Skip to main content

OPINION article

Front. Pharmacol., 29 January 2021
Sec. Experimental Pharmacology and Drug Discovery
This article is part of the Research Topic Purinergic Signaling 2020: the State-of-The-Art Commented by the Members of the Italian Purine Club View all 41 articles

New Insight into the Role of Adenosine in Demyelination, Stroke and Neuropathic Pain

  • Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), Section of Pharmacology and Toxicology, University of Florence, Florence, Italy

Introduction

Our group of research has a long-time experience in studying the role/s of adenosine P1 receptors in health and disease conditions of the central nervous system (CNS). The effects of adenosine were investigated in our laboratory by the use of in vitro or in vivo models.

In most recent years, main topics of our research concerned the study of adenosine and its receptors in the physiological processes of myelination and in pathological conditions of ischemia. Most recently, the group addressed the study of adenosine-mediated mechanisms in pain control.

Here, we are going to summarize our recent findings.

Areas of Interest

Role of A2ARs and A2BRs in Oligodendrogliogenesis

Oligodendrocytes (OLs) are the only myelinating cells in the CNS and differentiate throughout adult life from their progenitors, the oligodendrocyte progenitor cells (OPCs), to produce myelin sheets around neuronal axons. Growing evidence indicates that failure of myelin formation in demyelinating diseases, i.e., multiple sclerosis (MS), arises from the disruption of OPC differentiation process (Levine et al., 2001). Hence, therapeutic strategies aimed at fostering this process are largely attractive.

Purines are important modulators of cell development in different cellular systems (Coppi et al., 2007b; Coppi et al., 2012; Burnstock and Dale, 2015). Concerning oligodendrogliogenesis, it is now generally recognized that adenosine plays a key role in OPC maturation. All subtypes of adenosine P1 receptors are expressed during the entire process of OPC maturation (Stevens et al., 2002; Coppi et al., 2013a; Coppi et al., 2020a). Indeed, adenosine is known to be released upon electrical stimulation of nearby neurons to acts as a neuron-glial transmitter that inhibits OPC proliferation and facilitates their differentiation (Stevens et al., 2002). Adenosine A1R stimulation is also known to promote OPC migration (Othman et al., 2003), another important process aimed to repair brain damage and repopulate the injured site (de Castro and Bribian, 2005).

Our attention was devoted to study the role of the different subtypes of adenosine P1 receptors, the adenosine A2ARs and A2BRs, in this process. Our group of research significantly contributed to gain insight into the field by demonstrating that selective adenosine A2AR stimulation inhibits OPC maturation by reducing voltage-dependent, sustained, outward K+ currents (IK) sensitive to tetraethyilammonium (TEA) (Coppi et al., 2013a; Coppi et al., 2015; Cherchi et al., 2020). These data are well in keeping with original research from Gallo and coworkers who first demonstrated that TEA, incubated in OPC cultures, reduces the expression of myelin proteins (Gallo et al., 1996).

Successive studies by our laboratory demonstrated that adenosine A2BR agonists, either the prototypical compound BAY60-6583 or the recently synthetized BAY60-6583-analogue P456 (Betti et al., 2018), not only inhibit sustained IK but also transient IA conductances in cultured OPCs (Coppi et al., 2020a). The effect on IK was mimicked and occluded by forskolin, demonstrating its dependency upon intracellular cAMP increase, consistently with Gs-coupling of A2AR and A2BR and with the fact that the Gi-coupled GPR17 (Pugliese et al., 2009b) elicits the opposite effect in OPCs (Coppi et al., 2013b). Importantly, in the same work, we reported that A2BR agonists activate sphingosine-kinase-1 (SphK1), an ubiquitous enzyme responsible for the formation of sphingosine-1-phosphate (S1P). Finally, in vitro silencing of A2BR was sufficient to increase OPC maturation, to inhibit SphK1 expression, and to strikingly rise the levels of S1P lyase, the enzyme devoted to irreversible degradation of S1P. In conclusion, our data demonstrate that A2BR stimulation enhances intracellular S1P levels, whereas A2BR silencing triggers S1P catabolism. Since S1P acts as a mitogen in a variety of cells including OPCs (Jung et al., 2007), we postulate that the antidifferentiative role of A2BR in oligodendrogliogenesis is exerted by increasing intracellular S1P levels. These results are particularly attractive since the first approved oral agent for MS treatment is the S1P analogue fingolimod (FTY720). Thus, our results may open interesting possibilities about a pharmacological control of myelin formation and repair during demyelinating pathologies by A2BR ligands, possibly in combination with S1P modulators.

