2AAR stimulation with polydeoxyribonucleotide in two experimental models of colitis. In the DSS model polydeoxyribonucleotide could counteract the hemorrhagic diarrhea, improve the weight loss, and restore the anatomic integrity of damaged epithelial and mucosal layers. In the DNBS model, polydeoxyribonucleotide markedly reduced the inflammatory response as well as the granulocytic infiltration into the mucosal and submucosal layers and, therefore, decreased the pro-inflammatory cytokines TNF and IL-1β, MPO activity and lipid peroxidation in colon samples. Noteworthy, polydeoxyribonucleotide treatment also affected Bax and Bcl-2 expression, reducing apoptotic and necrotic cells in all tissue layers. By contrast, no beneficial effects have been reported upon administration of CGS21680 in mouse model of DSS-induced colitis (Selmeczy et al., 2007). Further investigations are needed to better characterize the therapeutic potential of A2AAR agonists in IBDs. A number of evidences reported that aging is often associated with a chronic, low-grade systemic inflammatory condition (Laflamme et al., 2017), that could predispose to the gastrointestinal alterations typical of the elderly subject (Remond et al., 2015). Recently, it has been demonstrated that the reduction of A2AAR in the digestive tract of aged mice, contributes to an increased inflammation and lower ability to counteract gut infection with deleterious effects in the elderly (Rodrigues et al., 2016).

Another promising option aimed at counteracting the bowel inflammation is the pharmacological stimulation of A3AR (Gessi et al., 2008; Antonioli et al., 2014a). The administration of IB-MECA revealed to afford a protective role in murine models of intestinal inflammation (Mabley et al., 2003; Guzman et al., 2006). In detail, the pharmacological engagement of A3AR determined the inhibition of several cytokine/chemokine/inflammatory genes, thus promoting a marked down-regulation of several pro-inflammatory mediators (MIP-1α and MIP-2, IL-1, IL-6, IL-12) and the production of reactive species of oxygen, determining an improvement of the intestinal damage (Guzman et al., 2006). A recent preclinical study by Ren et al. (2015) showed that the pharmacological stimulation of A3AR with Cl-IB-MECA inhibited the NF-κB pathway in the colonic epithelia of DSS colitis mice. The inhibition of both NF-κB activation and IκBa phosphorylation caused a reduction of pro-inflammatory cytokines expression in colonic epithelia of inflamed animals.

Finally, the evidence supporting an over-expression of A2BAR in experimental colitis has sparked interest on the potential therapeutic implications of these intriguing receptor subtype (Kolachala et al., 2005). The same research group (Kolachala V. et al., 2008; Kolachala V.L. et al., 2008) demonstrated also a critical role of intestinal epithelial A2BAR in the pro-inflammatory activity exerted by adenosine in animals with TNBS or DSS colitis. The treatment with the A2BAR antagonist ATL-801 to mice with experimental colitis ameliorated several inflammatory parameters, leading to a beneficial impact on the disease progression (Kolachala V. et al., 2008). By contrast, Frick et al. (2009) reported a detrimental effect exerted by the A2BAR inhibitor PSB1115 in the acute phase of DSS-induced colitis, thus questioning the beneficial effect of A2BAR blockade in the management of IBDs.

Regulation of Extracellular Adenosine Levels

Several authors investigated the efficacy of pharmacological treatments aimed at increasing the levels of endogenous adenosine, through the blockade of pivotal catabolic enzymes, as an alternative way to counteract intestinal inflammation. eATP is rapidly degraded to adenosine by ectonucleotidases CD73 and CD39. Genetic deletion of these enzymes prompts a higher susceptibility to inflammatory states or more severe progression of inflammation in IBD experimental models (Idzko et al., 2014; Longhi et al., 2017). Polymorphisms taking to lower ectonucleotidases expression takes to analog scenarios (Idzko et al., 2014). By contrast, blockade of nucleoside transporters leads to an increase of extracellular adenosine levels, with a consequent improvement of the inflammation course in IBD models (Ye and Rajendran, 2009). The extracellular levels of adenosine are also regulated by the activity of metabolic enzymes like ADK and ADA (Cristalli et al., 2001). The blockade of these enzymes is associated to anti-inflammatory effects and was preclinically tested as strategy to ameliorate intestinal inflammation-related conditions. Siegmund et al. (2001) reported, for the first time, an anti-inflammatory effect of the ADK inhibitor GP515 in a mouse model of colitis. Encouraging ameliorative effects were observed also following the inhibition of ADA in murine models of intestinal inflammation (Antonioli et al., 2007, 2010, 2012; La Motta et al., 2009), with reduction of the colonic inflammatory damage and decrease in tissue levels of pro-inflammatory cytokines (IL-1β, IL-6, TNF-α, IFN-γ, and chemokine C-X-C motif ligand 10), as well as a reduction of neutrophil infiltration and ROS production (Antonioli et al., 2007, 2010; Brown et al., 2008; La Motta et al., 2009). Noteworthy, it was observed that the attenuation of colonic injury, following ADA blockade, was mediated mainly by the engagement of A2AAR and, to a lesser extent, A3AR (Antonioli et al., 2010).

Conclusion

Several pre-clinical studies pointed out a key role for the purinergic system in the modulation of inflammatory and immune responses. Furthermore, clinical evaluation of purinergic ligands for the treatment of inflammation-related conditions (i.e., RA and psoriasis but also IBDs for a P2X7R inhibitor) showed a good tolerability of these molecules and suggests further investigations for this strategy. Figure 1 and Table 1 provide a schematic overview of the purinergic signal contributions in the intestinal tract and the most promising purine-based therapeutic strategies for the intestinal disorders.

