This is an open access publication, available online and distributed under the terms of a Creative Commons Attribution-Non Commercial-No Derivatives 4.0 International licence (CC BY-NC-ND 4.0), a copy of which is available at https://creativecommons.org/licenses/by-nc-nd/4.0/. Subject to this license, all rights are reserved.
NCBI Bookshelf. A service of the National Library of Medicine, National Institutes of Health.
Noebels JL, Avoli M, Rogawski MA, et al., editors. Jasper's Basic Mechanisms of the Epilepsies. 5th edition. New York: Oxford University Press; 2024. doi: 10.1093/med/9780197549469.003.0073
Abstract
Despite the availability of numerous antiseizure drugs (ASDs), treatment of epilepsy remains a clinical challenge, with over 30% of patients not responding to any pharmacological intervention. In addition, ASDs are mainly symptomatic without significantly impacting on disease progression. Thus, there is a strong focus on the identification of drug targets with a novel mechanism of action that are independent on GABAergic and glutamatergic neurotransmission and that possess a disease-modifying potential. That extracellular released purines such as adenosine triphosphate (ATP) and adenosine act as signaling molecules in the brain has been known for decades. While the anticonvulsant potential of adenosine is well established, increasing evidence suggests also an important role for ATP during the generation of seizures and the development of epilepsy. Usually present at low extracellular concentrations, ATP is released in response to several pathological stimuli including increased neuronal activity acting as danger signal and activating specific purinergic P2 receptors divided into the metabotropic P2Y and ionotropic P2X receptors. Suggesting a role during seizures and epilepsy, data from experimental models and patients show widespread expression changes of both P2Y and P2X receptor family members during epilepsy and, critically, drugs targeting both receptor subtypes, in particular, the P2Y1 and P2X7 receptors, confer both anticonvulsive and antiepileptic effects. This chapter provides a detailed summary of the current evidence suggesting ATP-gated receptors as novel drug targets for epilepsy, discusses the possible mechanisms for how these receptors contribute to increased hyperexcitability, and briefly outlines how the purinergic system can be used as novel tool for the diagnosis of epilepsy.
Purinergic Signaling in the Brain
Compelling and exciting evidence gathered over the past decades has demonstrated a role for purinergic signaling in the generation of seizures and the development of epilepsy. These discoveries provide new promising therapeutic avenues for the treatment of drug-refractory seizures and epilepsy. As the field of purinergic signaling is fast moving and many readers may not be familiar with it, we start our review by considering the mechanisms of purinergic signaling that are likely to be relevant to the development and regulation of seizures.
The purinergic signaling system is similar in some ways to other neurotransmitter systems in the brain. However, it is marked out from other possibly more familiar systems by a complexity that encompasses the multiplicity of possible ligands, release pathways, receptor systems, and the fact that the inactivation products are signaling agents in their own right (Fig. 73–1). Adenosine triphosphate (ATP) and adenosine comprise the canonical purinergic ligands. However, there are other compounds present in the central nervous system (CNS) that can be released or generated in a regulated fashion and can activate some of the purinergic receptors: Adenosine diphosphate (ADP), diadenosine polyphosphates (Pintor et al., 1992; Miras-Portugal et al., 1999), uridine triphosphate (UTP), uridine diphosphate (UDP), and UDP-glucose (Moore et al., 2003; Lazarowski, 2006).

Figure 73–1.
Scheme that shows the major elements of purinergic signaling. There are two major mechanisms of release: via gap junction hemichannels (HC); and exocytosis. ATP is sequestered in vesicles via the vesicular nucleotide transporter. ATP can also be released (more...)
ATP Receptors
The basic outlines of the pharmacology of ATP receptors were drawn in 1985 by Burnstock and Kennedy, who proposed that there were two types of receptors for ATP, which they called P2X receptors (P2XRs) and P2Y receptors (P2YRs) (Burnstock and Kennedy, 1985; Kennedy, 2021). Nevertheless, it took nearly a decade to achieve the first molecular cloning of ATP receptors: in 1993 for the first P2YR (Webb et al., 1993); and in 1994 for the first P2XR (Valera et al., 1994; Brake et al., 1994). After these initial breakthroughs, cloning through sequence homology of other related receptors rapidly took place. We now recognize eight different P2YR subtypes that have the typical seven-transmembrane segment of G protein–coupled receptors (GPCRs). Of these P2Y2, P2Y11 are activated best by ATP, and others are selective for ADP (P2Y1, P2Y12, and P2Y13), UTP/UDP (P2Y4, P2Y6), and the ligand UDP-glucose (P2Y14) (Ralevic and Burnstock, 1998; von Kugelgen, 2021). The P2XRs form a ligand-gated ion channel that has significant Ca2+ permeability. Unlike the multiplicity of endogenous ligands that can act on the P2YR family, ATP is the unique endogenous ligand for the P2XR. There are seven different subtypes of the P2XR family (Khakh et al., 2001), which form a trimeric structure that can be either homomeric or heteromeric. There are now high-resolution structures for P2 receptors. The first X-ray crystal structure for a P2XR was published in 2009 (Kawate et al., 2009), confirming its unique trimeric structure among the ligand-gated ion channels, and further work on this structure has revealed interesting and unusual features of ion permeation through this channel (Hattori and Gouaux, 2012; Karasawa and Kawate, 2016; Mansoor et al., 2016). X-ray structures have also been obtained for P2YRs (Zhang et al., 2014, 2015). These advances mean that the rational design of drugs targeting receptors for ATP is now possible (Ciancetta and Jacobson, 2018).
Adenosine Receptors
Adenosine receptors (ARs), alternatively called P1 receptors, are a family of G protein–coupled receptors comprised of four subtypes: A1R, A2AR, A2BR, and A3R (Dunwiddie et al., 1997; B. B. Fredholm, 1999; Benarroch, 2008). ARs are widely distributed throughout the body, including the CNS, with each receptor, however, showing differences in expression (tissue and cell type), downstream signaling, physiological effects, and responses during pathology when compared to the remaining ARs (Peleli et al., 2017). All ARs are activated by their endogenous ligand, adenosine, with each AR showing, however, slight differences in their affinity for adenosine [A1Rs (1–10 nM) > A2ARs (30 nM) > A3Rs (100 nM) > A2BR (1000 nM)] (Fredholm et al., 2011; Borea et al., 2018). In terms of downstream signaling, whereas A1Rs and A3Rs are coupled to inhibitory Gi/o proteins, A2ARs and A2BRs are coupled to stimulatory Gs proteins (Borea et al., 2018). Unlike for ATP, there is no ligand-gated receptor for adenosine.
Interconversion of Endogenous Purinergic Ligands
The actions of a neurotransmitter must be terminated to give temporal specificity of signaling. This was recognized from the earliest development of the chemical hypothesis of neurotransmission (Werman, 1966). For some transmitters this is simply diffusion away from the synaptic cleft (the site of release and action) and reuptake into neurons and glia. Burnstock observed in his 1972 review, which set the stage for purinergic signaling, that an inactivating mechanism is essential to terminate the actions of ATP as a neurotransmitter (Burnstock, 1972). There is now known to be an imposing array of extracellular enzymes capable of metabolizing ATP and its downstream products. CD39, a lymphocyte marker, was cloned in 1994 (Maliszewski et al., 1994). At that time, however, its biological function was not established, although the authors drew attention to similarities with proteins known to hydrolyze GTP. In 1996, Robson and colleagues realized that CD39 was an ectoATPase and established the ENTPDase (ecto-nucleoside triphosphate diphosphohydrolase) family (Kaczmarek et al., 1996; Chadwick and Frischauf, 1998). This family has eight members, of which four are found on the plasma membrane (Robson et al., 2006). NTPDase1 has roughly equal enzymatic activity for ATP and ADP; that is, it will convert ATP to AMP. However, NTPDase2 acts only on ATP and has no activity toward ADP. NTPDase 3 falls between these two extremes. It should be remembered that the ENTPDases have quite broad activity and will hydrolyze any nucleoside tri- or diphosphate. There are a number of circumstances during development and in regions of the brain where alkaline phosphatases (e.g., phosphatidate phosphatase [PAP] and tissue-nonspecific alkaline phosphatase [TNAP]) have significant roles (Langer et al., 2007; Zimmermann, 2009; Diez-Zaera et al., 2011). Interestingly TNAP and PAP were first cloned in 1988 (Henthorn et al., 1988; Vihko et al., 1988) and the first nucleotide pyrophosphatase (NPPase) in 1992 (Funakoshi et al., 1992). The NPPase family (Stefan et al., 2005) is now known to have seven members with very broad substrate specificity. The final enzyme of interest is CD73 or the ecto-5 nucleotidase which converts a nucleoside monophosphate such as AMP to the corresponding nucleoside. CD73 is important in brain and, in several regions, can mediate most of the generation of adenosine from previously released ATP (Klyuch et al., 2012; Lovatt et al., 2012; Wall and Dale, 2013).
It is too simplistic to regard the function of this army of ectoenzymes capable of hydrolyzing ATP as being only to terminate the actions of ATP. It is now clear that they initiate the downstream actions of the products at their cognate receptors—notably ADP, which can act at P2Y1, P2Y12, and P2Y13Rs, and adenosine, which has its own receptor system. The interconversion of ATP to adenosine is particularly relevant in the context of epilepsy. ATP is excitatory (via P2XRs, and some subtypes of P2YRs) and may be pro-convulsant (as discussed in the next section). By contrast, adenosine acting via A1Rs is inhibitory and has anticonvulsant actions. The spatial and temporal dynamics of the interconversion of ATP to adenosine can be unexpectedly complex, given that ATP and ADP can inhibit CD73—known as feed-forward inhibition. This gives a delay in the production of adenosine from the previously released ATP (James and Richardson, 1993; Dale, 1998), which in the context of generation of motor rhythms in the spinal cord separates the excitatory actions of ATP from the later inhibitory actions of adenosine (Dale and Gilday, 1996; Dale, 1998). More recently it has been shown via simulations of the diffusion and production of adenosine in the extracellular space that the enzymatic interconversion of ATP and adenosine, especially when combined with the feed-forward inhibition of CD73, can physically separate the actions of ATP and adenosine, such that adenosine reaches its highest concentrations some considerable distance away from the source of ATP release from which it originated (Masse and Dale, 2012; Dale, 2021). Given the respectively pro- and anticonvulsant actions of ATP and adenosine, the spatial and temporal dynamics of ATP breakdown to ADP and adenosine is likely to be important in the regulation of seizure activity. Microglia express both CD39 and CD73 (Dalmau et al., 1998). Microglia are highly motile cells that migrate and send processes toward sites of neural insult. ATP released from distressed cells acts as a microglial chemoattractant (Davalos et al., 2005). CD39 on microglial processes metabolizes the released ATP to AMP with further conversion to adenosine via CD73. Because microglia are motile, this mechanism gives dynamic control to the interconversion of ATP and adenosine, and this is important in mediating activity-dependent feedback via adenosine A1Rs (Badimon et al., 2020), a concept further explored later in this chapter.
Sources and Release
ATP is present in every cell at millimolar concentrations. In principle, therefore, any cell in the CNS could potentially serve as a source of extracellular ATP. The sources and mechanisms of ATP release have remained an active area of investigation, and here we summarize the current state of knowledge.
Vesicular Release of ATP
The identification of the orphan transporter SLC17A9 as a vesicular nucleotide transporter (VNUT) (Sawada et al., 2008) confirms the importance of vesicular exocytosis as a release mechanism for ATP and provides a unique marker to identify cells that use ATP as a transmitter. Surprisingly, however, release from vesicles is not the only exocytotic pathway—there may also be exocytosis of ATP from lysosomes (Zhang et al., 2007).
The first description of ATP acting as a principal transmitter in the CNS was published in 1992 (Edwards et al., 1992). Despite this development, most think now that this synapse is rather unusual and that ATP signaling in the CNS most often occurs as a cotransmitter (originally proposed by Burnstock [1976]) or is involved in nonsynaptic signaling between a variety of non-neuronal cells. ATP has been described as a cotransmitter with glutamate (Pankratov et al., 2006, 2007) and γ-aminobutyric acid (GABA) (Jo and Schlichter, 1999; Hugel and Schlichter, 2000). ATP is also released from non-neuronal cells. In the CNS, astrocytes can release ATP, definitively demonstrated by the Haydon group in 2000 (Wang et al., 2000b), most probably by exocytosis. This ATP release stimulates Ca2+ signaling waves through neighboring astrocytes which can in turn trigger release of other gliotransmitters (e.g., glutamate, D-serine) from astrocytes. ATP signaling is intimately bound up with the concept of the tripartite synapse—that central synapses consist of the pre- and postsynaptic element plus a closely associated astrocytic element that effectively “listens” to the synaptic conversation and can, through release of ATP and other transmitters, modulate this conversation (Haydon, 2001). A key aspect of this modulatory action by astrocytic release of ATP concerns conversion of ATP to adenosine, which in turn impacts on synaptic function and integrative behaviors such as the control of sleep (Halassa et al., 2009). Other glial cells also release and respond to ATP; these include microglia and oligodendrocytes.
Channel-Mediated Release of ATP
Although exocytotic release of ATP is well established, it is now known that a variety of channels can release ATP directly: connexin hemichannels, pannexins, calcium homeostasis modulator 1 (CalHM1), volume-regulated anion channels, and even the P2X7R itself. For channel-mediated release, the driving force is the chemical gradient for the released moiety across the membrane. ATP is a favorable candidate for two reasons: (1) it is present in the cytosol at high concentrations (probably three orders of magnitude greater than that of the extracellular space); and (2) it is negatively charged: as cells have negative resting potentials, this will increase the driving force of ATP through a permeable membrane channel. The Nernstian equilibrium potential for ATP is +97 mV, which would give a driving force on ATP of about 150 mV at a typical resting membrane potential. Indeed, ATP may be preferentially released at potentials near the resting potential rather than during depolarization (Nualart-Marti et al., 2013).
In 2002, Stout et al. (2002, 2004) demonstrated that connexin gap junction hemichannels (a connexon comprised of six connexin subunits) could have functions of their own. Although it was disputed at the time that hemichannels had a distinctive signaling function, Stout et al. showed for the first time that connexin hemichannels could release ATP from cultured astrocytes (Stout et al., 2002). Following this, several other papers demonstrated hemichannel-mediated release of ATP under more physiological circumstances such as cortical development (Weissman et al., 2004) and retinal development (Pearson et al., 2005). At the same time, the number of hemichannels capable of releasing ATP has grown, with the addition of pannexin-1 and a number of connexin hemichannels (e.g., Cx43 and Cx26). In the taste bud sensory cells, there is strong evidence for the release of ATP to be mediated via pannexin-1 (Huang et al., 2007) and CalHM1 (Taruno et al., 2013). Hypothalamic tanycytes, a type of glial cell, sense glucose and amino acids. Their response to glucose is mediated partly via the sweet taste receptors described in the tongue (Benford et al., 2017) and involves release of ATP and activation of P2Y1Rs (Frayling et al., 2011; Benford et al., 2017). The ATP release appears to occur via gating of Cx43 hemichannels (Orellana et al., 2012). By contrast, amino acid sensing involves umami taste receptors and ATP release mediated via pannexin-1 and CalHM1 channels (Lazutkaite et al., 2017). ATP release via pannexin-1 also plays an important role in regulating vascular tone and in neurovascular coupling (Billaud et al., 2012; Thakore et al., 2021) and has been implicated in driving seizure-like activity in the hippocampus (Lopatar et al., 2015; Scemes et al., 2019). The physiological importance of connexin-mediated ATP release has been demonstrated in the context of CO2 chemosensitivity and the regulation of breathing. Here, a particular connexin, Cx26, serves not only as the conduit for ATP release but is itself directly sensitive to CO2 (Huckstepp et al., 2010a, 2010b; Meigh et al., 2013). CO2 is able to bind to a specific lysine residue via a labile carbamylation reaction (Meigh et al., 2013; Dospinescu et al., 2019). Cx26 can, therefore, be considered as a receptor for CO2, which when stimulated by CO2 opens to release ATP. By mutating the key residues that bind CO2 and expressing mutated Cx26 in vivo, it has been shown that this binding of CO2 to Cx26 is an important contributor to the chemosensory regulation of breathing (van de Wiel et al., 2020).