Role of Adenosine A2ARs and A2BRs in Brain Ischemia

A main topic of our research group concerns the role of different P1 receptor subtypes in modulating the damage inflicted to neurons and glia by ischemia. To this purpose, both the in vitro model of oxygen and glucose deprivation (OGD) in rat hippocampal slices or the in vivo model of ischemia induced by middle cerebral artery occlusion (MCAo) in the rat were used. It is well accepted that adenosine released during brain ischemia exerts a neuroprotective role by A1R-mediated mechanisms (Pedata et al., 2016). However, as A1R-selective agonists became clinically less and less attractive due to important side effects such as bradycardia and sedation (Sollevi, 1986; Vonlubitz et al., 1994), our research focused on A2AR and A2BR subtypes.

In the in vitro OGD model in acute rat hippocampal slices, we monitored the excitotoxic damage mediated by excessive glutamate release during a severe OGD insult of 7 or 9 min (Pugliese et al., 2006; Pugliese et al., 2009a; Colotta et al., 2012). Purines are important modulators of neuronal damage during OGD (Coppi et al., 2007a; Maraula et al., 2014). Selective antagonists of adenosine A2ARs, the SCH58261 and SCH442416, protect from the OGD-induced irreversible disappearance of field excitatory post-synaptic potentials (fEPSPs) and from the appearance of anoxic depolarization (AD), an unequivocal sign of neuronal injury. The A2AR antagonists also delay AD onset invariably induced during a long period, 30 min, OGD in the CA1 region (Pugliese et al., 2009a) or in the dentate gyrus (DG) (Maraula et al., 2013) of the hippocampus. In the DG, A2AR antagonists also restore the number of 5-bromo-20-deoxyuridine-positive (BrdU+) newborn neurons 6 h after the end of the insult (Maraula et al., 2013), limit the extent of CA1 damage to neurons and glia assessed by propidium iodide permeability, and reduce astrocyte activation (Pugliese et al., 2009a).

Similar results were later found with antagonists to the “partner” adenosinergic receptor, i.e., the A2BR subtype. The compounds MRS1754 and PSB603, by blocking the A2BR, protected CA1 hippocampal slices from OGD-induced damage by preventing irreversible synaptic failure and AD appearance induced by OGD and counteracted CA1 neuronal loss, astrocyte activation, and cytochrome C release (Fusco et al., 2018). In line with previous literature (Goncalves et al., 2015), we confirmed that protection by the adenosine A2BR antagonists is attributable to diminished glutamate release since the A2BR agonist BAY60-6583, and its analogue P456, significantly decreased hippocampal paired pulse facilitation (PPF) (Fusco et al., 2019), an electrophysiological paradigm used to detect presynaptic neuromodulation of glutamate release (Regehr, 2012).

Consistent with previous results, we found that the selective A2AR antagonist, SCH58261, acutely (5 min) or subchronically (5 min, 6 h, and 20 h) administered in the in vivo model of permanent MCAo (pMCAo) in the rat, 24 h thereafter, was protective against neurological deficit and brain damage (Melani et al., 2003; Melani et al., 2006; Melani et al., 2009). Such protection can be related to the activation of deleterious pathways of MAPK expressed in microglia, such as p38, or JNK, expressed in mature OLs and in OPCs (Melani et al., 2006), which is considered a inhibitory molecule that can hinder myelin reconstitution and neuron functionality (Melani et al., 2009). Such a protective effect induced by adenosine A2AR antagonism in the first hours after ischemia found a valuable explanation in the ability to control excessive glutamate extracellular concentrations as detected by the technique of cerebral microdialysis (Melani et al., 2003) and the ensuing acute excitoxicity after ischemia.