FIGURE 1
www.frontiersin.org

FIGURE 1. Schematic figure with the putative contribution of the main purinergic signals (ATP and adenosine) and their different receptors (P2X3, P2X7, P2Y1,2,6, A2A, and A3) in different cell types of the intestinal tract.

TABLE 1
www.frontiersin.org

TABLE 1. Promising pharmacological tools acting on purinergic receptors to manage intestinal disorders.

Despite clinical studies demonstrated an encouraging profile in terms of tolerability for AZD9056, a novel P2X7R antagonist, it displayed a limited efficacy in the management of patients affected by moderate/severe Crohn’s disease. In parallel, an increasing interest has been payed toward the A2AAR (and the A3AR) agonists as viable way to manage digestive dysfunctions. However, despite promising the available evidences are limited to the pre-clinical phase and deserve further translational and clinical insights before highlighting their therapeutic potential. In this context, Michael Sitkovsky (Fredholm et al., 2007; Ohta and Sitkovsky, 2009) undoubtedly provided several of the most eminent scientific evidences about the therapeutic potential of ligands acting on this receptor subtype, paving the way toward their next clinical employment.

Apart from animal studies, the unique window of opportunity to grasp the relevance of part of the purinergic system, operated by adenosine receptors, in intestine function, is offered by the consumption of caffeine, which only known target at non-toxic conditions is the antagonism of adenosine receptors. In this regard, there are evidences, despite conflicting, about the association between caffeine consumption and the onset and development of gastrointestinal-related disorders. In particular, several authors reported a direct association between coffee consumption and some functional digestive disorders (i.e., gastro-oesophageal reflux, dyspepsia, irritable bowel syndrome) (Boekema et al., 1999; DiBaise, 2003).

These data boost the medicinal chemistry toward the synthesis of novel pharmacological entities acting selectively on specific purinergic receptors/enzymes and endowed with improved pharmacodynamic and pharmacokinetic profiles. These molecules would definitively help to clearly depict the pathophysiological role and the therapeutic potential of these proteins and would represent a key step for the development of useful tools for the management of intestinal inflammatory disorders (Burnstock, 2017a,b; Burnstock et al., 2017).

Author Contributions

DDB and LA drafted this manuscript. DDB, LA, CL, MF, CB, and RV were responsible for idea conception, critical evaluation, and manuscript review.

Conflict of Interest Statement

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

The handling Editor declared a past co-authorship with one of the authors DDB.

Acknowledgments

The authors acknowledge the contributions of University of Camerino (Progetto FAR FPI000042) in supporting the works.

References

2005-004110-32_Clinical_Trial_Results (2005). Clinical Trials for 2005-004110-32: A Randomised, Double-blind, Placebo-controlled, Parallel Group, Multicentre, Phase II Study to Assess The Efficacy of AZD9056 (single oral 400 mg dose) When Administered for 4 Weeks in Patients with Moderate to Severe COPD [Online]. Available at: https://www.clinicaltrialsregister.eu/ctr-search/search?query=2005-004110-32

Google Scholar

Allard, B., Longhi, M. S., Robson, S. C., and Stagg, J. (2017). The ectonucleotidases CD39 and CD73: novel checkpoint inhibitor targets. Immunol. Rev. 276, 121–144. doi: 10.1111/imr.12528

PubMed Abstract | CrossRef Full Text | Google Scholar

Antonioli, L., Colucci, R., La Motta, C., Tuccori, M., Awwad, O., Da Settimo, F., et al. (2012). Adenosine deaminase in the modulation of immune system and its potential as a novel target for treatment of inflammatory disorders. Curr. Drug Targets 13, 842–862. doi: 10.2174/138945012800564095

PubMed Abstract | CrossRef Full Text | Google Scholar

Antonioli, L., Colucci, R., Pellegrini, C., Giustarini, G., Tuccori, M., Blandizzi, C., et al. (2013). The role of purinergic pathways in the pathophysiology of gut diseases: pharmacological modulation and potential therapeutic applications. Pharmacol. Ther. 139, 157–188. doi: 10.1016/j.pharmthera.2013.04.002

PubMed Abstract | CrossRef Full Text | Google Scholar

Antonioli, L., Csoka, B., Fornai, M., Colucci, R., Kokai, E., Blandizzi, C., et al. (2014a). Adenosine and inflammation: what’s new on the horizon? Drug Discov. Today 19, 1051–1068. doi: 10.1016/j.drudis.2014.02.010

PubMed Abstract | CrossRef Full Text | Google Scholar

Antonioli, L., Fornai, M., Colucci, R., Awwad, O., Ghisu, N., Tuccori, M., et al. (2010). The blockade of adenosine deaminase ameliorates chronic experimental colitis through the recruitment of adenosine A2A and A3 receptors. J. Pharmacol. Exp. Ther. 335, 434–442. doi: 10.1124/jpet.110.171223

PubMed Abstract | CrossRef Full Text | Google Scholar

Antonioli, L., Fornai, M., Colucci, R., Awwad, O., Ghisu, N., Tuccori, M., et al. (2011). Differential recruitment of high affinity A1 and A2A adenosine receptors in the control of colonic neuromuscular function in experimental colitis. Eur. J. Pharmacol. 650, 639–649. doi: 10.1016/j.ejphar.2010.10.041

PubMed Abstract | CrossRef Full Text | Google Scholar

Antonioli, L., Fornai, M., Colucci, R., Ghisu, N., Da Settimo, F., Natale, G., et al. (2007). Inhibition of adenosine deaminase attenuates inflammation in experimental colitis. J. Pharmacol. Exp. Ther. 322, 435–442. doi: 10.1124/jpet.107.122762