Volume-regulated anion channels are also an important conduit for ATP release. Recently a very elegant mechanism of communication in myelinated axons, from axon to oligodendrocyte, has been shown to be mediated by ATP released via a mechanosensitive channel present in the axon and which opens to release ATP during the action potential (Fields and Ni, 2010; Fields, 2011), thus providing a signal to link axon activity with activation of the oligodendrocytes that ensheath the axon. Interestingly, the Schwann cells also appear to release ATP via Cx32 hemichannels (Carrer et al., 2018; Nualart-Marti et al., 2013), suggesting complex two-way communication between axon and myelin sheath.
Mechanisms of Release and Production of Adenosine
Activity-dependent adenosine release is well established in the brain (Mitchell et al., 1993; Wall and Dale, 2007, 2008; Klyuch et al., 2012; Wall and Dale, 2013; Sims et al., 2013). As outlined above, an important source of extracellular adenosine derives from previously released ATP and its conversion via ectoenzymes in the extracellular space. However, adenosine can also be release from neurons directly via the equilibrative transporters ENT1 and ENT2 (Baldwin et al., 2004). This is important in a number of contexts: for example, during neuronal activity, adenosine can arise via this route as well as from the previously released ATP (Lovatt et al., 2012; Wall and Dale, 2013). In the hippocampus, this direct release of adenosine is Ca2+-dependent and comprises about half of the activity-dependent adenosine release. In other brain areas, there is a Na+ dependence to activity-dependent adenosine release (Sims et al., 2013; Sims and Dale, 2014). This appears to arise from activation of the Na+-K+ ATPase required to extrude the Na+ ions that have entered during signaling activity. The resulting consumption of ATP causes an intracellular accumulation of adenosine that then crosses the membrane via equilibrative transport (Sims and Dale, 2014). This mechanism of adenosine production may be significant in the context of seizure activity as the intense neuronal firing would cause very significant activation of the Na+-K+ ATPase and, hence, release of the adenosine and enhancement of its anticonvulsant action.
In cerebellum, there is evidence that adenosine may be released via exocytosis (Wall and Dale, 2007; Klyuch et al., 2012). The activity-dependent adenosine release is both tetrodotoxin- (TTX) and Ca2+-dependent, and, while some of it arises from previously released ATP, about half of activity-dependent adenosine release remains when CD73, the ecto-5′ nucleotidase, has been deleted (Klyuch et al., 2012). This remaining component of release appears to depend on vesicles as bafilomycin, the inhibitor of the vesicular proton pump that provides the electromotive force for vesicular transporters, greatly reduces activity-dependent adenosine release. This phenomenon has not yet been documented in other areas of the brain.
Targeting Purinergic Receptors for Seizure Control and the Treatment of Epilepsy
To capture the heterologous nature of epilepsy, researchers have developed a plethora of different approaches to model seizures and epilepsy, which includes the use of seizure-inducing chemoconvulsants (e.g., kainic acid [KA], pilocarpine or pentylenetetrazole [PTZ]), electrical stimulation (e.g., perforant pathway, hippocampus, amygdala), genetic mutations and injury models (e.g., TBI, WAG/Rij rats) with species spanning from flies to primates (Loscher, 2011, 2017) with each model mimicking different aspects of the disease. To date, however, investigation of the impact of purinergic signaling on seizures and epilepsy has largely been restricted to the use of chemoconvulsants and electrical stimulation in rodents using models of acute seizures (i.e., status epilepticus) and drug-refractory epilepsy. Of note, although some studies have started to investigate the role of purinergic signaling in epilepsies with a genetic cause (e.g., Dogan et al., 2020), temporal lobe epilepsy (TLE), which represents the most common form of epilepsy in adults, involving structures of the limbic system such as the hippocampus, and which is particularly prone to drug refractoriness (Thijs et al., 2019), has been the most widely studied. Current principal approaches include administration of the glutamate agonist KA, the cholinergic agonist pilocarpine, and the GABAA receptor inhibitor PTZ. For a detailed description of different models of epilepsy, please refer to Loscher (2011, 2017) or other reviews written on this topic. During this section we will first describe evidence showing how ATP is released during seizures and epilepsy. Next, we will provide a brief summary of the role of P1 adenosine receptors during epilepsy, which is then followed by a detailed description of the current knowledge on targeting ATP-gated P2 receptors.
ATP Release during Seizures and Epilepsy
A link between increased extracellular ATP levels and hyperexcitability was first provided in 1978 by Wu et al., showing that microinjections of ATP into the prepiriform cortex of rats caused motor seizures (Wu and Phillis, 1978) and in 1989 by Wierasko et al. by showing increased extracellular ATP concentrations in the brain of a seizure-prone strain of mice (inbred DBA/2 [D2]) (Wieraszko and Seyfried, 1989). In 2012, Heinrich et al. showed that ATP can be released during increased neuronal activity by using depolarizing high K+ concentrations in slices of rat hippocampus (Heinrich et al., 2012). Later, Lopartar et al. suggested this increase to be mediated via Pannexin-1 in rat hippocampal slices treated with the glutamate agonist ((S)-3,5-Dihydroxyphenylglycine) (Lopatar et al., 2015). Other methods, such as the stimulation of the Schaffer collateral, however, failed to increase extracellular ATP (Lopatar et al., 2011). Further evidence suggesting that Pannexin-1 activation contributes to increases in extracellular ATP during seizures has been provided by Dossie et al., who recently showed, using resected tissue from patients with epilepsy, that extracellular ATP increased 80% during high K+-induced ictal discharges and was suppressed by blocking Pannexin-1. The increased extracellular ATP may have a proconvulsant function as blockade of Pannexin-1 channels provided potent anticonvulsive effects during KA-induced seizures in mice (Dossi et al., 2018). In agreement with a proconvulsant function of extracellular adenosine nucleotides, injection of ATP, the ATP analog 2 ,3 -O-(4-benzoylbenzoyl) ATP (BzATP) or ADP into the lateral ventricle of mice caused high-amplitude, high-frequency polyspiking on the electroencephalogram (EEG) (ATP) (Sebastian-Serrano et al., 2016) and increased seizure severity during intra-amygdala KA-induced status epilepticus (BzATP and ADP) (Engel et al., 2012; Alves et al., 2017). By contrast, intracerebroventricular injection of uridine-5′-triphosphate (UTP) reduced seizure severity during intra-amygdala KA-induced status epilepticus in mice (Alves et al., 2017). In vivo evidence demonstrating increased extracellular ATP during seizures/epilepsy was provided by a study from Dona et al. Here, by using a rat model of intraperitoneal pilocarpine, the authors observed increased extracellular levels of ADP, adenosine monophosphate (AMP), and adenosine, but not ATP post status epilepticus. While the same purines were found to be decreased during epilepsy, their levels increased, including ATP, following an epileptic seizure (Dona et al., 2016). While it is widely accepted now that ATP is released during increased neuronal activity, it is important to keep in mind that, once released into the extracellular space, ATP is rapidly metabolized via several ectonucleotidases into different breakdown products including adenosine (Zimmermann, 2006). Thus, while ATP seems to exert a mainly pro-convulsant function, released ATP may also increase the extracellular pool of adenosine, thereby acting as anticonvulsant, as discussed in the next section.
Targeting Adenosine and P1 Receptors
While nucleotide-sensitive P2 receptors are the main topic of the present book chapter, we will provide a brief overview of what is known on the role of adenosine and adenosine-sensitive P1 receptors during seizures and epilepsy. For a more in-depth discussion on adenosine and epilepsy, please refer to these excellent reviews (Boison and Rho, 2019; Weltha et al., 2019; Tescarollo et al., 2020; Murugan et al., 2021).
Among the different ARs, it is well established that A1Rs exert a mainly anticonvulsive function, whereas A2ARs seem to be mainly proconvulsive. In contrast, the role of A2B and A3Rs remains to be fully established. Evidence suggesting a role of adenosine during seizures stems from early studies by During et al. in 1992 showing a 6- to 31-fold increase in extracellular adenosine levels in patients with epilepsy following a seizure when compared to baseline (During and Spencer, 1992). Subsequent studies confirmed this seizure-induced release of adenosine, which is now believed to constitute a protective feedback mechanism to limit seizure duration and the intensity or spread of focal seizures (Dulla et al., 2009; Ilie et al., 2012; Lovatt et al., 2012; Van Gompel et al., 2014). The anticonvulsant function of A1Rs is supported in mice in which the gene for A1Rs has been deleted. These mice exhibit spontaneous electrographic seizures and develop lethal status epilepticus following intrahippocampal KA administration (Fedele et al., 2006; Masino et al., 2011). A1R knockout mice also display increased seizure and TBI-induced neurodegeneration (Fedele et al., 2006; Kochanek et al., 2006), suggesting that these receptors play an important neuroprotective role. Pharmacological evidence was provided by showing seizure-suppressive effects of A1Rs agonists in several mouse models (Gouder et al., 2003; Li and Zhang, 2011; Li et al., 2013; Mares, 2010; Muzzi et al., 2013; Tosh et al., 2012; Vianna et al., 2005). Conversely, A1R antagonists increase seizure activity and decrease responsiveness to ASDs (Fukuda et al., 2010; Chwalczuk et al., 2008). In contrast to A1Rs, A2ARs seem to contribute to seizures rather than blocking them. A2ARs have been found upregulated at excitatory glutamatergic terminals in rodent models of epilepsy (i.e., amygdala-kindled rats and systemic KA-induced status epilepticus mouse model) and in hippocampal astrocytes of patients with mesial TLE (Barros-Barbosa et al., 2016a; Rebola et al., 2005). In line with a proconvulsive action of A2ARs, mice with a genetic inactivation of A2ARs have a reduced seizure susceptibility (El Yacoubi et al., 2001, 2008, 2009). Moreover, Canas et al. showed, by using a rat and mouse KA model of TLE, A2ARs contributing to neurodegeneration via the modulation of synaptic excitability following KA-induced seizures in rats and mice (Canas et al., 2018). While the majority of studies support a proconvulsive function, others have, however, also suggested an anticonvulsive function of A2ARs (Vianna et al., 2005; Adami et al., 1995). The exact roles of both A2B and A3Rs in epilepsy remain to be established. A3Rs have been suggested to counteract the anticonvulsive effects provided by A1Rs (Dunwiddie et al., 1997). Both A2B and A3Rs have been linked to the modulation of GABA currents. A2BR and A3R antagonists reduce the rundown of GABA currents (Roseti et al., 2008), thereby potentially reducing brain excitability and the occurrence of seizures.
Proposed adenosine-based therapies for epilepsy are, however, not restricted to the targeting of adenosine-sensitive P1 receptors. For example, one of the major routes for the removal of anticonvulsive adenosine is via the action of adenosine kinase (ADK), which is highly expressed on astrocytes and which catalyzes the phosphorylation of adenosine to 5′-AMP with the subsequent decreased concentration of adenosine in the extracellular space. Supporting a pro-convulsive role of ADK, inhibition of ADK suppresses acute, evoked seizures and seizures once epilepsy is established (Boison, 2013; Boison and Yegutkin, 2019; Sandau et al., 2019). Of note, adenosine and adenosine metabolizing enzymes have also been associated with epigenetic alterations (e.g., DNA methylation), which have been linked to the development of human and experimental epilepsy (Kobow et al., 2013; Miller-Delaney et al., 2015; Murugan et al., 2021).
P2 Receptors
P2 Receptor Expression Following Seizures and during Epilepsy
P2Rs are widely distributed throughout the brain where they are expressed and functional on both neurons and glial cells (Burnstock, 2013; Guzman and Gerevich, 2016). While a detailed description of potential expression changes during epilepsy is still missing for most of the P2Rs, substantial progress has been made for the P2Y1R and P2X7R subtype with data stemming from both experimental models and patients (Table 73–1). Expression analysis of brain tissue was, however, mainly limited to the hippocampus and the cortex.

Table 73–1
P2 Expression Changes and Impact of P2 Receptor-Targeting during Acute Seizures, Epileptogenesis, and Epilepsy.
Changes in the expression for P2XRs following seizures (i.e., status epilepticus) or during epilepsy have been reported for the P2X2, P2X4, and P2X7R, while no changes have been found so far in the expression for the remaining P2XRs (i.e., P2X1R, P2X3R, P2X5R, and P2X6R). Regarding the P2X2R and P2X4R, whereas hippocampal P2X2R expression has been shown to be downregulated following intra-amygdala KA (Engel et al., 2012), P2X4R expression increases in the hippocampus following systemic KA injections (Avignone et al., 2008; Ulmann et al., 2013), but not intra-amygdala KA- or pilocarpine-induced status epilepticus (Dona et al., 2009; Engel et al., 2012). In contrast, P2X4R expression was found decreased in the hippocampus of pilocarpine-treated epileptic rats (Dona et al., 2009).
Among the P2XR family, most data are available on the P2X7R subtype. P2X7R expression has consistently been reported to be increased following status epilepticus including intraperitoneal KA in mice (hippocampus) (Avignone et al., 2008), intra-amygdala KA-induced status epilepticus in mice (hippocampus, cortex, striatum, thalamus, cerebellum) (Engel et al., 2012; Jimenez-Pacheco et al., 2013; Morgan et al., 2020) and pilocarpine-induced status epilepticus in rats (hippocampus) (Dona et al., 2009). While increases in P2X7R expression are well established, the cell-type-specific expression pattern of P2X7R in the setting of seizures and epilepsy remains controversial, in line with a wider ongoing debate surrounding its expression in the CNS (Illes et al., 2017; Miras-Portugal et al., 2017). Using immunohistochemistry approaches, P2X7R expression has been detected at glutamatergic nerve terminals (Dona et al., 2009). Neuronal P2X7R expression has also been detected using a soluble enhanced green fluorescent protein (EGFP) bacterial artificial chromosome (BAC) transgenic P2X7R reporter mouse model (Engel et al., 2012; Jimenez-Pacheco et al., 2013). In this model EGFP is under the transcriptional control of the P2rx7 promoter. These results have been challenged by the use of a P2X7R reporter mice in which P2X7R is fused to EGFP. This approach detected P2X7R expression within the CNS mainly in microglia and oligodendrocytes (Kaczmarek-Hajek et al., 2018; Morgan et al., 2020). A recent study comparing both transgenic P2X7R reporter mouse lines has shown abnormal P2X7R expression and increased P2X4R expression in the soluble EGFP BAC transgenic P2X7R mouse model, questioning the validity of this reporter mouse (Ramirez-Fernandez et al., 2020). P2X7R expression has not been detected on astrocytes (Engel et al., 2012; Morgan et al., 2020). Similarly to status epilepticus, there is wide consensus that increased P2X7R expression occurs during epilepsy in the brain (hippocampus and cortex) with evidence from both rodent models and patients with TLE (Vianna et al., 2002; Dona et al., 2009; Barros-Barbosa et al., 2016b; Jimenez-Pacheco et al., 2013, 2016). Again, as in status epilepticus, P2X7R expression increases in microglia, neurons, and oligodendrocytes, but was not detected in astrocytes. Vianna et al. found P2X7R expression increased on mossy fibers and glutamatergic nerve terminals of epileptic pilocarpine-treated rats (Vianna et al., 2002; Dona et al., 2009). A neuronal P2X7R expression during epilepsy was confirmed later using the soluble EGFP BAC transgenic P2X7R mouse model (Jimenez-Pacheco et al., 2013, 2016). However, as for status epilepticus, neuronal expression of P2X7R during epilepsy could not be confirmed in a more recent study using the fused P2X7R-EGFP reporter mice (Morgan et al., 2020). Thus, while compelling evidence suggests P2X7R to be increased in microglia following status epilepticus and during epilepsy, it is still a matter of debate as to whether P2X7R expression increases in neurons.