In an apparent paradoxical manner, we found that, in the model of transient, 1 h, MCAo (tMCAo), not the antagonist but the agonist at the A2AR, CGS21680, exerted protective effects. However, this observation was made later from ischemia induction, i.e., 7 days after ischemia. Indeed, the chronic administration (twice/day for 7 days) with CGS21680 induced protection from neurological deficit, weight loss, cortical infarct volume, myelin disorganization, and glial activation (Melani et al., 2014). Without excluding that protection by A2AR agonists is attributable to central effects, such as increasing expression and release of neurotrophic factors (Sebastiao and Ribeiro, 2009), data reported by our research group point toward a protective effect due to peripheral A2AR activation. A2ARs are expressed on blood cells where they definitely reduce adhesion cell factor production, platelet aggregation and neutrophil activation, exerting, therefore, an antithrombotic, antioxidant and anti-inflammatory effect. Accordingly, two days after tMCAo, chronic treatment with CGS21680 reduced the number of infiltrated blood cells in the ischemic areas (Melani et al., 2014).

Most recently, we found similar results by studying the effect of BAY60-6583 (administered twice/day for 7 days in the tMCAo model), a selective agonist of the other A2R subtype, that the A2BR in most cases is coexpressed with A2ARs on hematic cells where it inhibits vascular adhesion (Yang et al., 2006) and migration of inflammatory cells (Wakai et al., 2001; Konrad et al., 2012). Two days after ischemia, the A2BR agonist reduced blood cell infiltration in the ischemic cortex (Dettori et al., 2020). Interestingly, 7 days after ischemia, the A2BR agonist also decreased TNF-α and increased IL-10 levels in the blood (Dettori et al., 2020). Both factors are considered valuable blood markers of the brain damage following an ischemic insult (Jickling and Sharp, 2011). These results stress the key research questions of the predictive value of blood biomarkers in stroke.

By and large, results underlie that, after hypoxia/ischemia, brain injury results from a complex sequence of pathophysiological events that evolve over time: a primary acute mechanism of excitotoxicity and periinfarct depolarizations followed by a secondary brain injury activation triggered by protracted neuroinflammation (Coppi et al., 2020b). Information acquired up to now indicate that adenosine A2Rs located on any cell type of the brain and on vascular and blood cells partake in either salvage or demise of the tissue after a stroke. Thus, they all represent important targets for drugs having different therapeutic time-windows after stroke.

Role of Adenosine A3Rs in Pain Control

Most recently, our group addressed the study of adenosine-mediated mechanisms of pain control. It is known that agonists at A3R subtype are effective pain suppressors in animal models of chronic constriction injury, chemotherapic pain (Janes et al., 2016), and also in colitis-induced visceral hypersensitivity (Lucarini et al., 2020). Their safe pharmacological profile, as shown by clinical trials for other pathologies, i.e., rheumatoid arthritis, psoriasis, and cancer, confers a realistic translational potential, thus encouraging research studies on the molecular mechanisms underpinning their antinociceptive actions. A number of pathways, involving central or peripheral mechanisms, have been proposed.

Our group recently demonstrated that the prototypical A3R agonist Cl-IB-MECA and the new, highly selective, A3R agonist MRS5980 (Tosh et al., 2014) inhibit the neuronal N-type voltage-dependent Ca2+ current in DRG neurons, a known pain-related current, more efficiently than the A1R agonist CPA (Coppi et al., 2019). Indeed, current-clamp experiments confirmed that Cl-IB-MECA significantly decreased the DRG neuronal firing (Coppi et al., 2019).