PubMed Abstract | CrossRef Full Text | Google Scholar

Antonioli, L., Giron, M. C., Colucci, R., Pellegrini, C., Sacco, D., Caputi, V., et al. (2014b). Involvement of the P2X7 purinergic receptor in colonic motor dysfunction associated with bowel inflammation in rats. PLoS One 9:e116253. doi: 10.1371/journal.pone.0116253

PubMed Abstract | CrossRef Full Text | Google Scholar

Arulkumaran, N., Unwin, R. J., and Tam, F. W. (2011). A potential therapeutic role for P2X7 receptor (P2X7R) antagonists in the treatment of inflammatory diseases. Expert Opin. Investig. Drugs 20, 897–915. doi: 10.1517/13543784.2011.578068

PubMed Abstract | CrossRef Full Text | Google Scholar

Beko, K., Kovanyi, B., Goloncser, F., Horvath, G., Denes, A., Kornyei, Z., et al. (2017). Contribution of platelet P2Y12 receptors to chronic Complete Freund’s adjuvant-induced inflammatory pain. J. Thromb. Haemost. 15, 1223–1235. doi: 10.1111/jth.13684

PubMed Abstract | CrossRef Full Text | Google Scholar

Boekema, P. J., Samsom, M., van Berge Henegouwen, G. P., and Smout, A. J. (1999). Coffee and gastrointestinal function: facts and fiction. A review. Scand. J. Gastroenterol. Suppl. 230, 35–39. doi: 10.1080/003655299750025525

PubMed Abstract | CrossRef Full Text | Google Scholar

Borea, P. A., Varani, K., Vincenzi, F., Baraldi, P. G., Tabrizi, M. A., Merighi, S., et al. (2015). The A3 adenosine receptor: history and perspectives. Pharmacol. Rev. 67, 74–102. doi: 10.1124/pr.113.008540

PubMed Abstract | CrossRef Full Text | Google Scholar

Brown, J. B., Lee, G., Grimm, G. R., and Barrett, T. A. (2008). Therapeutic benefit of pentostatin in severe IL-10-/- colitis. Inflamm. Bowel Dis. 14, 880–887. doi: 10.1002/ibd.20410

PubMed Abstract | CrossRef Full Text | Google Scholar

Burnstock, G. (2011). Purinergic signaling in the gastrointestinal tract. World J. Gastrointest. Pathophysiol. 2, 31–34. doi: 10.4291/wjgp.v2.i2.31

PubMed Abstract | CrossRef Full Text | Google Scholar

Burnstock, G. (2014). Purinergic signalling in the gastrointestinal tract and related organs in health and disease. Purinergic Signal. 10, 3–50. doi: 10.1007/s11302-013-9397-9

PubMed Abstract | CrossRef Full Text | Google Scholar

Burnstock, G. (2017a). Purinergic Signalling: therapeutic developments. Front. Pharmacol. 8:661. doi: 10.3389/fphar.2017.00661

CrossRef Full Text | Google Scholar

Burnstock, G. (2017b). The therapeutic potential of purinergic signalling. Biochem. Pharmacol. doi: 10.1016/j.bcp.2017.07.016 [Epub ahead of print].

PubMed Abstract | CrossRef Full Text | Google Scholar

Burnstock, G., Jacobson, K. A., and Christofi, F. L. (2017). Purinergic drug targets for gastrointestinal disorders. Curr. Opin. Pharmacol. 37, 131–141. doi: 10.1016/j.coph.2017.10.011

PubMed Abstract | CrossRef Full Text | Google Scholar

Burnstock, G., and Kennedy, C. (2011). P2X receptors in health and disease. Adv. Pharmacol. 61, 333–372. doi: 10.1016/B978-0-12-385526-8.00011-4

PubMed Abstract | CrossRef Full Text | Google Scholar

Burnstock, G., and Knight, G. E. (2017). The potential of P2X7 receptors as a therapeutic target, including inflammation and tumour progression. Purinergic Signal. doi: 10.1007/s11302-017-9593-0 [Epub ahead of print].

PubMed Abstract | CrossRef Full Text | Google Scholar

Cavalcante, I. C., Castro, M. V., Barreto, A. R., Sullivan, G. W., Vale, M., Almeida, P. R., et al. (2006). Effect of novel A2A adenosine receptor agonist ATL 313 on Clostridium difficile toxin A-induced murine ileal enteritis. Infect. Immun. 74, 2606–2612. doi: 10.1128/IAI.74.5.2606-2612.2006

PubMed Abstract | CrossRef Full Text | Google Scholar

Cesaro, A., Brest, P., Hofman, V., Hebuterne, X., Wildman, S., Ferrua, B., et al. (2010). Amplification loop of the inflammatory process is induced by P2X7R activation in intestinal epithelial cells in response to neutrophil transepithelial migration. Am. J. Physiol. Gastrointest. Liver Physiol. 299, G32–G42. doi: 10.1152/ajpgi.00282.2009

PubMed Abstract | CrossRef Full Text | Google Scholar

Ciruela, F. (2011). Adenosine receptors. Biochim. Biophys. Acta 1808, 1231–1232. doi: 10.1016/j.bbamem.2011.03.007

PubMed Abstract | CrossRef Full Text | Google Scholar

Coddou, C., Yan, Z., Obsil, T., Huidobro-Toro, J. P., and Stojilkovic, S. S. (2011). Activation and regulation of purinergic P2X receptor channels. Pharmacol. Rev. 63, 641–683. doi: 10.1124/pr.110.003129