Although less investigated, changes in the expression of the different P2YR subtypes have also been reported in both rodent models and patients (Alves et al., 2018). P2ry6, P2ry12, and P2ry13 mRNA transcripts were found increased in the hippocampus post-status epilepticus using the intraperitoneal KA mouse model (Avignone et al., 2008). A later study carried out by Alves et al. using the intra-amygdala KA mouse model reported that while mRNA levels of adenine nucleotide-sensitive receptors were decreased (P2ry1, P2ry12, and P2ry13), transcript levels of uracil nucleotide-sensitive receptors were increased (P2ry2, P2ry4, and P2ry6) in the hippocampus. When looking at the protein level, P2YRs coupled to Gq were increased (P2Y1R, P2Y2R, P2Y4R, and P2Y6R), and P2YRs coupled to Gi were downregulated or not changed (P2Y12R, P2Y13R, P2Y14R) (Alves et al., 2017), suggesting that receptor expression is dependent on substrate (mRNA) and receptor-downstream signaling (protein). In contrast to the hippocampus, however, P2YRs expression in the cortex was mainly upregulated post status epilepticus (Alves et al., 2019b). An upregulation of P2YRs was also the predominant response during epilepsy in both epileptic mice and patients (Sukigara et al., 2014; Alves et al., 2017). Very little is known regarding the cell-type-specific expression. Whereas P2Y1Rs have been shown to be increased on microglia post-intra-amygdala KA-induced status epilepticus (Alves et al., 2019a), P2Y2Rs and P2Y4Rs have been detected on astrocytes in brain tissue from patients with intractable epilepsy (Sukigara et al., 2014).
Targeting P2 Receptors during Epilepsy
P2X Receptors
Among the P2XRs, the P2X7R subtype has attracted by far the most attention as a potential therapeutic target for seizure suppression and treatment of epilepsy (Beamer et al., 2017) (Table 73–1). The only other two P2XRs for which functional data are available are P2X3R and P2X4R.
The first study suggesting anticonvulsive potential of targeting the P2X7R was published by Engel et al. in 2012. Here the authors showed that P2X7R deletion or blocking of the P2X7R (antagonists and P2X7R-targeting antibodies) reduced both seizure severity during status epilepticus and resulting neurodegeneration in the intra-amygdala KA mouse model (Engel et al., 2012; Jimenez-Pacheco et al., 2013). Suggesting the potential of P2X7R antagonists as adjunctive treatment for pharmaco-resistant status epilepticus, P2X7R antagonists potentiated effects of the anticonvulsant lorazepam (Engel et al., 2012). An anticonvulsive effect of P2X7R antagonism was confirmed in a more recent study using a rat model of coriaria lactone-induced status epilepticus (Huang et al., 2017). This anticonvulsive potential of targeting P2X7R, seems, however, heavily dependent on the model used. Fischer et al. showed no anticonvulsant effects provided by P2X7R antagonism using the maximal electroshock seizure threshold test and the PTZ seizure threshold test in mice. In the same study, the authors, however, observed that P2X7R antagonists potentiated anticonvulsant effects of carbamazepine in the maximal electroshock seizure test, supporting the potential of P2X7R-based treatment as adjunctive therapy (Fischer et al., 2016). Similar to these results, in a study published one year later, Nieoczym et al. (2017) only found a weak anticonvulsant effect of targeting the P2X7R in the intravenous pentylenetetrazole (PTZ) seizure threshold, maximal electroshock seizure threshold, and 6 Hz psychomotor seizure threshold tests. More recently, Dogan et al. showed no effect of P2X7R antagonism on seizures in WAG/Rij rats, a model of genetic absence epilepsy (Dogan et al., 2020). On the other hand, a pro-convulsive function of P2X7R is suggested by Kim et al., who reported in 2012 that seizure severity was exacerbated in P2X7R knockout mice subjected to intraperitoneal pilocarpine. This may be unique to the pilocarpine model, as P2X7 knockout mice treated with intraperitoneal KA showed no increase in seizure severity (Kim and Kang, 2011). In subsequent studies the same group showed, using a pilocarpine rat model, that P2X7R antagonism protected from astroglial cell death, reduced the infiltration of neutrophils into the frontoparietal cortex and increased status epilepticus–induced hippocampal neurodegeneration (Kim et al., 2009, 2010, 2011).
In addition to anticonvulsive effects during acute seizures, an impact of targeting the P2X7R has also been investigated during the development of epilepsy and on established epilepsy. In 2015, Soni et al. showed that P2X7R antagonism decreased the mean kindling score and restored cognitive deficits and motor coordination in the PTZ kindling model in rats, suggesting antiepileptogenic potential (Soni et al., 2015). These antiepileptogenic effects were confirmed in two more recent studies by Fischer et al. using the same model (Fischer et al., 2016) and by Amorim et al. using P2X7R-targeting siRNA post status epilepticus in the pilocarpine rat model (Amorim et al., 2017). Jamali-Raeuly et al., meanwhile, showed that treatment with a P2X7R antagonist and linagliptin, which blocks the enzyme dipeptidyl peptidase-4 and is used for the treatment of diabetes, reduced more efficiently seizure severity and neuronal cell death when given in combination in a KA rat model (Jamali-Raeufy et al., 2020). Similar, however, to status epilepticus, treatment with a P2X7R antagonist post pilocarpine-induced status epilepticus in mice resulted in the development of a more severe epileptic phenotype (Rozmer et al., 2017).
Less conflicting results have been obtained during epilepsy with two studies, published both in 2016, demonstrating antiepileptic effects of P2X7R antagonism. In the study carried out by Amhaoul et al., P2X7R antagonism during epilepsy reduced seizure severity, without, however, affecting the total number of seizures (Amhaoul et al., 2016). Jimenez-Pacheco et al. meanwhile showed that mice subjected to intra-amygdala KA and treated with a P2X7R antagonists starting 10 days post status epilepticus, when mice usually have experienced their first epileptic seizure, experienced fewer seizures during treatment and, remarkably, during a 1-week drug-washout period, suggesting disease-modifying potential (Jimenez-Pacheco et al., 2016).
As mentioned before, the only other P2XRs for which a functional role has been investigated in the setting of seizures and epilepsy include the P2X3R and P2X4R subtype. Here, Xia et al. showed, using a PTZ-induced kindling rat model, that treatment with a P2X3R antagonist reduced the mean kindling score and improved other pathological parameters such as memory deficits, motor activity, neuronal damage, and hippocampal inflammation (Xia et al., 2018). Using P2X4R knockout mice subjected to intraperitoneal KA-induced status epilepticus, Ulmann et al. showed that, despite experiencing no changes in seizure severity during status epilepticus, P2X4R deficiency partially protects from seizure-induced neurodegeneration (Ulmann et al., 2013).
P2Y Receptors
While less investigated, increasing evidence also suggests a role of the metabotropic P2YRs during epilepsy (Engel et al., 2016; Alves et al., 2018) with the P2Y1R and P2Y12R being the most studied (Alves et al., 2018; Table 73–1). In one of the first studies published in 2014, Eyo et al. demonstrated increased seizure severity in P2Y12R knockout mice subjected to intraperitoneal KA which was accompanied by a reduction in microglia process extension in the hippocampus (Eyo et al., 2014). Meanwhile, more recently, Milior et al. showed, using resected hippocampal tissue from patients with TLE, that while low doses of ADP induce microglial process extension, which was suppressed by treatment with P2Y12R antagonists, high doses of ADP cause microglia process retraction and membrane ruffling, blocked by the co-application of antagonists against the P2Y1R and P2Y13R (Milior et al., 2020).
One of the first investigations suggesting a role for the P2Y1R during seizures stems from a study carried out by Simoes et al. in 2018. Here, the authors showed that blocking of the P2Y1R reduced neuronal cell death following intraperitoneal KA-induced status epilepticus in rats. Seizure severity was, however, not affected by treatment (Simoes et al., 2018). A more complex role of P2Y1R was, however, suggested by a more recent study published by Alves et al. in 2019 in which mice deficient for P2Y1R experienced more severe seizures during intra-amygdala KA-induced status epilepticus with an associated increase in seizure-induced neurodegeneration (Alves et al., 2019a). These results were confirmed via pharmacological approaches where specific P2Y1R antagonists prior to intra-amygdala KA led to a more severe seizure phenotype. Conversely pretreatment with a P2Y1R agonist reduced seizure severity during status epilepticus (Alves et al., 2019a). However, and in contrast to their pretreatment regime, treatment of mice with P2Y1R agonists and antagonists post intra-amygdala KA injection, after the occurrence of the first seizure burst, showed the opposite result. In this case, P2Y1R antagonists reduced seizure severity and neurodegeneration, while treatment with a P2Y1R agonist increased seizure severity. Further confirming the anticonvulsant potential of P2Y1R antagonists once pathological changes are ongoing, data from the same study showed that treatment with a P2Y1R antagonist post-status epilepticus delayed the onset of epilepsy, and, when applied during epilepsy, suppressed epileptic seizures. However, in contrast to targeting the P2X7R, this effect did not persist following treatment withdrawal (Alves et al., 2019a). P2Y1R-based treatment is therefore highly dependent on the time of intervention relative to the onset of epilepsy. Whether this context-specific role during seizures is also true for other purinergic receptors remains to be established.
P2 Receptor–Mediated Molecular Mechanisms Contributing to Seizures and Epilepsy
P2 receptors have been implicated in multiple cellular processes pertinent to seizures and epilepsy, including synaptic reorganization, the regulation of the blood–brain barrier (BBB), cellular survival, circadian rhythms, neurogenesis, and inflammation (Burnstock and Ralevic, 2014; Pitkanen et al., 2015; Khan et al., 2018; Klein et al., 2018; Ali et al., 2020). While the exact mechanism(s) of how P2 receptors contribute to increased hyperexcitability in the brain remain to be established, inflammation has attracted particular attention. In this section we will briefly summarize evidence suggesting that P2 receptors impact on inflammatory processes during epilepsy. As mentioned before, it is, however, important to keep in mind that purinergic signaling is not restricted to driving inflammatory processes and that its impact on seizures is most likely the combination of several altered cellular pathways rather than a single one.
A role for inflammation on neuronal excitability is well established. Cytokines, such as the P2X7R downstream target Interleukin (IL)-1β, have been shown to be upregulated during seizures and epilepsy and to modulate GABAergic and NMDA receptor-dependent synaptic transmission (e.g., Wang et al., 2000a; Yang et al., 2005; Roseti et al., 2015; Stellwagen et al., 2005; Vezzani et al., 2019). Critically, drugs targeting inflammatory signaling pathways have shown promising effects during acute seizures and epilepsy (Vezzani et al., 2019).
One of the P2 receptors which has been most closely associated with inflammatory processes is the P2X7R. The P2X7R is highly expressed on microglia under both physiological conditions and during epilepsy where it contributes to microglial activation and proliferation (Monif et al., 2009; Kaczmarek-Hajek et al., 2018; Morgan et al., 2020). Evidence suggesting regulation of inflammatory processes during seizures and epilepsy by P2X7Rs stems from studies showing that blockade of the P2X7R leads to decreased hippocampal IL-1β levels post intra-amygdala KA-induced status epilepticus (Engel et al., 2012) and reduced microgliosis and astrogliosis during epilepsy (Jimenez-Pacheco et al., 2016). Although P2X7Rs are not detected on astrocytes during epilepsy, P2X7R antagonism protects against astrocyte death following pilocarpine-induced status epilepticus (Kim et al., 2009). Astrocytes can reduce seizure threshold via various mechanisms (e.g., dysregulation of the extracellular ionic balance, impaired neurotransmitter reuptake, release of pro-inflammatory cytokines and purines [e.g., ATP], removal of extracellular adenosine via ADK) (Bedner et al., 2015; Robel et al., 2015; Illes et al., 2019; Boison, 2008). Supporting this indirect role for P2X7Rs in astrocyte function, P2X7R-deficient mice subjected to intraperitoneal KA showed decreased astroglial autophagy through the regulation of FAK- and PHLPP1/2-mediated AKT-S473 phosphorylation (Lee and Kim, 2020).
The P2X4R subtype, primarily expressed on activated microglia where it drives microglia motility, process extension, activation, and recruitment to the site of damage (Raouf et al., 2007, Ohsawa et al., 2007, Montilla et al., 2020), represents another receptor subtype possibly contributing to inflammatory processes during seizures. Its contribution to seizure-induced inflammation is supported by Ullman et al., who found that P2X4Rs localize to activated microglia during intraperitoneal KA-induced status epilepticus and that mice deficient in P2X4R showed an impairment in several characteristics of microglia activation, including cell recruitment and upregulation of voltage-dependent potassium channels (Ulmann et al., 2013).
In contrast to the P2X7R and P2X4R, the P2Y12R seems to perform a protective function during seizures. In the brain, P2Y12R expression is mainly restricted to microglia (Haynes et al., 2006), where it has been involved in several processes, including microglial migration, the regulation of microglial-neuron interactions, and microglia injury detection (Gomez Morillas et al., 2021). While P2Y12R immunoreactivity has been widely accepted as a marker of resting microglia (Mildner et al., 2017), that is, microglia in nonpathological conditions, the primary role of P2Y12R activation is to induce reactivity of microglia and to stimulate their migratory movement via PI3k/PLC signaling to sites of inflammation (Ohsawa et al., 2007; Irino et al., 2008). In this line, Eyo et al. and, more recently, Milior et al. showed by using mouse models of epilepsy and resected brain tissue from TLE patients that both the deletion of P2Y12 (Eyo et al., 2014) and treatment with P2Y12R antagonists (Milior et al., 2020) reduced microglia process extension thereby preventing possible seizure-suppressive microglia-neuron interactions. Avignone et al. observed an increase in microglia motility following treatment with the P2Y12R agonist 2-Me-ADP following intraperitoneal KA-induced status epilepticus when compared to control (Avignone et al., 2015). This further supports a role for P2Y12Rs in regulating microglia during seizures.
In the case of the P2Y1R, findings are more complex with cell-specific expression changes and anti- and pro-convulsive functions that depend on the disease stage and model used. Whereas Alves et al. suggested that the proconvulsant effects of P2Y1R are mediated via P2Y1R acting on microglia, others have suggested these effects are mediated via astrocytes (Alvarez-Ferradas et al., 2015; Martorell et al., 2020). In support of a pro-convulsive role of P2Y1Rs via activation of microglia, Alves et al. showed that P2Y1Rs become rapidly up-regulated in microglia following intra-amygdala KA and that P2Y1R antagonism suppressed seizures. Thus, a likely explanation is that P2Y1Rs contribute to increased hyperexcitability via driving inflammatory processes (Alves et al., 2019a). In the same study, P2Y1R was mainly found to be expressed on neurons during physiological conditions. Because P2Y1R antagonism applied before intra-amygdala KA injection increased seizure severity, the authors concluded that, in contrast to P2Y1R on microglia, neuronal P2Y1R expression may have anticonvulsive effects. By contrast, and supporting the hypothesis that P2Y1R acts via astrocytes, P2Y1R has been suggested to contribute to seizures and epilepsy via the mediation of astrocytic-Ca2+ oscillations (Alvarez-Ferradas et al., 2015). More recently, and again in agreement with P2Y1R acting on astrocytes, Martorell et al. showed that P2Y1R inhibition rescues both the abnormal pattern of astroglial Ca2+-activity and plastic properties of CA3-CA1 synapses in the epileptic hippocampus (Martorell et al., 2020).