Our findings contributed to unveil one of the mechanisms of A3R-based pain control and reinforce the concept of A3R agonists as novel, promising, nonnarcotic agents for chronic pain relief.

Discussion

On the whole, our recent findings support the notion of adenosine receptors being involved in a number of central and peripheral nervous system diseases, from stroke and demyelination to neuropathic pain. Beyond the well-known protective effect of A1Rs, whose agonists are unfortunately not devoid of side effects, A2R- and A3R-selective ligands are attractive tools to develop innovative therapeutic strategies. Indeed, A2R antagonists exert significant neuroprotection in the initial (within 24 h) postischemic damage in the brain due to inhibition of glutamate excitotoxicity. Additional neuroprotection by A2AR and/or A2BR antagonists could be due to a promyelinating effect exerted by these compounds by stimulating OPC differentiation. On the other hand, at later phases (i.e., 7 days) after stroke, A2R agonists may attenuate neuroinflammation and immune cell infiltration to reduce brain damage. Finally, recent results emerging from our group revealed one of the mechanisms responsible for A3R-mediated pain control, i.e., inhibition of N-type Ca2+ currents and electrical activity in primary sensory neurons of the DRG, thus supporting these receptors as innovative nonopioid compounds for the treatment of chronic pain.

Author Contributions

EC, ID, AMP, and FP wrote the paper. FC, EC, ID, MV, and IB performed and analyzed experiments described in the paper. AMP, EC, and FP provided funding. AMP and FP supervised the work.

Funding

The study was supported by the University of Florence (Fondi Ateneo Ricerca) to AMP, the Ministry of Education, Universities and Research, Italy: MIUR-PRIN 2017CY3J3W_003 to FP, Fondazione Umberto Veronesi to EC (FUV 2020-3299), and Fondazione Italiana Sclerosi Multipla (FISM): 2019/R-Single/036 (AMP) to AMP and EC.

Conflict of Interest

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.

References

Betti, M., Catarzi, D., Varano, F., Falsini, M., Varani, K., Vincenzi, F., et al. (2018). The aminopyridine-3,5-dicarbonitrile core for the design of new non-nucleoside-like agonists of the human adenosine A(2B) receptor. Eur. J. Med. Chem. 150, 127–139. doi:10.1016/j.ejmech.2018.02.081

PubMed Abstract | CrossRef Full Text | Google Scholar

Burnstock, G., and Dale, N. (2015). Purinergic signalling during development and ageing. Purinergic Signal. 11, 277–305. doi:10.1007/s11302-015-9452-9

PubMed Abstract | CrossRef Full Text | Google Scholar

Cherchi, F., Pugliese, A. M., and Coppi, E. (2020). Oligodendrocyte precursor cell maturation: role of adenosine receptors. Neural. Regen. Res. 9, 155. doi:10.3389/fncel.2015.00155

CrossRef Full Text | Google Scholar

Colotta, V., Lenzi, O., Catarzi, D., Varano, F., Squarcialupi, L., Costagli, C., et al. (2012). 3-Hydroxy-1H-quinazoline-2,4-dione derivatives as new antagonists at ionotropic glutamate receptors: molecular modeling and pharmacological studies. Eur. J. Med. Chem. 54, 470–482. doi:10.1016/j.ejmech.2012.05.036

PubMed Abstract | CrossRef Full Text | Google Scholar

Coppi, E., Cellai, L., Maraula, G., Dettori, I., Melani, A., Pugliese, A. M., et al. (2015). Role of adenosine in oligodendrocyte precursor maturation. Front. Cell. Neurosci. 9, 155. doi:10.3389/fncel.2015.00155

PubMed Abstract | CrossRef Full Text | Google Scholar

Coppi, E., Cellai, L., Maraula, G., Pugliese, A. M., and Pedata, F. (2013a). Adenosine A(2A) receptors inhibit delayed rectifier potassium currents and cell differentiation in primary purified oligodendrocyte cultures. Neuropharmacology. 73, 301–310. doi:10.1016/j.neuropharm.2013.05.035