PubMed Abstract | CrossRef Full Text | Google Scholar

Conroy, S., Kindon, N., Kellam, B., and Stocks, M. J. (2016). Drug-like antagonists of P2Y receptors-from lead identification to drug development. J. Med. Chem. 59, 9981–10005. doi: 10.1021/acs.jmedchem.5b01972

PubMed Abstract | CrossRef Full Text | Google Scholar

Cristalli, G., Costanzi, S., Lambertucci, C., Lupidi, G., Vittori, S., Volpini, R., et al. (2001). Adenosine deaminase: functional implications and different classes of inhibitors. Med. Res. Rev. 21, 105–128. doi: 10.1002/1098-1128(200103)21:2<105::AID-MED1002>3.0.CO;2-U

PubMed Abstract | CrossRef Full Text | Google Scholar

Cristalli, G., and Volpini, R. (2003). Adenosine receptors: chemistry and pharmacology. Curr. Top. Med. Chem. 3, 355–469. doi: 10.2174/1568026033392282

CrossRef Full Text | Google Scholar

Dal Ben, D., Buccioni, M., Lambertucci, C., Marucci, G., Thomas, A., and Volpini, R. (2015). Purinergic P2X receptors: structural models and analysis of ligand-target interaction. Eur. J. Med. Chem. 89, 561–580. doi: 10.1016/j.ejmech.2014.10.071

PubMed Abstract | CrossRef Full Text | Google Scholar

Dal Ben, D., Marchenkova, A., Thomas, A., Lambertucci, C., Spinaci, A., Marucci, G., et al. (2017). 2’,3’-O-Substituted ATP derivatives as potent antagonists of purinergic P2X3 receptors and potential analgesic agents. Purinergic Signal. 13, 61–74. doi: 10.1007/s11302-016-9539-y

PubMed Abstract | CrossRef Full Text | Google Scholar

de Lera Ruiz, M., Lim, Y. H., and Zheng, J. (2014). Adenosine A2A receptor as a drug discovery target. J. Med. Chem. 57, 3623–3650. doi: 10.1021/jm4011669

PubMed Abstract | CrossRef Full Text | Google Scholar

De Marchi, E., Orioli, E., Dal Ben, D., and Adinolfi, E. (2016). P2X7 receptor as a therapeutic target. Adv. Protein Chem. Struct. Biol. 104, 39–79. doi: 10.1016/bs.apcsb.2015.11.004

PubMed Abstract | CrossRef Full Text | Google Scholar

Degagne, E., Degrandmaison, J., Grbic, D. M., Vinette, V., Arguin, G., and Gendron, F. P. (2013). P2Y2 receptor promotes intestinal microtubule stabilization and mucosal re-epithelization in experimental colitis. J. Cell. Physiol. 228, 99–109. doi: 10.1002/jcp.24109

PubMed Abstract | CrossRef Full Text | Google Scholar

Deiteren, A., van der Linden, L., de Wit, A., Ceuleers, H., Buckinx, R., Timmermans, J. P., et al. (2015). P2X3 receptors mediate visceral hypersensitivity during acute chemically-induced colitis and in the post-inflammatory phase via different mechanisms of sensitization. PLoS One 10:e0123810. doi: 10.1371/journal.pone.0123810

PubMed Abstract | CrossRef Full Text | Google Scholar

Di Virgilio, F., Dal Ben, D., Sarti, A. C., Giuliani, A. L., and Falzoni, S. (2017). The P2X7 receptor in infection and inflammation. Immunity 47, 15–31. doi: 10.1016/j.immuni.2017.06.020

PubMed Abstract | CrossRef Full Text | Google Scholar

DiBaise, J. K. (2003). A randomized, double-blind comparison of two different coffee-roasting processes on development of heartburn and dyspepsia in coffee-sensitive individuals. Dig. Dis. Sci. 48, 652–656. doi: 10.1023/A:1022860019852

PubMed Abstract | CrossRef Full Text | Google Scholar

Donnelly-Roberts, D. L., Namovic, M. T., Surber, B., Vaidyanathan, S. X., Perez-Medrano, A., Wang, Y., et al. (2009). [3H]A-804598 ([3H]2-cyano-1-[(1S)-1-phenylethyl]-3-quinolin-5-ylguanidine) is a novel, potent, and selective antagonist radioligand for P2X7 receptors. Neuropharmacology 56, 223–229. doi: 10.1016/j.neuropharm.2008.06.012

PubMed Abstract | CrossRef Full Text | Google Scholar

Eltzschig, H. K., Sitkovsky, M. V., and Robson, S. C. (2012). Purinergic signaling during inflammation. N. Engl. J. Med. 367, 2322–2333. doi: 10.1056/NEJMra1205750

PubMed Abstract | CrossRef Full Text | Google Scholar

Eser, A., Colombel, J. F., Rutgeerts, P., Vermeire, S., Vogelsang, H., Braddock, M., et al. (2015). Safety and efficacy of an oral inhibitor of the purinergic receptor P2X7 in adult patients with moderately to severely active Crohn’s disease: a randomized placebo-controlled, double-blind, phase IIa study. Inflamm. Bowel Dis. 21, 2247–2253. doi: 10.1097/MIB.0000000000000514

PubMed Abstract | CrossRef Full Text | Google Scholar

Fantoni, E. R., Dal Ben, D., Falzoni, S., Di Virgilio, F., Lovestone, S., and Gee, A. (2017). Design, synthesis and evaluation in an LPS rodent model of neuroinflammation of a novel 18F-labelled PET tracer targeting P2X7. EJNMMI Res. 7:31. doi: 10.1186/s13550-017-0275-2