In a nutshell, while P2X4, P2X7, and P2Y1Rs seem to activate microglia, thereby lowering seizure threshold and contributing to the development of epilepsy, P2Y12Rs seem to exert an anticonvulsive function maintaining microglia in a nonactivated state. In addition, P2Y1Rs may also contribute to seizures via the activation of astrocytes (Fig. 73–2). It is, however, important to keep in mind that both cell types, astrocytes and microglia, maintain a tight relationship during seizures and epilepsy, thereby impacting on each other’s activation status and function (Sano et al., 2021). Microglial inflammatory action may precede astrocytic action, with microglia known to stimulate the transition of astrocytes to a neuroprotective phenotype (Shinozaki et al., 2017) and to tightly regulate their proliferation (Quintas et al., 2011). Moreover, while microglia were previously believed to have a mainly pro-convulsant function via the secretion of pro-inflammatory cytokines (e.g., IL-1β) (Vezzani et al., 1999), more recent research now has suggested also a protective function of microglia. In this line, the depletion of microglia leads to an amplification and synchronization of neuronal activity culminating in the generation of epileptic seizures. Interestingly, this effect was highly dependent on microglia’s ability to sense and catabolize extracellular ATP, which is released by neurons and astrocytes following neuronal activation (Badimon et al., 2020). This detrimental effect was further confirmed by another two recent studies where microglia were depleted in a similar fashion (Zhao et al., 2020; Liu et al., 2020).

Figure 73–2.
Positive feedforward loop of inflammation and P2 signaling underlying epileptogenesis. ATP is released from cells via exocytotic mechanisms, through leakage across damaged membranes and through purinergic channels such as the P2X7R and Pannexin-1 following (more...)
In summary, increasing evidence suggests P2 receptors as important contributors to inflammatory processes during epilepsy. Whether P2 receptor-driven changes in inflammation are the main signaling pathway of how these receptors impact on seizures and epilepsy and exactly which cell types are involved remain to be fully established. This will, however, most likely require the use of cell-specific knockout mice (e.g., Cre-LoxP system) or cell-type-specific targeting of different receptors (e.g., via viral vectors).
Purines as Diagnostics for Epilepsy
Together with the lack of effective pharmacological approaches in over 30% of patients and the absence of disease-modifying treatments, epilepsy diagnosis remains a clinical challenge, adding significantly to the disease burden (Moshe et al., 2015; Thijs et al., 2019). A correct diagnosis of epilepsy is critical to inform treatment. To date, epilepsy diagnosis relies, however, heavily on clinical examination and history and, patient monitoring via long-term video-EEG recording at hospitals remains the gold standard for the diagnosis of epilepsy. This is, however, time-consuming, costly, low-throughput, and requires a high level of specialist expertise (Engel et al., 2013; Moshe et al., 2015). Misdiagnosis rates are high and clinical signs can easily be confused with disorders, which present in a similar way, such as psychogenic nonepileptic attacks (Benbadis, 2009). Accordingly, there is significant interest in the discovery and validation of novel molecular biomarkers to identify patients with epilepsy. Ideally, these biomarkers should be noninvasive, be measured via a reproducible, easy-to-use, and economically feasible analysis platform and associated with cellular and molecular changes occurring during seizures and epilepsy (Engel et al., 2013).
Our laboratories have pioneered the development and use of biosensors for purines, and biosensors for adenosine and the downstream purines inosine and hypoxanthine have been developed to allow rapid and convenient measurement of purines in whole blood (Beamer et al., 2021; Martin et al., 2019; Fisher et al., 2019; Dale et al., 2019; Tian et al., 2007, 2017). As we have discussed previously, purines are produced in the brain during seizure activity. They also readily cross the BBB and can thus be detected in whole blood (Tian et al., 2017). This make purines a potentially valuable and convenient biomarker that could be elevated in the blood of epilepsy patients.
We have explored this concept in both mouse models of epilepsy and human patients. In a recent study, we found that induction of status epilepticus in mice rapidly elevated whole blood purine levels by nearly 4-fold (Beamer et al., 2021). Importantly, the extent of elevation correlated with the intensity of seizures and the degree of neurodegeneration in the hippocampus that followed induction of seizures, suggesting that the magnitude of the purine signal could indicate the severity of the epileptic insult. The sensitivity and specificity of these measurements had excellent performance in the mouse model with an area under the receiver operating characteristic (ROC) curve of 0.95.
Based on these results, we performed an initial study on 26 epileptic patients and 13 controls. Despite the patients not having experienced a seizure within 24 h of the measurement, we found that the baseline level of whole blood purines of epileptic patients was elevated by 2-fold relative to comparable healthy controls (Beamer et al., 2021). Overall, the diagnostic performance (area under the ROC curve of 0.79) in this study was highly promising and suggests that whole blood purine measurements could provide a rapid biomarker test suitable for triaging suspected epilepsy patients for more detailed clinical assessment (Beamer et al., 2021). Further trials are needed to explore this further and to study the extent to which, like in the mouse model, the level of whole-blood purines in human patients could indicate the severity of the condition.
Conclusion
We now have a substantial body of evidence demonstrating the therapeutic potential of targeting the purinergic system during epilepsy. This includes the well-known anticonvulsive and antiepileptogenic effects of targeting adenosine-dependent pathways, and more recently also ATP-dependent receptors (Fig. 73–3). While there are still important questions to be answered, such as possible cell-type-specific effects of certain receptors (e.g., P2Y1R) or the confirmation of results in animal models mimicking closer the human condition (e.g., TBI-induced epilepsy, genetic models), we have come a long way and it is now clear that the purinergic signaling system represents numerous promising targets for seizure control and treatment of epilepsy and, as shown more recently, as a diagnostic tool. Of particular promise are the disease-modifying effects observed via targeting of the ATP-gated P2X7R or ADK (Jimenez-Pacheco et al., 2016; Sandau et al., 2019). With drugs targeting the purinergic signaling system quickly moving from preclinical to clinical trials stage for CNS diseases (Timmers et al., 2018; Bhattacharya, 2018), the first clinical trials targeting this system in epilepsy can be expected in the near future.

Figure 73–3.
Targeting purinergic signaling to treat seizures and epilepsy. Schematic illustrating possible therapeutic intervention points targeting the purinergic system to suppress seizures and to treat epilepsy. (1) Purine release: Purines (e.g., ATP and adenosine) (more...)
Acknowledgments
This work was supported by funding from Science Foundation Ireland (17/CDA/4708) and Epilepsy Research UK (A1830).
Disclosure Statement
The authors declare no relevant conflicts.
References
- Adami, M., Bertorelli, R., Ferri, N., Foddi, M. C. & Ongini, E. 1995. Effects of repeated administration of selective adenosine A1 and A2A receptor agonists on pentylenetetrazole-induced convulsions in the rat. Eur J Pharmacol, 294, 383–9. [PubMed: 8750698]
- Ali, A. A. H., Avakian, G. A. & Gall, C. V. 2020. The Role of Purinergic Receptors in the Circadian System. Int J Mol Sci, 12;21(10):3423. [PMC free article: PMC7279285] [PubMed: 32408622]
- Alvarez-Ferradas, C., Morales, J. C., Wellmann, M., Nualart, F., Roncagliolo, M., Fuenzalida, M. & Bonansco, C. 2015. Enhanced astroglial Ca2+ signaling increases excitatory synaptic strength in the epileptic brain. Glia, 63, 1507–21. [PubMed: 25980474]
- Alves, M., Beamer, E. & Engel, T. 2018. The Metabotropic Purinergic P2Y Receptor Family as Novel Drug Target in Epilepsy. Front Pharmacol, 9, 193. [PMC free article: PMC5851315] [PubMed: 29563872]
- Alves, M., De Diego Garcia, L., Conte, G., Jimenez-Mateos, E. M., D’orsi, B., Sanz-Rodriguez, A., Prehn, J. H. M., Henshall, D. C. & Engel, T. 2019a. Context-Specific Switch from Anti- to Pro-epileptogenic Function of the P2Y1 Receptor in Experimental Epilepsy. J Neurosci, 39, 5377–5392. [PMC free article: PMC6607746] [PubMed: 31048325]
- Alves, M., Gomez-Villafuertes, R., Delanty, N., Farrell, M. A., O’brien, D. F., Miras-Portugal, M. T., Hernandez, M. D., Henshall, D. C. & Engel, T. 2017. Expression and function of the metabotropic purinergic P2Y receptor family in experimental seizure models and patients with drug-refractory epilepsy. Epilepsia, 58, 1603–1614. [PubMed: 28733972]
- Alves, M., Smith, J. & Engel, T. 2019b. Differential Expression of the Metabotropic P2Y Receptor Family in the Cortex Following Status Epilepticus and Neuroprotection via P2Y1 Antagonism in Mice. Front Pharmacol, 10, 1558. [PMC free article: PMC6976538] [PubMed: 32009961]
- Amhaoul, H., Ali, I., Mola, M., Van Eetveldt, A., Szewczyk, K., Missault, S., Bielen, K., Kumar-Singh, S., Rech, J., Lord, B., Ceusters, M., Bhattacharya, A. & Dedeurwaerdere, S. 2016. P2X7 receptor antagonism reduces the severity of spontaneous seizures in a chronic model of temporal lobe epilepsy. Neuropharmacology, 105, 175–185. [PubMed: 26775823]
- Amorim, R. P., Araujo, M. G. L., Valero, J., Lopes-Cendes, I., Pascoal, V. D. B., Malva, J. O. & Da Silva Fernandes, M. J. 2017. Silencing of P2X7R by RNA interference in the hippocampus can attenuate morphological and behavioral impact of pilocarpine-induced epilepsy. Purinergic Signal, 13, 467–478. [PMC free article: PMC5714836] [PubMed: 28707031]
- Avignone, E., Lepleux, M., Angibaud, J. & Nagerl, U. V. 2015. Altered morphological dynamics of activated microglia after induction of status epilepticus. J Neuroinflammation, 12, 202. [PMC free article: PMC4634193] [PubMed: 26538404]
- Avignone, E., Ulmann, L., Levavasseur, F., Rassendren, F. & Audinat, E. 2008. Status epilepticus induces a particular microglial activation state characterized by enhanced purinergic signaling. J Neurosci, 28, 9133–44. [PMC free article: PMC6670931] [PubMed: 18784294]
- B.B. Fredholm, K. B., J. Holmén, A. Nehlig, E.E 1999. Actions of caffeine in the brain with special reference to factors that contribute to its widespread use. Pharmacol Rev, 51, 83–133. [PubMed: 10049999]
- Badimon, A., Strasburger, H. J., Ayata, P., Chen, X., Nair, A., Ikegami, A., Hwang, P., Chan, A. T., Graves, S. M., Uweru, J. O., Ledderose, C., Kutlu, M. G., Wheeler, M. A., Kahan, A., Ishikawa, M., Wang, Y. C., Loh, Y. E., Jiang, J. X., Surmeier, D. J., Robson, S. C., Junger, W. G., Sebra, R., Calipari, E. S., Kenny, P. J., Eyo, U. B., Colonna, M., Quintana, F. J., Wake, H., Gradinaru, V. & Schaefer, A. 2020. Negative feedback control of neuronal activity by microglia. Nature, 586, 417–423. [PMC free article: PMC7577179] [PubMed: 32999463]
- Baldwin, S. A., Beal, P. R., Yao, S. Y., King, A. E., Cass, C. E. & Young, J. D. 2004. The equilibrative nucleoside transporter family, SLC29. Pflugers Arch, 447, 735–43. [PubMed: 12838422]
- Barros-Barbosa, A. R., Ferreirinha, F., Oliveira, A., Mendes, M., Lobo, M. G., Santos, A., Rangel, R., Pelletier, J., Sevigny, J., Cordeiro, J. M. & Correia-De-Sa, P. 2016a. Adenosine A2A receptor and ecto-5’-nucleotidase/CD73 are upregulated in hippocampal astrocytes of human patients with mesial temporal lobe epilepsy (MTLE). Purinergic Signal, 12, 719–734. [PMC free article: PMC5124012] [PubMed: 27650530]
- Barros-Barbosa, A. R., Fonseca, A. L., Guerra-Gomes, S., Ferreirinha, F., Santos, A., Rangel, R., Lobo, M. G., Correia-De-Sa, P. & Cordeiro, J. M. 2016b. Up-regulation of P2X7 receptor-mediated inhibition of GABA uptake by nerve terminals of the human epileptic neocortex. Epilepsia, 57, 99–110. [PubMed: 26714441]
- Beamer, E., Fischer, W. & Engel, T. 2017. The ATP-Gated P2X7 Receptor As a Target for the Treatment of Drug-Resistant Epilepsy. Front Neurosci, 11, 21. [PMC free article: PMC5288361] [PubMed: 28210205]