PubMed Abstract | CrossRef Full Text | Google Scholar

Coppi, E., Cherchi, F., Fusco, I., Failli, P., Vona, A., Dettori, I., et al. (2019). Adenosine A3 receptor activation inhibits pronociceptive N-type Ca2+ currents and cell excitability in dorsal root ganglion neurons. Pain 160 (5), 1103–1118. doi:10.1097/j.pain.0000000000001488

PubMed Abstract | CrossRef Full Text | Google Scholar

Coppi, E., Cherchi, F., Fusco, I., Dettori, I., Gaviano, L., Magni, G., et al. (2020a). Adenosine A2B receptors inhibit K+ currents and cell differentiation in cultured oligodendrocyte precursor cells and modulate sphingosine-1-phosphate signaling pathway. Biochem. Pharmacol. 177, 113956. doi:10.1016/j.bcp.2020.113956

PubMed Abstract | CrossRef Full Text | Google Scholar

Coppi, E., Dettori, I., Cherchi, F., Bulli, I., Venturini, M., Lana, D., et al. (2020b). A2B adenosine receptors: when outsiders may become an attractive target to treat brain ischemia or demyelination. Int. J. Mol. Sci. doi:10.3390/ijms21249697

CrossRef Full Text | Google Scholar

Coppi, E., Maraula, G., Fumagalli, M., Failli, P., Cellai, L., Bonfanti, E., et al. (2013b). UDP-glucose enhances outward K plus currents necessary for cell differentiation and stimulates cell migration by activating the GPR17 receptor in oligodendrocyte precursors. Glia. 61, 1155–1171. doi:10.1002/glia.22506

PubMed Abstract | CrossRef Full Text | Google Scholar

Coppi, E., Pedata, F., and Gibb, A. J. (2012). P2Y(1) receptor modulation of Ca2+-activated K+ currents in medium-sized neurons from neonatal rat striatal slices. J. Neurophysiol. 107, 1009–1021. doi:10.1152/jn.00816.2009

PubMed Abstract | CrossRef Full Text | Google Scholar

Coppi, E., Pugliese, A. M., Stephan, H., Muller, C. E., and Pedata, F. (2007a). Role of P2 purinergic receptors in synaptic transmission under normoxic and ischaemic conditions in the CA1 region of rat hippocampal slices. Purinergic Signal. 3, 203–219. doi:10.1007/s11302-006-9049-4

PubMed Abstract | CrossRef Full Text | Google Scholar

Coppi, E., Pugliese, A. M., Urbani, S., Melani, A., Cerbai, E., Mazzanti, B., et al. (2007b). ATP modulates cell proliferation and elicits two different electrophysiological responses in human mesenchymal stem cells. Stem Cell. 25, 1840–1849. doi:10.1634/stemcells.2006-0669

PubMed Abstract | CrossRef Full Text | Google Scholar

de Castro, F., and Bribian, A. (2005). The molecular orchestra of the migration of oligodendrocyte precursors during development. Brain Res. Rev. 49, 227–241. doi:10.1016/j.brainresrev.2004.12.034

PubMed Abstract | CrossRef Full Text | Google Scholar

Dettori, I., Gaviano, L., Ugolini, F., Lana, D., Bulli, I., Magni, G., et al. (2020). Protective effect of adenosine A2B receptor agonist, BAY60-6583, against transient focal brain ischemia in rat. Front. Pharmacol. 11, 1639. doi:10.1016/j.nbd.2017.08.011

CrossRef Full Text | Google Scholar

Fusco, I., Ugolini, F., Lana, D., Coppi, E., Dettori, I., Gaviano, , et al. (2018). The selective antagonism of adenosine A 2B receptors reduces the synaptic failure and neuronal death induced by oxygen and glucose deprivation in Rat CA1 hippocampus in vitro. Front. Pharmacol. 9, 399. doi:10.3389/fphar.2018.00399