PubMed Abstract | CrossRef Full Text | Google Scholar

Fredholm, B. B., Chern, Y., Franco, R., and Sitkovsky, M. (2007). Aspects of the general biology of adenosine A2A signaling. Prog. Neurobiol. 83, 263–276. doi: 10.1016/j.pneurobio.2007.07.005

PubMed Abstract | CrossRef Full Text | Google Scholar

Frick, J. S., MacManus, C. F., Scully, M., Glover, L. E., Eltzschig, H. K., and Colgan, S. P. (2009). Contribution of adenosine A2B receptors to inflammatory parameters of experimental colitis. J. Immunol. 182, 4957–4964. doi: 10.4049/jimmunol.0801324

PubMed Abstract | CrossRef Full Text | Google Scholar

Gessi, S., Merighi, S., Varani, K., Leung, E., Mac Lennan, S., and Borea, P. A. (2008). The A3 adenosine receptor: an enigmatic player in cell biology. Pharmacol. Ther. 117, 123–140. doi: 10.1016/j.pharmthera.2007.09.002

PubMed Abstract | CrossRef Full Text | Google Scholar

Grbic, D. M., Degagne, E., Langlois, C., Dupuis, A. A., and Gendron, F. P. (2008). Intestinal inflammation increases the expression of the P2Y6 receptor on epithelial cells and the release of CXC chemokine ligand 8 by UDP. J. Immunol. 180, 2659–2668. doi: 10.4049/jimmunol.180.4.2659

PubMed Abstract | CrossRef Full Text | Google Scholar

Grbic, D. M., Degagne, E., Larrivee, J. F., Bilodeau, M. S., Vinette, V., Arguin, G., et al. (2012). P2Y6 receptor contributes to neutrophil recruitment to inflamed intestinal mucosa by increasing CXC chemokine ligand 8 expression in an AP-1-dependent manner in epithelial cells. Inflamm. Bowel Dis. 18, 1456–1469. doi: 10.1002/ibd.21931

PubMed Abstract | CrossRef Full Text | Google Scholar

Gulbransen, B. D., Bashashati, M., Hirota, S. A., Gui, X., Roberts, J. A., MacDonald, J. A., et al. (2012). Activation of neuronal P2X7 receptor-pannexin-1 mediates death of enteric neurons during colitis. Nat. Med. 18, 600–604. doi: 10.1038/nm.2679

PubMed Abstract | CrossRef Full Text | Google Scholar

Guo, W., Sui, Q. Q., Gao, X. F., Feng, J. F., Zhu, J., He, C., et al. (2016). Co-localization of Pirt protein and P2X2 receptors in the mouse enteric nervous system. Purinergic Signal. 12, 489–496. doi: 10.1007/s11302-016-9515-6

PubMed Abstract | CrossRef Full Text | Google Scholar

Guzman, J., Yu, J. G., Suntres, Z., Bozarov, A., Cooke, H., Javed, N., et al. (2006). ADOA3R as a therapeutic target in experimental colitis: proof by validated high-density oligonucleotide microarray analysis. Inflamm. Bowel Dis. 12, 766–789. doi: 10.1097/00054725-200608000-00014

PubMed Abstract | CrossRef Full Text | Google Scholar

Hasko, G., and Cronstein, B. N. (2004). Adenosine: an endogenous regulator of innate immunity. Trends Immunol. 25, 33–39. doi: 10.1016/j.it.2003.11.003

CrossRef Full Text | Google Scholar

Hasko, G., Linden, J., Cronstein, B., and Pacher, P. (2008). Adenosine receptors: therapeutic aspects for inflammatory and immune diseases. Nat. Rev. Drug Discov. 7, 759–770. doi: 10.1038/nrd2638

PubMed Abstract | CrossRef Full Text | Google Scholar

Hasko, G., and Pacher, P. (2008). A2A receptors in inflammation and injury: lessons learned from transgenic animals. J. Leukoc. Biol. 83, 447–455. doi: 10.1189/jlb.0607359

PubMed Abstract | CrossRef Full Text | Google Scholar

Idzko, M., Ferrari, D., and Eltzschig, H. K. (2014). Nucleotide signalling during inflammation. Nature 509, 310–317. doi: 10.1038/nature13085

PubMed Abstract | CrossRef Full Text | Google Scholar

Jacobson, K. A., Balasubramanian, R., Deflorian, F., and Gao, Z. G. (2012). G protein-coupled adenosine (P1) and P2Y receptors: ligand design and receptor interactions. Purinergic Signal. 8, 419–436. doi: 10.1007/s11302-012-9294-7

PubMed Abstract | CrossRef Full Text | Google Scholar

Jacobson, K. A., and Gao, Z. G. (2006). Adenosine receptors as therapeutic targets. Nat. Rev. Drug. Discov. 5, 247–264. doi: 10.1038/nrd1983

PubMed Abstract | CrossRef Full Text | Google Scholar

Jacobson, K. A., Merighi, S., Varani, K., Borea, P. A., Baraldi, S., Aghazadeh Tabrizi, M., et al. (2017). A3 adenosine receptors as modulators of inflammation: from medicinal chemistry to therapy. Med. Res. Rev. doi: 10.1002/med.21456 [Epub ahead of print].