- Beamer, E., Lacey, A., Alves, M., Conte, G., Tian, F., De Diego-Garcia, L., Khalil, M., Rosenow, F., Delanty, N., Dale, N., El-Naggar, H., Henshall, D. C. & Engel, T. 2021. Elevated blood purine levels as a biomarker of seizures and epilepsy. Epilepsia. 62(3):817–828. [PubMed: 33599287]
- Bedner, P., Dupper, A., Huttmann, K., Muller, J., Herde, M. K., Dublin, P., Deshpande, T., Schramm, J., Haussler, U., Haas, C. A., Henneberger, C., Theis, M. & Steinhauser, C. 2015. Astrocyte uncoupling as a cause of human temporal lobe epilepsy. Brain, 138, 1208–22. [PMC free article: PMC5963418] [PubMed: 25765328]
- Benarroch, E. E. 2008. Adenosine and its receptors. Multiple modulatory functions and potential therapeutic targets for neurologic disease. Neurology, 70, 231–236. [PubMed: 18195269]
- Benbadis, S. 2009. The differential diagnosis of epilepsy: a critical review. Epilepsy Behav, 15, 15–21. [PubMed: 19236946]
- Benford, H., Bolborea, M., Pollatzek, E., Lossow, K., Hermans-Borgmeyer, I., Liu, B., Meyerhof, W., Kasparov, S. & Dale, N. 2017. A sweet taste receptor-dependent mechanism of glucosensing in hypothalamic tanycytes. Glia, 65, 773–789. [PMC free article: PMC5363357] [PubMed: 28205335]
- Bhattacharya, A. 2018. Recent Advances in CNS P2X7 Physiology and Pharmacology: Focus on Neuropsychiatric Disorders. Front Pharmacol, 9, 30. [PMC free article: PMC5799703] [PubMed: 29449810]
- Billaud, M., Sandilos, J. K. & Isakson, B. E. 2012. Pannexin 1 in the regulation of vascular tone. Trends Cardiovasc Med, 22, 68–72. [PMC free article: PMC3455115] [PubMed: 22841835]
- Boison, D. 2008. The adenosine kinase hypothesis of epileptogenesis. Prog Neurobiol, 84, 249–62. [PMC free article: PMC2278041] [PubMed: 18249058]
- Boison, D. 2013. Adenosine kinase: exploitation for therapeutic gain. Pharmacol Rev, 65, 906–43. [PMC free article: PMC3698936] [PubMed: 23592612]
- Boison, D. & Rho, J. M. 2019. Epigenetics and epilepsy prevention: The therapeutic potential of adenosine and metabolic therapies. Neuropharmacology, 107741. [PMC free article: PMC7220211] [PubMed: 31419398]
- Boison, D. & Yegutkin, G. G. 2019. Adenosine Metabolism: Emerging Concepts for Cancer Therapy. Cancer Cell, 36, 582–596. [PMC free article: PMC7224341] [PubMed: 31821783]
- Borea, P. A., Gessi, S., Merighi, S., Vincenzi, F. & Varani, K. 2018. Pharmacology of Adenosine Receptors: The State of the Art. Physiol Rev, 98, 1591–1625. [PubMed: 29848236]
- Brake, A. J., Wagenbach, M. J. & Julius, D. 1994. New structural motif for ligand-gated ion channels defined by an ionotropic ATP receptor. Nature, 371, 519–23. [PubMed: 7523952]
- Burnstock, G. 1972. Purinergic nerves. Pharmacol Rev, 24, 509–81. [PubMed: 4404211]
- Burnstock, G. 1976. Do some nerve cells release more than one transmitter? Neuroscience, 1, 239–48. [PubMed: 11370511]
- Burnstock, G. 2013. Introduction to purinergic signaling in the brain. Adv Exp Med Biol, 986, 1–12. [PubMed: 22879061]
- Burnstock, G. & Kennedy, C. 1985. Is there a basis for distinguishing two types of P2-purinoceptor? Gen Pharmacol, 16, 433–40. [PubMed: 2996968]
- Burnstock, G. & Ralevic, V. 2014. Purinergic signaling and blood vessels in health and disease. Pharmacol Rev, 66, 102–92. [PubMed: 24335194]
- Canas, P. M., Porciuncula, L. O., Simoes, A. P., Augusto, E., Silva, H. B., Machado, N. J., Goncalves, N., Alfaro, T. M., Goncalves, F. Q., Araujo, I. M., Real, J. I., Coelho, J. E., Andrade, G. M., Almeida, R. D., Chen, J. F., Kofalvi, A., Agostinho, P. & Cunha, R. A. 2018. Neuronal Adenosine A2A Receptors Are Critical Mediators of Neurodegeneration Triggered by Convulsions. eNeuro, 5. [PMC free article: PMC6325550] [PubMed: 30627646]
- Carrer, A., Leparulo, A., Crispino, G., Ciubotaru, C. D., Marin, O., Zonta, F. & Bortolozzi, M. 2018. Cx32 hemichannel opening by cytosolic Ca2+ is inhibited by the R220X mutation that causes Charcot-Marie-Tooth disease. Hum Mol Genet, 27, 80–94. [PubMed: 29077882]
- Chadwick, B. P. & Frischauf, A. M. 1998. The CD39-like gene family: identification of three new human members (CD39L2, CD39L3, and CD39L4), their murine homologues, and a member of the gene family from Drosophila melanogaster. Genomics, 50, 357–67. [PubMed: 9676430]
- Chwalczuk, K., Rubaj, A., Swiader, M. & Czuczwar, S. J. 2008. [Influence of the antagonist of adenosine A1 receptors, 8-cyclopentyl-1 ,3-dipropylxanthine, upon the anticonvulsant activity of antiepileptic drugs in mice]. Przegl Lek, 65, 759–63. [PubMed: 19205356]
- Ciancetta, A. & Jacobson, K. A. 2018. Breakthrough in GPCR Crystallography and Its Impact on Computer-Aided Drug Design. Methods Mol Biol, 1705, 45–72. [PMC free article: PMC5859332] [PubMed: 29188558]
- Dale, N. 1998. Delayed production of adenosine underlies temporal modulation of swimming in frog embryo. J Physiol, 511 (Pt 1), 265–72. [PMC free article: PMC2231108] [PubMed: 9679180]
- Dale, N. 2021. Biological insights from the direct measurement of purine release. Biochem Pharmacol, 187, 114416. [PubMed: 33444569]
- Dale, N. & Gilday, D. 1996. Regulation of rhythmic movements by purinergic neurotransmitters in frog embryos. Nature, 383, 259–63. [PubMed: 8805702]
- Dale, N., Tian, F., Sagoo, R., Phillips, N., Imray, C. & Roffe, C. 2019. Point-of-care measurements reveal release of purines into venous blood of stroke patients. Purinergic Signal, 15, 237–246. [PMC free article: PMC6635545] [PubMed: 30859371]
- Dalmau, I., Vela, J. M., Gonzalez, B. & Castellano, B. 1998. Expression of purine metabolism-related enzymes by microglial cells in the developing rat brain. J Comp Neurol, 398, 333–46. [PubMed: 9714147]
- Davalos, D., Grutzendler, J., Yang, G., Kim, J. V., Zuo, Y., Jung, S., Littman, D. R., Dustin, M. L. & Gan, W. B. 2005. ATP mediates rapid microglial response to local brain injury in vivo. Nat Neurosci, 8, 752–8. [PubMed: 15895084]
- Diez-Zaera, M., Diaz-Hernandez, J. I., Hernandez-Alvarez, E., Zimmermann, H., Diaz-Hernandez, M. & Miras-Portugal, M. T. 2011. Tissue-nonspecific alkaline phosphatase promotes axonal growth of hippocampal neurons. Mol Biol Cell, 22, 1014–24. [PMC free article: PMC3069005] [PubMed: 21289095]
- Dogan, E., Aygun, H., Arslan, G., Rzayev, E., Avci, B., Ayyildiz, M. & Agar, E. 2020. The Role of NMDA Receptors in the Effect of Purinergic P2X7 Receptor on Spontaneous Seizure Activity in WAG/Rij Rats With Genetic Absence Epilepsy. Front Neurosci, 14, 414. [PMC free article: PMC7218146] [PubMed: 32435183]
- Dona, F., Conceicao, I. M., Ulrich, H., Ribeiro, E. B., Freitas, T. A., Nencioni, A. L. & Da Silva Fernandes, M. J. 2016. Variations of ATP and its metabolites in the hippocampus of rats subjected to pilocarpine-induced temporal lobe epilepsy. Purinergic Signal, 12, 295–302. [PMC free article: PMC4854830] [PubMed: 26939579]
- Dona, F., Ulrich, H., Persike, D. S., Conceicao, I. M., Blini, J. P., Cavalheiro, E. A. & Fernandes, M. J. 2009. Alteration of purinergic P2X4 and P2X7 receptor expression in rats with temporal-lobe epilepsy induced by pilocarpine. Epilepsy Res, 83, 157–67. [PubMed: 19084381]
- Dospinescu, V. M., Nijjar, S., Spanos, F., Cook, J., De Wolf, E., Biscotti, M. A., Gerdol, M. & Dale, N. 2019. Structural determinants of CO2-sensitivity in the beta connexin family suggested by evolutionary analysis. Commun Biol, 2, 331. [PMC free article: PMC6726660] [PubMed: 31508505]
- Dossi, E., Blauwblomme, T., Moulard, J., Chever, O., Vasile, F., Guinard, E., Le Bert, M., Couillin, I., Pallud, J., Capelle, L., Huberfeld, G. & Rouach, N. 2018. Pannexin-1 channels contribute to seizure generation in human epileptic brain tissue and in a mouse model of epilepsy. Sci Transl Med, 10. (443):eaar3796. [PubMed: 29848662]
- Dulla, C. G., Frenguelli, B. G., Staley, K. J. & Masino, S. A. 2009. Intracellular acidification causes adenosine release during states of hyperexcitability in the hippocampus. J Neurophysiol, 102, 1984–93. [PMC free article: PMC2746788] [PubMed: 19625534]
- Dunwiddie, T. V., Diao, L., Kim, H. O., Jiang, J. L. & Jacobson, K. A. 1997. Activation of hippocampal adenosine A3 receptors produces a desensitization of A1 receptor-mediated responses in rat hippocampus. J Neurosci, 17, 607–14. [PMC free article: PMC5470729] [PubMed: 8987783]
- During, M. J. & Spencer, D. D. 1992. Adenosine: a potential mediator of seizure arrest and postictal refractoriness. Ann Neurol, 32, 618–24. [PubMed: 1449242]
- Edwards, F. A., Gibb, A. J. & Colquhoun, D. 1992. ATP receptor-mediated synaptic currents in the central nervous system. Nature, 359, 144–7. [PubMed: 1381811]
- El Yacoubi, M., Ledent, C., Parmentier, M., Bertorelli, R., Ongini, E., Costentin, J. & Vaugeois, J. M. 2001. Adenosine A2A receptor antagonists are potential antidepressants: evidence based on pharmacology and A2A receptor knockout mice. Br J Pharmacol, 134, 68–77. [PMC free article: PMC1572930] [PubMed: 11522598]
- El Yacoubi, M., Ledent, C., Parmentier, M., Costentin, J. & Vaugeois, J. M. 2008. Evidence for the involvement of the adenosine A(2A) receptor in the lowered susceptibility to pentylenetetrazol-induced seizures produced in mice by long-term treatment with caffeine. Neuropharmacology, 55, 35–40. [PubMed: 18486156]
- El Yacoubi, M., Ledent, C., Parmentier, M., Costentin, J. & Vaugeois, J. M. 2009. Adenosine A2A receptor deficient mice are partially resistant to limbic seizures. Naunyn Schmiedebergs Arch Pharmacol, 380, 223–32. [PubMed: 19488739]
- Engel, J., Jr., Pitkanen, A., Loeb, J. A., Dudek, F. E., Bertram, E. H., 3rd, Cole, A. J., Moshe, S. L., Wiebe, S., Jensen, F. E., Mody, I., Nehlig, A. & Vezzani, A. 2013. Epilepsy biomarkers. Epilepsia, 54 Suppl 4, 61–9. [PMC free article: PMC4131763] [PubMed: 23909854]
- Engel, T., Alves, M., Sheedy, C. & Henshall, D. C. 2016. ATPergic signaling during seizures and epilepsy. Neuropharmacology, 104, 140–53. [PubMed: 26549853]
- Engel, T., Gomez-Villafuertes, R., Tanaka, K., Mesuret, G., Sanz-Rodriguez, A., Garcia-Huerta, P., Miras-Portugal, M. T., Henshall, D. C. & Diaz-Hernandez, M. 2012. Seizure suppression and neuroprotection by targeting the purinergic P2X7 receptor during status epilepticus in mice. FASEB J, 26, 1616–28. [PubMed: 22198387]
- Eyo, U. B., Peng, J., Swiatkowski, P., Mukherjee, A., Bispo, A. & Wu, L. J. 2014. Neuronal hyperactivity recruits microglial processes via neuronal NMDA receptors and microglial P2Y12 receptors after status epilepticus. J Neurosci, 34, 10528–40. [PMC free article: PMC4200107] [PubMed: 25100587]
- Fedele, D. E., Li, T., Lan, J. Q., Fredholm, B. B. & Boison, D. 2006. Adenosine A1 receptors are crucial in keeping an epileptic focus localized. Exp Neurol, 200, 184–90. [PubMed: 16750195]
- Fields, R. D. 2011. Signaling by neuronal swelling. Sci Signal, 4, tr1. [PMC free article: PMC3201844] [PubMed: 21224445]
- Fields, R. D. & Ni, Y. 2010. Nonsynaptic communication through ATP release from volume-activated anion channels in axons. Sci Signal, 3, ra73. [PMC free article: PMC5023281] [PubMed: 20923934]
- Fischer, W., Franke, H., Krugel, U., Muller, H., Dinkel, K., Lord, B., Letavic, M. A., Henshall, D. C. & Engel, T. 2016. Critical Evaluation of P2X7 Receptor Antagonists in Selected Seizure Models. PLoS One, 11, e0156468. [PMC free article: PMC4900628] [PubMed: 27281030]
- Fisher, O., Benson, R. A., Tian, F., Dale, N. E. & Imray, C. H. 2019. Purine nucleoside use as surrogate markers of cerebral ischaemia during local and general anaesthetic carotid endarterectomy. SAGE Open Med, 7, 2050312119865120. [PMC free article: PMC6643180] [PubMed: 31367381]
- Frayling, C., Britton, R. & Dale, N. 2011. ATP-mediated glucosensing by hypothalamic tanycytes. J Physiol, 589, 2275–86. [PMC free article: PMC3098703] [PubMed: 21486800]
- Fredholm, B. B., Ap, I. J., Jacobson, K. A., Linden, J. & Muller, C. E. 2011. International Union of Basic and Clinical Pharmacology. LXXXI. Nomenclature and classification of adenosine receptors--an update. Pharmacol Rev, 63, 1–34. [PMC free article: PMC3061413] [PubMed: 21303899]
- Fukuda, M., Suzuki, Y., Hino, H., Kuzume, K., Morimoto, T. & Ishii, E. 2010. Adenosine A1 receptor blockage mediates theophylline-associated seizures. Epilepsia, 51, 483–7. [PubMed: 19845735]
- Funakoshi, I., Kato, H., Horie, K., Yano, T., Hori, Y., Kobayashi, H., Inoue, T., Suzuki, H., Fukui, S., Tsukahara, M. & et al. 1992. Molecular cloning of cDNAs for human fibroblast nucleotide pyrophosphatase. Arch Biochem Biophys, 295, 180–7. [PubMed: 1315502]
- Gomez Morillas, A., Besson, V. C. & Lerouet, D. 2021. Microglia and Neuroinflammation: What Place for P2RY12? Int J Mol Sci, 22. [PMC free article: PMC7915979] [PubMed: 33561958]
- Gouder, N., Fritschy, J. M. & Boison, D. 2003. Seizure suppression by adenosine A1 receptor activation in a mouse model of pharmacoresistant epilepsy. Epilepsia, 44, 877–85. [PubMed: 12823569]