PubMed Abstract | CrossRef Full Text | Google Scholar

Fusco, I., Cherchi, F., Catarzi, D., Colotta, V., Varano, F., Pedata, F., et al. (2019). Functional characterization of a novel adenosine A2B receptor agonist on short-term plasticity and synaptic inhibition during oxygen and glucose deprivation in the rat CA1 hippocampus. Brain Res. Bull. 151, 174–180. doi:10.1016/j.brainresbull.2019.05.018

PubMed Abstract | CrossRef Full Text | Google Scholar

Gallo, V., Zhou, J. M., McBain, C. J., Wright, P., Knutson, P. L., and Armstrong, R. C. (1996). Oligodendrocyte progenitor cell proliferation and lineage progression are regulated by glutamate receptor-mediated K+ channel block. J. Neurosci. 16, 2659–2670. doi:10.1523/JNEUROSCI.16-08-02659.1996

PubMed Abstract | CrossRef Full Text | Google Scholar

Gonçalves, F. Q., Pires, J., Pliassova, A., Beleza, R., Lemos, C., Marques, J. M., et al. (2015). Adenosine A2B receptors control A1 receptor-mediated inhibition of synaptic transmission in the mouse hippocampus. Eur. J. Neurosci. 41 (7), 878–88. doi:10.1111/ejn.12851

PubMed Abstract | CrossRef Full Text | Google Scholar

Janes, K., Symons-Liguori, A. M., Jacobson, K. A., and Salvemini, D. (2016). Identification of A(3) adenosine receptor agonists as novel non-narcotic analgesics. Br. J. Pharmacol. 173, 1253–1267. doi:10.1111/bph.13446

PubMed Abstract | CrossRef Full Text | Google Scholar

Jickling, G. C., and Sharp, F. R. (2011). Blood biomarkers of ischemic stroke. Neurotherapeutics. 8, 349–360. doi:10.1007/s13311-011-0050-4

PubMed Abstract | CrossRef Full Text | Google Scholar

Jung, C. G., Kim, H. J., Miron, V. E., Cook, S., Kennedy, T. E., Foster, C. A., et al. (2007). Functional consequences of S1P receptor modulation in rat oligodendroglial lineage cells. Glia. 55, 1656–1667. doi:10.1002/glia.20576

PubMed Abstract | CrossRef Full Text | Google Scholar

Konrad, F. M., Witte, E., Vollmer, I., Stark, S., and Reutershan, J. (2012). Adenosine receptor A2B on hematopoietic cells mediates LPS-induced migration of PMNs into the lung interstitium. Am. J. Physiol. Lung Cell Mol. Physiol. 303, L425–L438. doi:10.1152/ajplung.00387.2011

PubMed Abstract | CrossRef Full Text | Google Scholar

Levine, J. M., Reynolds, R., and Fawcett, J. W. (2001). The oligodendrocyte precursor cell in health and disease. Trends Neurosci. 24, 39–47. doi:10.1016/s0166-2236(00)01691-x

PubMed Abstract | CrossRef Full Text | Google Scholar

Lucarini, E., Coppi, E., Micheli, L., Parisio, C., Vona, A., Cherchi, F., et al. (2020). Acute visceral pain relief mediated by A(3)AR agonists in rats: involvement of N-type voltage-gated calcium channels. Pain. 161, 2179–2190. doi:10.1097/j.pain.0000000000001905

PubMed Abstract | CrossRef Full Text | Google Scholar

Maraula, G., Lana, D., Coppi, E., Gentile, F., Mello, T., Melani, A., et al. (2014). The selective antagonism of P2X(7) and P2Y(1) receptors prevents synaptic failure and affects cell proliferation induced by oxygen and glucose deprivation in rat dentate gyrus. PLoS One. 9, e115273. doi:10.1371/journal.pone.0115273