PubMed Abstract | CrossRef Full Text | Google Scholar

Jacobson, K. A., and Muller, C. E. (2016). Medicinal chemistry of adenosine, P2Y and P2X receptors. Neuropharmacology 104, 31–49. doi: 10.1016/j.neuropharm.2015.12.001

PubMed Abstract | CrossRef Full Text | Google Scholar

Jin, H., Han, J., Resing, D., Liu, H., Yue, X., Miller, R. L., et al. (2018). Synthesis and in vitro characterization of a P2X7 radioligand [123I]TZ6019 and its response to neuroinflammation in a mouse model of Alzheimer disease. Eur. J. Pharmacol. 820, 8–17. doi: 10.1016/j.ejphar.2017.12.006

PubMed Abstract | CrossRef Full Text | Google Scholar

Keystone, E. C., Wang, M. M., Layton, M., Hollis, S., and McInnes, I. B. (2012). Clinical evaluation of the efficacy of the P2X7 purinergic receptor antagonist AZD9056 on the signs and symptoms of rheumatoid arthritis in patients with active disease despite treatment with methotrexate or sulphasalazine. Ann. Rheum. Dis. 71, 1630–1635. doi: 10.1136/annrheumdis-2011-143578

PubMed Abstract | CrossRef Full Text | Google Scholar

Kiesman, W. F., Elzein, E., and Zablocki, J. (2009). A1 adenosine receptor antagonists, agonists, and allosteric enhancers. Handb. Exp. Pharmacol. 193, 25–58. doi: 10.1007/978-3-540-89615-9_2

PubMed Abstract | CrossRef Full Text | Google Scholar

Kim, H. O., Ji, X. D., Siddiqi, S. M., Olah, M. E., Stiles, G. L., and Jacobson, K. A. (1994). 2-Substitution of N6-benzyladenosine-5’-uronamides enhances selectivity for A3 adenosine receptors. J. Med. Chem. 37, 3614–3621. doi: 10.1021/jm00047a018

CrossRef Full Text | Google Scholar

Kolachala, V., Asamoah, V., Wang, L., Obertone, T. S., Ziegler, T. R., Merlin, D., et al. (2005). TNF-alpha upregulates adenosine 2b (A2b) receptor expression and signaling in intestinal epithelial cells: a basis for A2bR overexpression in colitis. Cell. Mol. Life Sci. 62, 2647–2657. doi: 10.1007/s00018-005-5328-4

PubMed Abstract | CrossRef Full Text | Google Scholar

Kolachala, V., Ruble, B., Vijay-Kumar, M., Wang, L., Mwangi, S., Figler, H., et al. (2008). Blockade of adenosine A2B receptors ameliorates murine colitis. Br. J. Pharmacol. 155, 127–137. doi: 10.1038/bjp.2008.227

PubMed Abstract | CrossRef Full Text | Google Scholar

Kolachala, V. L., Vijay-Kumar, M., Dalmasso, G., Yang, D., Linden, J., Wang, L., et al. (2008). A2B adenosine receptor gene deletion attenuates murine colitis. Gastroenterology 135, 861–870. doi: 10.1053/j.gastro.2008.05.049

PubMed Abstract | CrossRef Full Text | Google Scholar

La Motta, C., Sartini, S., Mugnaini, L., Salerno, S., Simorini, F., Taliani, S., et al. (2009). Exploiting the pyrazolo[3,4-d]pyrimidin-4-one ring system as a useful template to obtain potent adenosine deaminase inhibitors. J. Med. Chem. 52, 1681–1692. doi: 10.1021/jm801427r

PubMed Abstract | CrossRef Full Text | Google Scholar

Laflamme, C., Mailhot, G. B., and Pouliot, M. (2017). Age-related decline of the acute local inflammation response: a mitigating role for the adenosine A2A receptor. Aging 9, 2083–2097. doi: 10.18632/aging.101303

PubMed Abstract | CrossRef Full Text | Google Scholar

Lambertucci, C., Dal Ben, D., Buccioni, M., Marucci, G., Thomas, A., and Volpini, R. (2015). Medicinal chemistry of P2X receptors: agonists and orthosteric antagonists. Curr. Med. Chem. 22, 915–928. doi: 10.2174/0929867321666141215093513

PubMed Abstract | CrossRef Full Text | Google Scholar

Le Duc, D., Schulz, A., Lede, V., Schulze, A., Thor, D., Bruser, A., et al. (2017). P2Y receptors in immune response and inflammation. Adv. Immunol. 136, 85–121. doi: 10.1016/bs.ai.2017.05.006

PubMed Abstract | CrossRef Full Text | Google Scholar

Longhi, M. S., Moss, A., Jiang, Z. G., and Robson, S. C. (2017). Purinergic signaling during intestinal inflammation. J. Mol. Med. 95, 915–925. doi: 10.1007/s00109-017-1545-1

PubMed Abstract | CrossRef Full Text | Google Scholar

Lord, B., Ameriks, M. K., Wang, Q., Fourgeaud, L., Vliegen, M., Verluyten, W., et al. (2015). A novel radioligand for the ATP-gated ion channel P2X7: [3H] JNJ-54232334. Eur. J. Pharmacol. 765, 551–559. doi: 10.1016/j.ejphar.2015.09.026

PubMed Abstract | CrossRef Full Text | Google Scholar

Mabley, J., Soriano, F., Pacher, P., Hasko, G., Marton, A., Wallace, R., et al. (2003). The adenosine A3 receptor agonist, N6-(3-iodobenzyl)-adenosine-5’-N-methyluronamide, is protective in two murine models of colitis. Eur. J. Pharmacol. 466, 323–329. doi: 10.1016/S0014-2999(03)01570-X

CrossRef Full Text | Google Scholar

Marques, C. C., Castelo-Branco, M. T., Pacheco, R. G., Buongusto, F., do Rosário, A. Jr., Schanaider, A., et al. (2014). Prophylactic systemic P2X7 receptor blockade prevents experimental colitis. Biochim. Biophys. Acta 1842, 65–78. doi: 10.1016/j.bbadis.2013.10.012

PubMed Abstract | CrossRef Full Text | Google Scholar

Muller, C. E. (2015). Medicinal chemistry of P2X receptors: allosteric modulators. Curr. Med. Chem. 22, 929–941. doi: 10.2174/0929867322666141210155610

CrossRef Full Text | Google Scholar

Murgia, M., Hanau, S., Pizzo, P., Rippa, M., and Di Virgilio, F. (1993). Oxidized ATP. An irreversible inhibitor of the macrophage purinergic P2Z receptor. J. Biol. Chem. 268, 8199–8203.