- Guzman, S. J. & Gerevich, Z. 2016. P2Y Receptors in Synaptic Transmission and Plasticity: Therapeutic Potential in Cognitive Dysfunction. Neural Plast, 2016, 1207393. [PMC free article: PMC4812485] [PubMed: 27069691]
- Halassa, M. M., Florian, C., Fellin, T., Munoz, J. R., Lee, S. Y., Abel, T., Haydon, P. G. & Frank, M. G. 2009. Astrocytic modulation of sleep homeostasis and cognitive consequences of sleep loss. Neuron, 61, 213–9. [PMC free article: PMC2673052] [PubMed: 19186164]
- Hattori, M. & Gouaux, E. 2012. Molecular mechanism of ATP binding and ion channel activation in P2X receptors. Nature, 485, 207–12. [PMC free article: PMC3391165] [PubMed: 22535247]
- Haydon, P. G. 2001. GLIA: listening and talking to the synapse. Nat Rev Neurosci, 2, 185–93. [PubMed: 11256079]
- Haynes, S. E., Hollopeter, G., Yang, G., Kurpius, D., Dailey, M. E., Gan, W. B. & Julius, D. 2006. The P2Y12 receptor regulates microglial activation by extracellular nucleotides. Nat Neurosci, 9, 1512–9. [PubMed: 17115040]
- Heinrich, A., Ando, R. D., Turi, G., Rozsa, B. & Sperlagh, B. 2012. K+ depolarization evokes ATP, adenosine and glutamate release from glia in rat hippocampus: a microelectrode biosensor study. Br J Pharmacol, 167, 1003–20. [PMC free article: PMC3492983] [PubMed: 22394324]
- Henthorn, P. S., Raducha, M., Kadesch, T., Weiss, M. J. & Harris, H. 1988. Sequence and characterization of the human intestinal alkaline phosphatase gene. J Biol Chem, 263, 12011–9. [PubMed: 2841341]
- Huang, C., Chi, X. S., Li, R., Hu, X., Xu, H. X., Li, J. M. & Zhou, D. 2017. Inhibition of P2X7 Receptor Ameliorates Nuclear Factor-Kappa B Mediated Neuroinflammation Induced by Status Epilepticus in Rat Hippocampus. J Mol Neurosci, 63, 173–184. [PubMed: 28856625]
- Huang, Y. J., Maruyama, Y., Dvoryanchikov, G., Pereira, E., Chaudhari, N. & Roper, S. D. 2007. The role of pannexin 1 hemichannels in ATP release and cell-cell communication in mouse taste buds. Proc Natl Acad Sci U S A, 104, 6436–41. [PMC free article: PMC1851090] [PubMed: 17389364]
- Huckstepp, R. T., Eason, R., Sachdev, A. & Dale, N. 2010a. CO2-dependent opening of connexin 26 and related beta connexins. J Physiol, 588, 3921–31. [PMC free article: PMC3000582] [PubMed: 20736419]
- Huckstepp, R. T., Id Bihi, R., Eason, R., Spyer, K. M., Dicke, N., Willecke, K., Marina, N., Gourine, A. V. & Dale, N. 2010b. Connexin hemichannel-mediated CO2-dependent release of ATP in the medulla oblongata contributes to central respiratory chemosensitivity. J Physiol, 588, 3901–20. [PMC free article: PMC3000581] [PubMed: 20736421]
- Hugel, S. & Schlichter, R. 2000. Presynaptic P2X receptors facilitate inhibitory GABAergic transmission between cultured rat spinal cord dorsal horn neurons. J Neurosci, 20, 2121–30. [PMC free article: PMC6772506] [PubMed: 10704486]
- Ilie, A., Raimondo, J. V. & Akerman, C. J. 2012. Adenosine release during seizures attenuates GABAA receptor-mediated depolarization. J Neurosci, 32, 5321–32. [PMC free article: PMC6622092] [PubMed: 22496577]
- Illes, P., Burnstock, G. & Tang, Y. 2019. Astroglia-Derived ATP Modulates CNS Neuronal Circuits. Trends Neurosci, 42, 885–898. [PubMed: 31704181]
- Illes, P., Khan, T. M. & Rubini, P. 2017. Neuronal P2X7 Receptors Revisited: Do They Really Exist? J Neurosci, 37, 7049–7062. [PMC free article: PMC6705732] [PubMed: 28747388]
- Irino, Y., Nakamura, Y., Inoue, K., Kohsaka, S. & Ohsawa, K. 2008. Akt activation is involved in P2Y12 receptor-mediated chemotaxis of microglia. J Neurosci Res, 86, 1511–9. [PubMed: 18183622]
- Jamali-Raeufy, N., Barati, H., Baluchnejadmojarad, T., Roghani, M. & Goudarzi, M. 2020. Combination therapy with dipeptidyl peptidase-4 and P2X7 purinoceptor inhibitors gives rise to antiepileptic effects in rats. J Chem Neuroanat, 110, 101855. [PubMed: 33031915]
- James, S. & Richardson, P. J. 1993. Production of adenosine from extracellular ATP at the striatal cholinergic synapse. J Neurochem, 60, 219–27. [PubMed: 8417143]
- Jimenez-Mateos, E. M., Arribas-Blazquez, M., Sanz-Rodriguez, A., Concannon, C., Olivos-Ore, L. A., Reschke, C. R., Mooney, C. M., Mooney, C., Lugara, E., Morgan, J., Langa, E., Jimenez-Pacheco, A., Silva, L. F., Mesuret, G., Boison, D., Miras-Portugal, M. T., Letavic, M., Artalejo, A. R., Bhattacharya, A., Diaz-Hernandez, M., Henshall, D. C. & Engel, T. 2015. microRNA targeting of the P2X7 purinoceptor opposes a contralateral epileptogenic focus in the hippocampus. Sci Rep, 5, 17486. [PMC free article: PMC4668358] [PubMed: 26631939]
- Jimenez-Pacheco, A., Diaz-Hernandez, M., Arribas-Blazquez, M., Sanz-Rodriguez, A., Olivos-Ore, L. A., Artalejo, A. R., Alves, M., Letavic, M., Miras-Portugal, M. T., Conroy, R. M., Delanty, N., Farrell, M. A., O’brien, D. F., Bhattacharya, A., Engel, T. & Henshall, D. C. 2016. Transient P2X7 Receptor Antagonism Produces Lasting Reductions in Spontaneous Seizures and Gliosis in Experimental Temporal Lobe Epilepsy. J Neurosci, 36, 5920–32. [PMC free article: PMC6601816] [PubMed: 27251615]
- Jimenez-Pacheco, A., Mesuret, G., Sanz-Rodriguez, A., Tanaka, K., Mooney, C., Conroy, R., Miras-Portugal, M. T., Diaz-Hernandez, M., Henshall, D. C. & Engel, T. 2013. Increased neocortical expression of the P2X7 receptor after status epilepticus and anticonvulsant effect of P2X7 receptor antagonist A-438079. Epilepsia, 54, 1551–61. [PubMed: 23808395]
- Jo, Y. H. & Schlichter, R. 1999. Synaptic corelease of ATP and GABA in cultured spinal neurons. Nat Neurosci, 2, 241–5. [PubMed: 10195216]
- Kaczmarek-Hajek, K., Zhang, J., Kopp, R., Grosche, A., Rissiek, B., Saul, A., Bruzzone, S., Engel, T., Jooss, T., Krautloher, A., Schuster, S., Magnus, T., Stadelmann, C., Sirko, S., Koch-Nolte, F., Eulenburg, V. & Nicke, A. 2018. Re-evaluation of neuronal P2X7 expression using novel mouse models and a P2X7-specific nanobody. Elife, 7: e36217. [PMC free article: PMC6140716] [PubMed: 30074479]
- Kaczmarek, E., Koziak, K., Sevigny, J., Siegel, J. B., Anrather, J., Beaudoin, A. R., Bach, F. H. & Robson, S. C. 1996. Identification and characterization of CD39/vascular ATP diphosphohydrolase. J Biol Chem, 271, 33116–22. [PubMed: 8955160]
- Karasawa, A. & Kawate, T. 2016. Structural basis for subtype-specific inhibition of the P2X7 receptor. Elife, 9:5:e22153. [PMC free article: PMC5176352] [PubMed: 27935479]
- Kawate, T., Michel, J. C., Birdsong, W. T. & Gouaux, E. 2009. Crystal structure of the ATP-gated P2X(4) ion channel in the closed state. Nature, 460, 592–8. [PMC free article: PMC2720809] [PubMed: 19641588]
- Kennedy, C. 2021. The P2Y/P2X divide: How it began. Biochem Pharmacol, 187, 114408. [PubMed: 33444568]
- Khakh, B. S., Burnstock, G., Kennedy, C., King, B. F., North, R. A., Seguela, P., Voigt, M. & Humphrey, P. P. 2001. International union of pharmacology. XXIV. Current status of the nomenclature and properties of P2X receptors and their subunits. Pharmacol Rev, 53, 107–18. [PubMed: 11171941]
- Khan, S., Nobili, L., Khatami, R., Loddenkemper, T., Cajochen, C., Dijk, D. J. & Eriksson, S. H. 2018. Circadian rhythm and epilepsy. Lancet Neurol, 17, 1098–1108. [PubMed: 30366868]
- Kim, J. E. & Kang, T. C. 2011. The P2X7 receptor-pannexin-1 complex decreases muscarinic acetylcholine receptor-mediated seizure susceptibility in mice. J Clin Invest, 121, 2037–47. [PMC free article: PMC3083785] [PubMed: 21505260]
- Kim, J. E., Kwak, S. E., Jo, S. M. & Kang, T. C. 2009. Blockade of P2X receptor prevents astroglial death in the dentate gyrus following pilocarpine-induced status epilepticus. Neurol Res, 31, 982–8. [PubMed: 19138473]
- Kim, J. E., Ryu, H. J. & Kang, T. C. 2011. P2X7 receptor activation ameliorates CA3 neuronal damage via a tumor necrosis factor-alpha-mediated pathway in the rat hippocampus following status epilepticus. J Neuroinflammation, 8, 62. [PMC free article: PMC3123566] [PubMed: 21631954]
- Kim, J. E., Ryu, H. J., Yeo, S. I. & Kang, T. C. 2010. P2X7 receptor regulates leukocyte infiltrations in rat frontoparietal cortex following status epilepticus. J Neuroinflammation, 7, 65. [PMC free article: PMC2964655] [PubMed: 20939924]
- Klein, P., Dingledine, R., Aronica, E., Bernard, C., Blumcke, I., Boison, D., Brodie, M. J., Brooks-Kayal, A. R., Engel, J., Jr., Forcelli, P. A., Hirsch, L. J., Kaminski, R. M., Klitgaard, H., Kobow, K., Lowenstein, D. H., Pearl, P. L., Pitkanen, A., Puhakka, N., Rogawski, M. A., Schmidt, D., Sillanpaa, M., Sloviter, R. S., Steinhauser, C., Vezzani, A., Walker, M. C. & Loscher, W. 2018. Commonalities in epileptogenic processes from different acute brain insults: Do they translate? Epilepsia, 59, 37–66. [PMC free article: PMC5993212] [PubMed: 29247482]
- Klyuch, B. P., Dale, N. & Wall, M. J. 2012. Deletion of ecto-5’-nucleotidase (CD73) reveals direct action potential-dependent adenosine release. J Neurosci, 32, 3842–7. [PMC free article: PMC6703466] [PubMed: 22423104]
- Kobow, K., Kaspi, A., Harikrishnan, K. N., Kiese, K., Ziemann, M., Khurana, I., Fritzsche, I., Hauke, J., Hahnen, E., Coras, R., Muhlebner, A., El-Osta, A. & Blumcke, I. 2013. Deep sequencing reveals increased DNA methylation in chronic rat epilepsy. Acta Neuropathol, 126, 741–56. [PMC free article: PMC3825532] [PubMed: 24005891]
- Kochanek, P. M., Vagni, V. A., Janesko, K. L., Washington, C. B., Crumrine, P. K., Garman, R. H., Jenkins, L. W., Clark, R. S., Homanics, G. E., Dixon, C. E., Schnermann, J. & Jackson, E. K. 2006. Adenosine A1 receptor knockout mice develop lethal status epilepticus after experimental traumatic brain injury. J Cereb Blood Flow Metab, 26, 565–75. [PubMed: 16121125]
- Langer, D., Ikehara, Y., Takebayashi, H., Hawkes, R. & Zimmermann, H. 2007. The ectonucleotidases alkaline phosphatase and nucleoside triphosphate diphosphohydrolase 2 are associated with subsets of progenitor cell populations in the mouse embryonic, postnatal and adult neurogenic zones. Neuroscience, 150, 863–79. [PubMed: 18031938]
- Lazarowski, E. 2006. Regulated release of nucleotides and UDP sugars from astrocytoma cells. Novartis Found Symp, 276, 73–84; discussion 84-90, 107–12, 275–81. [PubMed: 16805424]
- Lazutkaite, G., Solda, A., Lossow, K., Meyerhof, W. & Dale, N. 2017. Amino acid sensing in hypothalamic tanycytes via umami taste receptors. Mol Metab, 6, 1480–1492. [PMC free article: PMC5681271] [PubMed: 29107294]
- Lee, D. S. & Kim, J. E. 2020. P2 x 7 Receptor Inhibits Astroglial Autophagy via Regulating FAK- and PHLPP1/2-Mediated AKT-S473 Phosphorylation Following Kainic Acid-Induced Seizures. Int J Mol Sci, 21(18):6476. [PMC free article: PMC7555659] [PubMed: 32899862]
- Li, M., Kang, R., Shi, J., Liu, G. & Zhang, J. 2013. Anticonvulsant activity of B2, an adenosine analog, on chemical convulsant-induced seizures. PLoS One, 8, e67060. [PMC free article: PMC3692431] [PubMed: 23825618]
- Li, W. & Zhang, J. J. 2011. [Sedative, hypnotic and anticonvulsive effects of an adenosine analogue WS090501]. Yao Xue Xue Bao, 46, 742–6. [PubMed: 21882539]
- Liu, M., Jiang, L., Wen, M., Ke, Y., Tong, X., Huang, W. & Chen, R. 2020. Microglia depletion exacerbates acute seizures and hippocampal neuronal degeneration in mouse models of epilepsy. Am J Physiol Cell Physiol, 319, C605-C610. [PubMed: 32783655]
- Lopatar, J., Dale, N. & Frenguelli, B. G. 2011. Minor contribution of ATP P2 receptors to electrically-evoked electrographic seizure activity in hippocampal slices: Evidence from purine biosensors and P2 receptor agonists and antagonists. Neuropharmacology, 61, 25–34. [PubMed: 21338615]
- Lopatar, J., Dale, N. & Frenguelli, B. G. 2015. Pannexin-1-mediated ATP release from area CA3 drives mGlu5-dependent neuronal oscillations. Neuropharmacology, 93, 219–28. [PubMed: 25645390]
- Loscher, W. 2011. Critical review of current animal models of seizures and epilepsy used in the discovery and development of new antiepileptic drugs. Seizure, 20, 359–68. [PubMed: 21292505]
- Loscher, W. 2017. Animal Models of Seizures and Epilepsy: Past, Present, and Future Role for the Discovery of Antiseizure Drugs. Neurochem Res, 42, 1873–1888. [PubMed: 28290134]
- Lovatt, D., Xu, Q., Liu, W., Takano, T., Smith, N. A., Schnermann, J., Tieu, K. & Nedergaard, M. 2012. Neuronal adenosine release, and not astrocytic ATP release, mediates feedback inhibition of excitatory activity. Proc Natl Acad Sci U S A, 109, 6265–70. [PMC free article: PMC3341061] [PubMed: 22421436]
- Maliszewski, C. R., Delespesse, G. J., Schoenborn, M. A., Armitage, R. J., Fanslow, W. C., Nakajima, T., Baker, E., Sutherland, G. R., Poindexter, K., Birks, C. & et al. 1994. The CD39 lymphoid cell activation antigen. Molecular cloning and structural characterization. J Immunol, 153, 3574–83. [PubMed: 7930580]