PubMed Abstract | CrossRef Full Text | Google Scholar

Maraula, G., Traini, C., Mello, T., Coppi, E., Galli, A., Pedata, F., et al. (2013). Effects of oxygen and glucose deprivation on synaptic transmission in rat dentate gyrus:.Role of A(2A) adenosine receptors. Neuropharmacology. 67, 511–520. doi:10.1016/j.neuropharm.2012.12.002

PubMed Abstract | CrossRef Full Text | Google Scholar

Melani, A., Cipriani, S., Vannucchi, M. G., Nosi, D., Donati, C., Bruni, P., et al. (2009). Selective adenosine A(2a) receptor antagonism reduces JNK activation in oligodendrocytes after cerebral ischaemia. Brain. 132, 1480–1495. doi:10.1093/brain/awp076

PubMed Abstract | CrossRef Full Text | Google Scholar

Melani, A., Corti, F., Cellai, L., Vannucchi, M. G., and Pedata, F. (2014). Low doses of the selective adenosine A(2A) receptor agonist CGS21680 are protective in a rat model of transient cerebral ischemia. Brain Res. 1551, 59–72. doi:10.1016/j.brainres.2014.01.014

PubMed Abstract | CrossRef Full Text | Google Scholar

Melani, A., Gianfriddo, M., Vannucchi, M. G., Cipriani, S., Baraldi, P. G., Giovannini, M. G., et al. (2006). The selective A(2A) receptor antagonist SCH 58261 protects from neurological deficit, brain damage and activation of p38 MAPK in rat focal cerebral ischemia. Brain Res. 1073, 470–480. doi:10.1016/j.brainres.2005.12.010

PubMed Abstract | CrossRef Full Text | Google Scholar

Melani, A., Pantoni, L., Bordoni, F., Gianfriddo, M., Bianchi, L., Vannucchi, M. G., et al. (2003). The selective A(2A) receptor antagonist SCH 58261 reduces striatal transmitter outflow, turning behavior and ischemic brain damage induced by permanent focal ischemia in the rat. Brain Res. 959, 243–250. doi:10.1016/s0006-8993(02)03753-8

PubMed Abstract | CrossRef Full Text | Google Scholar

Othman, T., Yan, H. L., and Rrvkees, S. A. (2003). Oligodendrocytes express functional A1 adenosine receptors that stimulate cellular migration. Glia. 44, 166–172. doi:10.1002/glia.10281

PubMed Abstract | CrossRef Full Text | Google Scholar

Pedata, F., Dettori, I., Coppi, E., Melani, A., Fusco, I., Corradetti, R., et al. (2016). Purinergic signalling in brain ischemia. Neuropharmacol. 104, 105–30. doi:10.1016/j.neuropharm.2015.11.007

CrossRef Full Text | Google Scholar

Pugliese, A. M., Coppi, E., Spalluto, G., Corradetti, R., and Pedata, F. (2006). A(3) adenosine receptor antagonists delay irreversible synaptic failure caused by oxygen and glucose deprivation in the rat CA1 hippocampus in vitro. Br. J. Pharmacol. 147, 524–532. doi:10.1038/sj.bjp.0706646

PubMed Abstract | CrossRef Full Text | Google Scholar

Pugliese, A. M., Traini, C., Cipriani, S., Gianfriddo, M., Mello, T., Giovannini, M. G., et al. (2009a). The adenosine A(2A) receptor antagonist ZM241385 enhances neuronal survival after oxygen-glucose deprivation in rat CA1 hippocampal slices. Br. J. Pharmacol. 157, 818–830. doi:10.1111/j.1476-5381.2009.00218.x

PubMed Abstract | CrossRef Full Text | Google Scholar

Pugliese, A. M., Trincavelli, M. L., Lecca, D., Coppi, E., Fumagalli, M., Ferrario, S., et al. (2009b). Functional characterization of two isoforms of the P2Y-like receptor GPR17: S-35 GTP gamma S binding and electrophysiological studies in 1321N1 cells. Am. J. Physiol. Cell Physiol. 297, C1028–C1040. doi:10.1152/ajpcell.00658.2008