Google Scholar

Naganuma, M., Wiznerowicz, E. B., Lappas, C. M., Linden, J., Worthington, M. T., and Ernst, P. B. (2006). Cutting edge: critical role for A2A adenosine receptors in the T cell-mediated regulation of colitis. J. Immunol. 177, 2765–2769. doi: 10.4049/jimmunol.177.5.2765

PubMed Abstract | CrossRef Full Text | Google Scholar

Neves, A. R., Castelo-Branco, M. T., Figliuolo, V. R., Bernardazzi, C., Buongusto, F., Yoshimoto, A., et al. (2014). Overexpression of ATP-activated P2X7 receptors in the intestinal mucosa is implicated in the pathogenesis of Crohn’s disease. Inflamm. Bowel Dis. 20, 444–457. doi: 10.1097/01.MIB.0000441201.10454.06

PubMed Abstract | CrossRef Full Text | Google Scholar

North, R. A. (2016). P2X receptors. Philos. Trans. R. Soc. Lond. B Biol. Sci. 371:20150427. doi: 10.1098/rstb.2015.0427

PubMed Abstract | CrossRef Full Text | Google Scholar

North, R. A., and Jarvis, M. F. (2013). P2X receptors as drug targets. Mol. Pharmacol. 83, 759–769. doi: 10.1124/mol.112.083758

PubMed Abstract | CrossRef Full Text | Google Scholar

Odashima, M., Bamias, G., Rivera-Nieves, J., Linden, J., Nast, C. C., Moskaluk, C. A., et al. (2005). Activation of A2A adenosine receptor attenuates intestinal inflammation in animal models of inflammatory bowel disease. Gastroenterology 129, 26–33. doi: 10.1053/j.gastro.2005.05.032

PubMed Abstract | CrossRef Full Text | Google Scholar

Odashima, M., Otaka, M., Jin, M., Horikawa, Y., Matsuhashi, T., Ohba, R., et al. (2006). A selective adenosine A2A receptor agonist, ATL-146e, prevents concanavalin A-induced acute liver injury in mice. Biochem. Biophys. Res. Commun. 347, 949–954. doi: 10.1016/j.bbrc.2006.06.185

PubMed Abstract | CrossRef Full Text | Google Scholar

Ohta, A., and Sitkovsky, M. (2009). The adenosinergic immunomodulatory drugs. Curr. Opin. Pharmacol. 9, 501–506. doi: 10.1016/j.coph.2009.05.005

PubMed Abstract | CrossRef Full Text | Google Scholar

Pallio, G., Bitto, A., Pizzino, G., Galfo, F., Irrera, N., Squadrito, F., et al. (2016). Adenosine receptor stimulation by polydeoxyribonucleotide improves tissue repair and symptomology in experimental colitis. Front. Pharmacol. 7:273. doi: 10.3389/fphar.2016.00273

PubMed Abstract | CrossRef Full Text | Google Scholar

Park, J. H., and Kim, Y. C. (2017). P2X7 receptor antagonists: a patent review (2010-2015). Expert Opin. Ther. Pat. 27, 257–267. doi: 10.1080/13543776.2017.1246538

PubMed Abstract | CrossRef Full Text | Google Scholar

Paulino, A. S., Palombit, K., Cavriani, G., Tavares-de-Lima, W., Mizuno, M. S., Marosti, A. R., et al. (2011). Effects of ischemia and reperfusion on P2X2 receptor expressing neurons of the rat ileum enteric nervous system. Dig. Dis. Sci. 56, 2262–2275. doi: 10.1007/s10620-011-1588-z

PubMed Abstract | CrossRef Full Text | Google Scholar

Rahimian, R., Fakhfouri, G., Daneshmand, A., Mohammadi, H., Bahremand, A., Rasouli, M. R., et al. (2010). Adenosine A2A receptors and uric acid mediate protective effects of inosine against TNBS-induced colitis in rats. Eur. J. Pharmacol. 649, 376–381. doi: 10.1016/j.ejphar.2010.09.044

PubMed Abstract | CrossRef Full Text | Google Scholar

Remond, D., Shahar, D. R., Gille, D., Pinto, P., Kachal, J., Peyron, M. A., et al. (2015). Understanding the gastrointestinal tract of the elderly to develop dietary solutions that prevent malnutrition. Oncotarget 6, 13858–13898. doi: 10.18632/oncotarget.4030

PubMed Abstract | CrossRef Full Text | Google Scholar

Ren, T., Tian, T., Feng, X., Ye, S., Wang, H., Wu, W., et al. (2015). An adenosine A3 receptor agonist inhibits DSS-induced colitis in mice through modulation of the NF-kappaB signaling pathway. Sci. Rep. 5:9047. doi: 10.1038/srep09047

PubMed Abstract | CrossRef Full Text | Google Scholar

Rodrigues, L., Miranda, I. M., Andrade, G. M., Mota, M., Cortes, L., Rodrigues, A. G., et al. (2016). Blunted dynamics of adenosine A2A receptors is associated with increased susceptibility to Candida albicans infection in the elderly. Oncotarget 7, 62862–62872. doi: 10.18632/oncotarget.11760