- Mansoor, S. E., Lu, W., Oosterheert, W., Shekhar, M., Tajkhorshid, E. & Gouaux, E. 2016. X-ray structures define human P2X(3) receptor gating cycle and antagonist action. Nature, 538, 66–71. [PMC free article: PMC5161641] [PubMed: 27626375]
- Mares, P. 2010. Anticonvulsant action of 2-chloroadenosine against pentetrazol-induced seizures in immature rats is due to activation of A1 adenosine receptors. J Neural Transm (Vienna), 117, 1269–77. [PubMed: 20809069]
- Martin, A. J., Dale, N., Imray, C. H. E., Roffe, C., Smith, C. J., Tian, F. & Price, C. I. 2019. The association between early neurological deterioration and whole blood purine concentration during acute stroke. Biomark Res, 7, 7. [PMC free article: PMC6448300] [PubMed: 30988953]
- Martorell, A., Wellmann, M., Guiffa, F., Fuenzalida, M. & Bonansco, C. 2020. P2Y1 receptor inhibition rescues impaired synaptic plasticity and astroglial Ca(2+)-dependent activity in the epileptic hippocampus. Neurobiol Dis, 146, 105132. [PubMed: 33049315]
- Masino, S. A., Li, T., Theofilas, P., Sandau, U. S., Ruskin, D. N., Fredholm, B. B., Geiger, J. D., Aronica, E. & Boison, D. 2011. A ketogenic diet suppresses seizures in mice through adenosine A(1) receptors. J Clin Invest, 121, 2679–83. [PMC free article: PMC3223846] [PubMed: 21701065]
- Masse, K. & Dale, N. 2012. Purines as potential morphogens during embryonic development. Purinergic Signal, 8, 503–21. [PMC free article: PMC3360092] [PubMed: 22270538]
- Meigh, L., Greenhalgh, S. A., Rodgers, T. L., Cann, M. J., Roper, D. I. & Dale, N. 2013. CO(2)directly modulates connexin 26 by formation of carbamate bridges between subunits. Elife, 2, e01213. [PMC free article: PMC3821526] [PubMed: 24220509]
- Mildner, A., Huang, H., Radke, J., Stenzel, W. & Priller, J. 2017. P2Y12 receptor is expressed on human microglia under physiological conditions throughout development and is sensitive to neuroinflammatory diseases. Glia, 65, 375–387. [PubMed: 27862351]
- Milior, G., Morin-Brureau, M., Chali, F., Le Duigou, C., Savary, E., Huberfeld, G., Rouach, N., Pallud, J., Capelle, L., Navarro, V., Mathon, B., Clemenceau, S. & Miles, R. 2020. Distinct P2Y Receptors Mediate Extension and Retraction of Microglial Processes in Epileptic and Peritumoral Human Tissue. J Neurosci, 40, 1373–1388. [PMC free article: PMC7044731] [PubMed: 31896671]
- Miller-Delaney, S. F., Bryan, K., Das, S., Mckiernan, R. C., Bray, I. M., Reynolds, J. P., Gwinn, R., Stallings, R. L. & Henshall, D. C. 2015. Differential DNA methylation profiles of coding and non-coding genes define hippocampal sclerosis in human temporal lobe epilepsy. Brain, 138, 616–31. [PMC free article: PMC4408428] [PubMed: 25552301]
- Miras-Portugal, M. T., Gualix, J., Mateo, J., Diaz-Hernandez, M., Gomez-Villafuertes, R., Castro, E. & Pintor, J. 1999. Diadenosine polyphosphates, extracellular function and catabolism. Prog Brain Res, 120, 397–409. [PubMed: 10551014]
- Miras-Portugal, M. T., Sebastian-Serrano, A., De Diego Garcia, L. & Diaz-Hernandez, M. 2017. Neuronal P2X7 Receptor: Involvement in Neuronal Physiology and Pathology. J Neurosci, 37, 7063–7072. [PMC free article: PMC6705729] [PubMed: 28747389]
- Mitchell, J. B., Lupica, C. R. & Dunwiddie, T. V. 1993. Activity-dependent release of endogenous adenosine modulates synaptic responses in the rat hippocampus. J Neurosci, 13, 3439–47. [PMC free article: PMC6576537] [PubMed: 8393482]
- Monif, M., Reid, C. A., Powell, K. L., Smart, M. L. & Williams, D. A. 2009. The P2X7 receptor drives microglial activation and proliferation: a trophic role for P2X7R pore. J Neurosci, 29, 3781–91. [PMC free article: PMC6665035] [PubMed: 19321774]
- Montilla, A., Mata, G. P., Matute, C. & Domercq, M. 2020. Contribution of P2X4 Receptors to CNS Function and Pathophysiology. Int J Mol Sci, 21(15):5562. [PMC free article: PMC7432758] [PubMed: 32756482]
- Moore, D. J., Murdock, P. R., Watson, J. M., Faull, R. L., Waldvogel, H. J., Szekeres, P. G., Wilson, S., Freeman, K. B. & Emson, P. C. 2003. GPR105, a novel Gi/o-coupled UDP-glucose receptor expressed on brain glia and peripheral immune cells, is regulated by immunologic challenge: possible role in neuroimmune function. Brain Res Mol Brain Res, 118, 10–23. [PubMed: 14559350]
- Morgan, J., Alves, M., Conte, G., Menendez-Mendez, A., De Diego-Garcia, L., De Leo, G., Beamer, E., Smith, J., Nicke, A. & Engel, T. 2020. Characterization of the Expression of the ATP-Gated P2X7 Receptor Following Status Epilepticus and during Epilepsy Using a P2X7-EGFP Reporter Mouse. Neurosci Bull36(11):1242–1258. [PMC free article: PMC7674533] [PubMed: 32895896]
- Moshe, S. L., Perucca, E., Ryvlin, P. & Tomson, T. 2015. Epilepsy: new advances. Lancet, 385, 884–98. [PubMed: 25260236]
- Murugan, M., Fedele, D., Millner, D., Alharfoush, E., Vegunta, G. & Boison, D. 2021. Adenosine Kinase: An Epigenetic Modulator in Development and Disease. Neurochem Int, 105054. [PMC free article: PMC8178237] [PubMed: 33961946]
- Muzzi, M., Coppi, E., Pugliese, A. M. & Chiarugi, A. 2013. Anticonvulsant effect of AMP by direct activation of adenosine A1 receptor. Exp Neurol, 250, 189–93. [PubMed: 24056265]
- Nieoczym, D., Socala, K. & Wlaz, P. 2017. Evaluation of the Anticonvulsant Effect of Brilliant Blue G, a Selective P2X7 Receptor Antagonist, in the iv PTZ-, Maximal Electroshock-, and 6 Hz-Induced Seizure Tests in Mice. Neurochem Res, 42, 3114–3124. [PMC free article: PMC5649599] [PubMed: 28702712]
- Nualart-Marti, A., Del Molino, E. M., Grandes, X., Bahima, L., Martin-Satue, M., Puchal, R., Fasciani, I., Gonzalez-Nieto, D., Ziganshin, B., Llobet, A., Barrio, L. C. & Solsona, C. 2013. Role of connexin 32 hemichannels in the release of ATP from peripheral nerves. Glia, 61, 1976–89. [PubMed: 24123415]
- Ohsawa, K., Irino, Y., Nakamura, Y., Akazawa, C., Inoue, K. & Kohsaka, S. 2007. Involvement of P2X4 and P2Y12 receptors in ATP-induced microglial chemotaxis. Glia, 55, 604–16. [PubMed: 17299767]
- Orellana, J. A., Saez, P. J., Cortes-Campos, C., Elizondo, R. J., Shoji, K. F., Contreras-Duarte, S., Figueroa, V., Velarde, V., Jiang, J. X., Nualart, F., Saez, J. C. & Garcia, M. A. 2012. Glucose increases intracellular free Ca(2+) in tanycytes via ATP released through connexin 43 hemichannels. Glia, 60, 53–68. [PMC free article: PMC3417330] [PubMed: 21987367]
- Pankratov, Y., Lalo, U., Verkhratsky, A. & North, R. A. 2006. Vesicular release of ATP at central synapses. Pflugers Arch, 452, 589–97. [PubMed: 16639550]
- Pankratov, Y., Lalo, U., Verkhratsky, A. & North, R. A. 2007. Quantal release of ATP in mouse cortex. J Gen Physiol, 129, 257–65. [PMC free article: PMC2151610] [PubMed: 17325196]
- Pearson, R. A., Dale, N., Llaudet, E. & Mobbs, P. 2005. ATP released via gap junction hemichannels from the pigment epithelium regulates neural retinal progenitor proliferation. Neuron, 46, 731–44. [PubMed: 15924860]
- Peleli, M., Fredholm, B. B., Sobrevia, L. & Carlstrom, M. 2017. Pharmacological targeting of adenosine receptor signaling. Mol Aspects Med, 55, 4–8. [PubMed: 28088486]
- Pintor, J., Diaz-Rey, M. A., Torres, M. & Miras-Portugal, M. T. 1992. Presence of diadenosine polyphosphates--Ap4A and Ap5A--in rat brain synaptic terminals. Ca2+ dependent release evoked by 4-aminopyridine and veratridine. Neurosci Lett, 136, 141–4. [PubMed: 1641181]
- Pitkanen, A., Lukasiuk, K., Dudek, F. E. & Staley, K. J. 2015. Epileptogenesis. Cold Spring Harb Perspect Med, 5(10):a022822. [PMC free article: PMC4588129] [PubMed: 26385090]
- Quintas, C., Fraga, S., Goncalves, J. & Queiroz, G. 2011. P2Y receptors on astrocytes and microglia mediate opposite effects in astroglial proliferation. Purinergic Signal, 7, 251–63. [PMC free article: PMC3146643] [PubMed: 21559785]
- Ralevic, V. & Burnstock, G. 1998. Receptors for purines and pyrimidines. Pharmacol Rev, 50, 413–92. [PubMed: 9755289]
- Ramirez-Fernandez, A., Urbina-Trevino, L., Conte, G., Alves, M., Rissiek, B., Durner, A., Scalbert, N., Zhang, J., Magnus, T., Koch-Nolte, F., Plesnila, N., Deussing, J. M., Engel, T., Kopp, R. & Nicke, A. 2020. Deviant reporter expression and P2X4 passenger gene overexpression in the soluble EGFP BAC transgenic P2X7 reporter mouse model. Sci Rep, 10, 19876. [PMC free article: PMC7669894] [PubMed: 33199725]
- Raouf, R., Chabot-Dore, A. J., Ase, A. R., Blais, D. & Seguela, P. 2007. Differential regulation of microglial P2X4 and P2X7 ATP receptors following LPS-induced activation. Neuropharmacology, 53, 496–504. [PubMed: 17675190]
- Rebola, N., Porciuncula, L. O., Lopes, L. V., Oliveira, C. R., Soares-Da-Silva, P. & Cunha, R. A. 2005. Long-term effect of convulsive behavior on the density of adenosine A1 and A 2A receptors in the rat cerebral cortex. Epilepsia, 46 Suppl 5, 159–65. [PubMed: 15987272]
- Robel, S., Buckingham, S. C., Boni, J. L., Campbell, S. L., Danbolt, N. C., Riedemann, T., Sutor, B. & Sontheimer, H. 2015. Reactive astrogliosis causes the development of spontaneous seizures. J Neurosci, 35, 3330–45. [PMC free article: PMC4339349] [PubMed: 25716834]
- Robson, S. C., Sevigny, J. & Zimmermann, H. 2006. The E-NTPDase family of ectonucleotidases: Structure function relationships and pathophysiological significance. Purinergic Signal, 2, 409–30. [PMC free article: PMC2254478] [PubMed: 18404480]
- Roseti, C., Martinello, K., Fucile, S., Piccari, V., Mascia, A., Di Gennaro, G., Quarato, P. P., Manfredi, M., Esposito, V., Cantore, G., Arcella, A., Simonato, M., Fredholm, B. B., Limatola, C., Miledi, R. & Eusebi, F. 2008. Adenosine receptor antagonists alter the stability of human epileptic GABAA receptors. Proc Natl Acad Sci U S A, 105, 15118–23. [PMC free article: PMC2567502] [PubMed: 18809912]
- Roseti, C., Van Vliet, E. A., Cifelli, P., Ruffolo, G., Baayen, J. C., Di Castro, M. A., Bertollini, C., Limatola, C., Aronica, E., Vezzani, A. & Palma, E. 2015. GABAA currents are decreased by IL-1beta in epileptogenic tissue of patients with temporal lobe epilepsy: implications for ictogenesis. Neurobiol Dis, 82, 311–320. [PubMed: 26168875]
- Rozmer, K., Gao, P., Araujo, M. G. L., Khan, M. T., Liu, J., Rong, W., Tang, Y., Franke, H., Krugel, U., Fernandes, M. J. S. & Illes, P. 2017. Pilocarpine-Induced Status Epilepticus Increases the Sensitivity of P2X7 and P2Y1 Receptors to Nucleotides at Neural Progenitor Cells of the Juvenile Rodent Hippocampus. Cereb Cortex, 27, 3568–3585. [PubMed: 27341850]
- Sandau, U. S., Yahya, M., Bigej, R., Friedman, J. L., Saleumvong, B. & Boison, D. 2019. Transient use of a systemic adenosine kinase inhibitor attenuates epilepsy development in mice. Epilepsia, 60, 615–625. [PMC free article: PMC6713278] [PubMed: 30815855]
- Sano, F., Shigetomi, E., Shinozaki, Y., Tsuzukiyama, H., Saito, K., Mikoshiba, K., Horiuchi, H., Cheung, D. L., Nabekura, J., Sugita, K., Aihara, M. & Koizumi, S. 2021. Reactive astrocyte-driven epileptogenesis is induced by microglia initially activated following status epilepticus. JCI Insight6(9): e135391. [PMC free article: PMC8262323] [PubMed: 33830944]
- Sawada, K., Echigo, N., Juge, N., Miyaji, T., Otsuka, M., Omote, H., Yamamoto, A. & Moriyama, Y. 2008. Identification of a vesicular nucleotide transporter. Proc Natl Acad Sci U S A, 105, 5683–6. [PMC free article: PMC2311367] [PubMed: 18375752]
- Scemes, E., Velisek, L. & Veliskova, J. 2019. Astrocyte and Neuronal Pannexin1 Contribute Distinctly to Seizures. ASN Neuro, 11, 1759091419833502. [PMC free article: PMC6415468] [PubMed: 30862176]
- Sebastian-Serrano, A., Engel, T., De Diego-Garcia, L., Olivos-Ore, L. A., Arribas-Blazquez, M., Martinez-Frailes, C., Perez-Diaz, C., Millan, J. L., Artalejo, A. R., Miras-Portugal, M. T., Henshall, D. C. & Diaz-Hernandez, M. 2016. Neurodevelopmental alterations and seizures developed by mouse model of infantile hypophosphatasia are associated with purinergic signaling deregulation. Hum Mol Genet, 25, 4143–4156. [PMC free article: PMC5291194] [PubMed: 27466191]
- Shinozaki, Y., Shibata, K., Yoshida, K., Shigetomi, E., Gachet, C., Ikenaka, K., Tanaka, K. F. & Koizumi, S. 2017. Transformation of Astrocytes to a Neuroprotective Phenotype by Microglia via P2Y1 Receptor Downregulation. Cell Rep, 19, 1151–1164. [PubMed: 28494865]
- Simoes, A. P., Silva, C. G., Marques, J. M., Pochmann, D., Porciuncula, L. O., Ferreira, S., Oses, J. P., Beleza, R. O., Real, J. I., Kofalvi, A., Bahr, B. A., Lerma, J., Cunha, R. A. & Rodrigues, R. J. 2018. Glutamate-induced and NMDA receptor-mediated neurodegeneration entails P2Y1 receptor activation. Cell Death Dis, 9, 297. [PMC free article: PMC5833818] [PubMed: 29463792]
- Sims, R. E. & Dale, N. 2014. Activity-dependent adenosine release may be linked to activation of Na(+)-K(+) ATPase: an in vitro rat study. PLoS One, 9, e87481. [PMC free article: PMC3906196] [PubMed: 24489921]