PubMed Abstract | CrossRef Full Text | Google Scholar

Regehr, W. G. (2012). Short-term presynaptic plasticity. Cold Spring Harb. Perspect. Biol. 4(7), a005702. doi:10.1101/cshperspect.a005702

PubMed Abstract | CrossRef Full Text | Google Scholar

Sebastiao, A. M., and Ribeiro, J. A. (2009). Triggering neurotrophic factor actions through adenosine A2A receptor activation: implications for neuroprotection. Br. J. Pharmacol. 158, 15–22. doi:10.1111/j.1476-5381.2009.00157.x

PubMed Abstract | CrossRef Full Text | Google Scholar

Sollevi, A. (1986) Cardiovascular effects OF adenosine IN man—possible clinical implications. Progress in Neurobiology. 27, 319-349. doi:10.1016/0301-0082(86)90005-5

PubMed Abstract | CrossRef Full Text | Google Scholar

Stevens, B., Porta, S., Haak, L. L., Gallo, V., and Fields, R. D. (2002). Adenosine: a neuron-glial transmitter promoting myelination in the CNS in response to action potentials. Neuron. 36, 855–868. doi:10.1016/s0896-6273(02)01067-x

PubMed Abstract | CrossRef Full Text | Google Scholar

Tosh, D. K., Finley, A., Paoletta, S., Moss, S. M., Gao, Z. G., Gizewski, E. T., et al. (2014). In Vivo phenotypic screening for treating chronic neuropathic pain: modification of C2-arylethynyl group of conformationally constrained A(3) adenosine receptor agonists. J. Med. Chem. 57, 9901–9914. doi:10.1021/jm501021n

PubMed Abstract | CrossRef Full Text | Google Scholar

Vonlubitz, D., Lin, R. C. S., Melman, N., Ji, X. D., Carter, M. F., and Jacobson, K. A. (1994). Chronic administration OF selective adenosine A(1) receptor agonist or antagonist IN cerebral-ischemia. Eur. J. Pharmacol. 256, 161–167. doi:10.1016/0014-2999(94)90241-0

PubMed Abstract | CrossRef Full Text | Google Scholar

Wakai, A., Wang, J. H., Winter, D. C., Street, J. T., O'Sullivan, R. G., and Redmond, H. P. (2001). Adenosine inhibits neutrophil vascular endothelial growth factor release and transendothelial migration via A(2B) receptor activation. Shock. 15, 297–301. doi:10.1097/00024382-200115040-00008

PubMed Abstract | CrossRef Full Text | Google Scholar

Yang, D., Zhang, Y., Nguyen, H. G., Koupenova, M., Chauhan, A. K., Makitalo, M., et al. (2006). The A(2B) adenosine receptor protects against inflammation and excessive vascular adhesion. J. Clin. Invest. 116, 1913–1923. doi:10.1172/JCI27933

PubMed Abstract | CrossRef Full Text | Google Scholar

Keywords: cerebral ischemia, oxygen-glucose deprivation, adenosine receptors, oligodendrocyte differentiation, neuropathic pain, dorsal root ganglion neurons, neuroinflammation

Citation: Coppi E, Dettori I, Cherchi F, Bulli I, Venturini M, Pedata F and Pugliese AM (2021) New Insight into the Role of Adenosine in Demyelination, Stroke and Neuropathic Pain. Front. Pharmacol. 11:625662. doi: 10.3389/fphar.2020.625662

Received: 06 November 2020; Accepted: 18 December 2020;
Published: 29 January 2021.

Edited by:

Francisco Ciruela, University of Barcelona, Spain

Reviewed by:

Filippo Caraci, University of Catania, Italy

Copyright © 2021 Coppi, Dettori, Cherchi, Bulli, Venturini, Pedata and Pugliese. 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(s) 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: Elisabetta Coppi, elisabetta.coppi@unifi.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.