PubMed Abstract | CrossRef Full Text | Google Scholar

Selmeczy, Z., Csoka, B., Pacher, P., Vizi, E. S., and Hasko, G. (2007). The adenosine A2A receptor agonist CGS 21680 fails to ameliorate the course of dextran sulphate-induced colitis in mice. Inflamm. Res. 56, 204–209. doi: 10.1007/s00011-006-6150-7

PubMed Abstract | CrossRef Full Text | Google Scholar

Siegmund, B., Rieder, F., Albrich, S., Wolf, K., Bidlingmaier, C., Firestein, G. S., et al. (2001). Adenosine kinase inhibitor GP515 improves experimental colitis in mice. J. Pharmacol. Exp. Ther. 296, 99–105. doi: 10.1016/S0016-5085(00)84448-0

PubMed Abstract | CrossRef Full Text | Google Scholar

Strong, D. S., Cornbrooks, C. F., Roberts, J. A., Hoffman, J. M., Sharkey, K. A., and Mawe, G. M. (2010). Purinergic neuromuscular transmission is selectively attenuated in ulcerated regions of inflamed guinea pig distal colon. J. Physiol. 588, 847–859. doi: 10.1113/jphysiol.2009.185082

PubMed Abstract | CrossRef Full Text | Google Scholar

Syed, N. I. H., and Kennedy, C. (2012). Pharmacology of P2X receptors. WIREs Membr. Transp. Signal. 1, 16–30. doi: 10.1002/wmts.1

CrossRef Full Text | Google Scholar

Territo, P. R., Meyer, J. A., Peters, J. S., Riley, A. A., McCarthy, B. P., Gao, M., et al. (2017). Characterization of 11C-GSK1482160 for targeting the P2X7 receptor as a biomarker for neuroinflammation. J. Nucl. Med. 58, 458–465. doi: 10.2967/jnumed.116.181354

PubMed Abstract | CrossRef Full Text | Google Scholar

Thomas, M. R., and Storey, R. F. (2015). Effect of P2Y12 inhibitors on inflammation and immunity. Thromb. Haemost. 114, 490–497. doi: 10.1160/TH14-12-1068

PubMed Abstract | CrossRef Full Text | Google Scholar

Virginio, C., Robertson, G., Surprenant, A., and North, R. A. (1998). Trinitrophenyl-substituted nucleotides are potent antagonists selective for P2X1, P2X3, and heteromeric P2X2/3 receptors. Mol. Pharmacol. 53, 969–973. doi: 10.1124/mol.54.6.979

PubMed Abstract | CrossRef Full Text | Google Scholar

Volpini, R., Buccioni, M., Dal Ben, D., Lambertucci, C., Lammi, C., Marucci, G., et al. (2009). Synthesis and biological evaluation of 2-alkynyl-N6-methyl-5′-N-methylcarboxamidoadenosine derivatives as potent and highly selective agonists for the human adenosine A3 receptor. J. Med. Chem. 52, 7897–7900. doi: 10.1021/jm900754g

PubMed Abstract | CrossRef Full Text | Google Scholar

von Kugelgen, I. (2006). Pharmacological profiles of cloned mammalian P2Y-receptor subtypes. Pharmacol. Ther. 110, 415–432. doi: 10.1016/j.pharmthera.2005.08.014

PubMed Abstract | CrossRef Full Text | Google Scholar

Wan, P., Liu, X., Xiong, Y., Ren, Y., Chen, J., Lu, N., et al. (2016). Extracellular ATP mediates inflammatory responses in colitis via P2X7 receptor signaling. Sci. Rep. 6:19108. doi: 10.1038/srep19108

PubMed Abstract | CrossRef Full Text | Google Scholar

Weng, Z. J., Wu, L. Y., Zhou, C. L., Dou, C. Z., Shi, Y., Liu, H. R., et al. (2015). Effect of electroacupuncture on P2X3 receptor regulation in the peripheral and central nervous systems of rats with visceral pain caused by irritable bowel syndrome. Purinergic Signal. 11, 321–329. doi: 10.1007/s11302-015-9447-6

PubMed Abstract | CrossRef Full Text | Google Scholar

Wilson, C. N., and Mustafa, S. J. (2009). Adenosine Receptors in Health and Disease: Handbook of Experimental Pharmacology, Vol. 193. Berlin: Springer. doi: 10.1007/978-3-540-89615-9

CrossRef Full Text | Google Scholar

Ye, J. H., and Rajendran, V. M. (2009). Adenosine: an immune modulator of inflammatory bowel diseases. World J. Gastroenterol. 15, 4491–4498. doi: 10.3748/wjg.15.4491

CrossRef Full Text | Google Scholar

Keywords: inflammation, intestinal diseases, intestinal immune system, modulators, purinergic receptors, purinergic ligands, adenosine, therapeutic tools

Citation: Dal Ben D, Antonioli L, Lambertucci C, Fornai M, Blandizzi C and Volpini R (2018) Purinergic Ligands as Potential Therapeutic Tools for the Treatment of Inflammation-Related Intestinal Diseases. Front. Pharmacol. 9:212. doi: 10.3389/fphar.2018.00212

Received: 16 October 2017; Accepted: 26 February 2018;
Published: 14 March 2018.

Edited by:

Kenneth A. Jacobson, National Institutes of Health (NIH), United States

Reviewed by:

Rodrigo A. Cunha, University of Coimbra, Portugal
Eberhard Weihe, Philipps University of Marburg, Germany

Copyright © 2018 Dal Ben, Antonioli, Lambertucci, Fornai, Blandizzi and Volpini. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Rosaria Volpini, rosaria.volpini@unicam.it

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.