- Sims, R. E., Wu, H. H. & Dale, N. 2013. Sleep-wake sensitive mechanisms of adenosine release in the basal forebrain of rodents: an in vitro study. PLoS One, 8, e53814. [PMC free article: PMC3543262] [PubMed: 23326515]
- Soni, N., Koushal, P., Reddy, B. V., Deshmukh, R. & Kumar, P. 2015. Effect of GLT-1 modulator and P2X7 antagonists alone and in combination in the kindling model of epilepsy in rats. Epilepsy Behav, 48, 4–14. [PubMed: 26037843]
- Stefan, C., Jansen, S. & Bollen, M. 2005. NPP-type ectophosphodiesterases: unity in diversity. Trends Biochem Sci, 30, 542–50. [PubMed: 16125936]
- Stellwagen, D., Beattie, E. C., Seo, J. Y. & Malenka, R. C. 2005. Differential regulation of AMPA receptor and GABA receptor trafficking by tumor necrosis factor-alpha. J Neurosci, 25, 3219–28. [PMC free article: PMC6725093] [PubMed: 15788779]
- Stout, C., Goodenough, D. A. & Paul, D. L. 2004. Connexins: functions without junctions. Curr Opin Cell Biol, 16, 507–12. [PubMed: 15363800]
- Stout, C. E., Costantin, J. L., Naus, C. C. & Charles, A. C. 2002. Intercellular calcium signaling in astrocytes via ATP release through connexin hemichannels. J Biol Chem, 277, 10482–8. [PubMed: 11790776]
- Sukigara, S., Dai, H., Nabatame, S., Otsuki, T., Hanai, S., Honda, R., Saito, T., Nakagawa, E., Kaido, T., Sato, N., Kaneko, Y., Takahashi, A., Sugai, K., Saito, Y., Sasaki, M., Goto, Y., Koizumi, S. & Itoh, M. 2014. Expression of astrocyte-related receptors in cortical dysplasia with intractable epilepsy. J Neuropathol Exp Neurol, 73, 798–806. [PubMed: 25003238]
- Sun, H., Ma, L., Zhang, Y., Pan, X., Wang, C., Zhang, J., Zhang, X., Sun, H., Wang, Q. & Zhu, W. 2018. A Purinergic P2 Receptor Family-Mediated Increase in Thrombospondin-1 Bolsters Synaptic Density and Epileptic Seizure Activity in the Amygdala-Kindling Rat Model. Front Cell Neurosci, 12, 302. [PMC free article: PMC6199899] [PubMed: 30386206]
- Taruno, A., Vingtdeux, V., Ohmoto, M., Ma, Z., Dvoryanchikov, G., Li, A., Adrien, L., Zhao, H., Leung, S., Abernethy, M., Koppel, J., Davies, P., Civan, M. M., Chaudhari, N., Matsumoto, I., Hellekant, G., Tordoff, M. G., Marambaud, P. & Foskett, J. K. 2013. CALHM1 ion channel mediates purinergic neurotransmission of sweet, bitter and umami tastes. Nature, 495, 223–6. [PMC free article: PMC3600154] [PubMed: 23467090]
- Tescarollo, F. C., Rombo, D. M., Deliberto, L. K., Fedele, D. E., Alharfoush, E., Tome, A. R., Cunha, R. A., Sebastiao, A. M. & Boison, D. 2020. Role of Adenosine in Epilepsy and Seizures. J Caffeine Adenosine Res, 10, 45–60. [PMC free article: PMC7301316] [PubMed: 32566903]
- Thakore, P., Alvarado, M. G., Ali, S., Mughal, A., Pires, P. W., Yamasaki, E., Pritchard, H. A., Isakson, B. E., Tran, C. H. T. & Earley, S. 2021. Brain endothelial cell TRPA1 channels initiate neurovascular coupling. Elife, 10. [PMC free article: PMC7935492] [PubMed: 33635784]
- Thijs, R. D., Surges, R., O’brien, T. J. & Sander, J. W. 2019. Epilepsy in adults. Lancet, 393, 689–701. [PubMed: 30686584]
- Tian, F., Bibi, F., Dale, N. & Imray, C. H. E. 2017. Blood purine measurements as a rapid real-time indicator of reversible brain ischaemia. Purinergic Signal, 13, 521–528. [PMC free article: PMC5714841] [PubMed: 28803399]
- Tian, F., Llaudet, E. & Dale, N. 2007. Ruthenium purple-mediated microelectrode biosensors based on sol-gel film. Anal Chem, 79, 6760–6. [PubMed: 17672522]
- Timmers, M., Ravenstijn, P., Xi, L., Triana-Baltzer, G., Furey, M., Van Hemelryck, S., Biewenga, J., Ceusters, M., Bhattacharya, A., Van Den Boer, M., Van Nueten, L. & De Boer, P. 2018. Clinical pharmacokinetics, pharmacodynamics, safety, and tolerability of JNJ-54175446, a brain permeable P2X7 antagonist, in a randomised single-ascending dose study in healthy participants. J Psychopharmacol, 32, 1341–1350. [PubMed: 30260294]
- Tosh, D. K., Paoletta, S., Deflorian, F., Phan, K., Moss, S. M., Gao, Z. G., Jiang, X. & Jacobson, K. A. 2012. Structural sweet spot for A1 adenosine receptor activation by truncated (N)-methanocarba nucleosides: receptor docking and potent anticonvulsant activity. J Med Chem, 55, 8075–90. [PMC free article: PMC3463139] [PubMed: 22921089]
- Ulmann, L., Levavasseur, F., Avignone, E., Peyroutou, R., Hirbec, H., Audinat, E. & Rassendren, F. 2013. Involvement of P2X4 receptors in hippocampal microglial activation after status epilepticus. Glia, 61, 1306–19. [PubMed: 23828736]
- Valera, S., Hussy, N., Evans, R. J., Adami, N., North, R. A., Surprenant, A. & Buell, G. 1994. A new class of ligand-gated ion channel defined by P2x receptor for extracellular ATP. Nature, 371, 516–9. [PubMed: 7523951]
- Van De Wiel, J., Meigh, L., Bhandare, A., Cook, J., Nijjar, S., Huckstepp, R. & Dale, N. 2020. Connexin26 mediates CO2-dependent regulation of breathing via glial cells of the medulla oblongata. Commun Biol, 3, 521. [PMC free article: PMC7505967] [PubMed: 32958814]
- Van Gompel, J. J., Bower, M. R., Worrell, G. A., Stead, M., Chang, S. Y., Goerss, S. J., Kim, I., Bennet, K. E., Meyer, F. B., Marsh, W. R., Blaha, C. D. & Lee, K. H. 2014. Increased cortical extracellular adenosine correlates with seizure termination. Epilepsia, 55, 233–44. [PMC free article: PMC4104491] [PubMed: 24483230]
- Vezzani, A., Balosso, S. & Ravizza, T. 2019. Neuroinflammatory pathways as treatment targets and biomarkers in epilepsy. Nat Rev Neurol, 15, 459–472. [PubMed: 31263255]
- Vezzani, A., Conti, M., De Luigi, A., Ravizza, T., Moneta, D., Marchesi, F. & De Simoni, M. G. 1999. Interleukin-1beta immunoreactivity and microglia are enhanced in the rat hippocampus by focal kainate application: functional evidence for enhancement of electrographic seizures. J Neurosci, 19, 5054–65. [PMC free article: PMC6782637] [PubMed: 10366638]
- Vianna, E. P., Ferreira, A. T., Dona, F., Cavalheiro, E. A. & Da Silva Fernandes, M. J. 2005. Modulation of seizures and synaptic plasticity by adenosinergic receptors in an experimental model of temporal lobe epilepsy induced by pilocarpine in rats. Epilepsia, 46 Suppl 5, 166–73. [PubMed: 15987273]
- Vianna, E. P., Ferreira, A. T., Naffah-Mazzacoratti, M. G., Sanabria, E. R., Funke, M., Cavalheiro, E. A. & Fernandes, M. J. 2002. Evidence that ATP participates in the pathophysiology of pilocarpine-induced temporal lobe epilepsy: fluorimetric, immunohistochemical, and Western blot studies. Epilepsia, 43 Suppl 5, 227–9. [PubMed: 12121326]
- Vihko, P., Virkkunen, P., Henttu, P., Roiko, K., Solin, T. & Huhtala, M. L. 1988. Molecular cloning and sequence analysis of cDNA encoding human prostatic acid phosphatase. FEBS Lett, 236, 275–81. [PubMed: 2842184]
- Von Kugelgen, I. 2021. Molecular pharmacology of P2Y receptor subtypes. Biochem Pharmacol, 187, 114361. [PubMed: 33309519]
- Wall, M. & Dale, N. 2008. Activity-dependent release of adenosine: a critical re-evaluation of mechanism. Curr Neuropharmacol, 6, 329–37. [PMC free article: PMC2701281] [PubMed: 19587854]
- Wall, M. J. & Dale, N. 2007. Auto-inhibition of rat parallel fibre-Purkinje cell synapses by activity-dependent adenosine release. J Physiol, 581, 553–65. [PMC free article: PMC2075183] [PubMed: 17347275]
- Wall, M. J. & Dale, N. 2013. Neuronal transporter and astrocytic ATP exocytosis underlie activity-dependent adenosine release in the hippocampus. J Physiol, 591, 3853–71. [PMC free article: PMC3764633] [PubMed: 23713028]
- Wang, S., Cheng, Q., Malik, S. & Yang, J. 2000a. Interleukin-1beta inhibits gamma-aminobutyric acid type A (GABA(A)) receptor current in cultured hippocampal neurons. J Pharmacol Exp Ther, 292, 497–504. [PubMed: 10640285]
- Wang, Z., Haydon, P. G. & Yeung, E. S. 2000b. Direct observation of calcium-independent intercellular ATP signaling in astrocytes. Anal Chem, 72, 2001–7. [PubMed: 10815957]
- Webb, T. E., Simon, J., Krishek, B. J., Bateson, A. N., Smart, T. G., King, B. F., Burnstock, G. & Barnard, E. A. 1993. Cloning and functional expression of a brain G-protein-coupled ATP receptor. FEBS Lett, 324, 219–25. [PubMed: 8508924]
- Weissman, T. A., Riquelme, P. A., Ivic, L., Flint, A. C. & Kriegstein, A. R. 2004. Calcium waves propagate through radial glial cells and modulate proliferation in the developing neocortex. Neuron, 43, 647–61. [PubMed: 15339647]
- Weltha, L., Reemmer, J. & Boison, D. 2019. The role of adenosine in epilepsy. Brain Res Bull, 151, 46–54. [PMC free article: PMC6527499] [PubMed: 30468847]
- Werman, R. 1966. Criteria for identification of a central nervous system transmitter. Comp Biochem Physiol, 18, 745–66. [PubMed: 5338553]
- Wieraszko, A. & Seyfried, T. N. 1989. ATP-induced synaptic potentiation in hippocampal slices. Brain Res, 491, 356–9. [PubMed: 2765890]
- Wu, P. H. & Phillis, J. W. 1978. Distribution and release of adenosine triphosphate in rat brain. Neurochem Res, 3, 563–71. [PubMed: 370672]
- Xia, J., Wang, H., Zhang, Q. & Han, Z. 2018. Modulation of P2X Purinoceptor 3 (P2X3) in Pentylenetetrazole-Induced Kindling Epilepsy in Rats. Med Sci Monit, 24, 6165–6177. [PMC free article: PMC6134887] [PubMed: 30181529]
- Yang, S., Liu, Z. W., Wen, L., Qiao, H. F., Zhou, W. X. & Zhang, Y. X. 2005. Interleukin-1beta enhances NMDA receptor-mediated current but inhibits excitatory synaptic transmission. Brain Res, 1034, 172–9. [PubMed: 15713269]
- Zhang, D., Gao, Z. G., Zhang, K., Kiselev, E., Crane, S., Wang, J., Paoletta, S., Yi, C., Ma, L., Zhang, W., Han, G. W., Liu, H., Cherezov, V., Katritch, V., Jiang, H., Stevens, R. C., Jacobson, K. A., Zhao, Q. & Wu, B. 2015. Two disparate ligand-binding sites in the human P2Y1 receptor. Nature, 520, 317–21. [PMC free article: PMC4408927] [PubMed: 25822790]
- Zhang, J., Zhang, K., Gao, Z. G., Paoletta, S., Zhang, D., Han, G. W., Li, T., Ma, L., Zhang, W., Muller, C. E., Yang, H., Jiang, H., Cherezov, V., Katritch, V., Jacobson, K. A., Stevens, R. C., Wu, B. & Zhao, Q. 2014. Agonist-bound structure of the human P2Y12 receptor. Nature, 509, 119–22. [PMC free article: PMC4128917] [PubMed: 24784220]
- Zhang, Z., Chen, G., Zhou, W., Song, A., Xu, T., Luo, Q., Wang, W., Gu, X. S. & Duan, S. 2007. Regulated ATP release from astrocytes through lysosome exocytosis. Nat Cell Biol, 9, 945–53. [PubMed: 17618272]
- Zhao, X. F., Liao, Y., Alam, M. M., Mathur, R., Feustel, P., Mazurkiewicz, J. E., Adamo, M. A., Zhu, X. C. & Huang, Y. 2020. Microglial mTOR is Neuronal Protective and Antiepileptogenic in the Pilocarpine Model of Temporal Lobe Epilepsy. J Neurosci, 40, 7593–7608. [PMC free article: PMC7531547] [PubMed: 32868461]
- Zimmermann, H. 2006. Ectonucleotidases in the nervous system. Novartis Found Symp, 276, 113–28; discussion 128-30, 233–7, 275–81. [PubMed: 16805426]
- Zimmermann, H. 2009. Prostatic acid phosphatase, a neglected ectonucleotidase. Purinergic Signal, 5, 273–5. [PMC free article: PMC2717315] [PubMed: 19322680]
- Review Targeting Neuroinflammation via Purinergic P2 Receptors for Disease Modification in Drug-Refractory Epilepsy.[J Inflamm Res. 2021]Review Targeting Neuroinflammation via Purinergic P2 Receptors for Disease Modification in Drug-Refractory Epilepsy.Engel T, Smith J, Alves M. J Inflamm Res. 2021; 14:3367-3392. Epub 2021 Jul 18.
- Review ATPergic signalling during seizures and epilepsy.[Neuropharmacology. 2016]Review ATPergic signalling during seizures and epilepsy.Engel T, Alves M, Sheedy C, Henshall DC. Neuropharmacology. 2016 May; 104:140-53. Epub 2015 Nov 6.
- Context-Specific Switch from Anti- to Pro-epileptogenic Function of the P2Y(1) Receptor in Experimental Epilepsy.[J Neurosci. 2019]Context-Specific Switch from Anti- to Pro-epileptogenic Function of the P2Y(1) Receptor in Experimental Epilepsy.Alves M, De Diego Garcia L, Conte G, Jimenez-Mateos EM, D'Orsi B, Sanz-Rodriguez A, Prehn JHM, Henshall DC, Engel T. J Neurosci. 2019 Jul 3; 39(27):5377-5392. Epub 2019 May 2.
- Review The Metabotropic Purinergic P2Y Receptor Family as Novel Drug Target in Epilepsy.[Front Pharmacol. 2018]Review The Metabotropic Purinergic P2Y Receptor Family as Novel Drug Target in Epilepsy.Alves M, Beamer E, Engel T. Front Pharmacol. 2018; 9:193. Epub 2018 Mar 7.
- Review ATP and adenosine-Two players in the control of seizures and epilepsy development.[Prog Neurobiol. 2021]Review ATP and adenosine-Two players in the control of seizures and epilepsy development.Beamer E, Kuchukulla M, Boison D, Engel T. Prog Neurobiol. 2021 Sep; 204:102105. Epub 2021 Jun 16.
- Purinergic Signaling in Epilepsy - Jasper's Basic Mechanisms of the EpilepsiesPurinergic Signaling in Epilepsy - Jasper's Basic Mechanisms of the Epilepsies
Your browsing activity is empty.
Activity recording is turned off.
See more...