Abstract
Adenosine is an endogenous anticonvulsant and neuroprotectant of the brain. Seizure activity produces large quantities of adenosine, and it is this seizure-induced adenosine surge that normally stops a seizure. However, within the context of epilepsy, adenosine plays a wide spectrum of different roles. It not only controls seizures (ictogenesis), but also plays a major role in processes that turn a normal brain into an epileptic brain (epileptogenesis). It is involved in the control of abnormal synaptic plasticity and neurodegeneration and plays a major role in the expression of comorbid symptoms and complications of epilepsy, such as sudden unexpected death in epilepsy (SUDEP). Given the important role of adenosine in epilepsy, therapeutic strategies are in development with the goal to utilize adenosine augmentation not only for the suppression of seizures but also for disease modification and epilepsy prevention, as well as strategies to block adenosine A2A receptor overfunction associated with neurodegeneration. This review provides a comprehensive overview of the role of adenosine in epilepsy.
Keywords: adenosine, adenosine receptors, adenosine kinase, epilepsy, seizure, epileptogenesis
Introduction
Because epilepsy has traditionally been considered a condition based on an imbalance of neuronal excitation and inhibition, conventional antiseizure drugs (ASDs) act by blunting neuronal transmission.1 Unfortunately, those treatments are exclusively symptomatic, they do not work in more than one third of all persons with epilepsy, fail to affect underlying pathogenic processes, and also fail to affect the comorbidities of epilepsy.2,3
In addition to the well-characterized impairment of neurotransmission and a shift in the excitatory/inhibitory balance toward more excitation, there is now growing consensus that epilepsy is a complex network disorder with a profound derailment of metabolic functions, as well as maladaptive responses of glial function and inflammatory processes which play a major role in the pathogenesis and pathophysiology of epilepsy.4–10 Therefore, novel therapeutic approaches are needed. Network pharmacology offers new options to treat epilepsy more holistically, with the prospect of affecting underlying pathogenic processes (i.e., epileptogenesis)11 and the potential to prevent the development of epilepsy or its progression.12
Because adenosine has multiple functions globally controlling allostasis,13 it is ideally suited to act as a prototypic network modulator. Through a combination of adenosine receptor (AR)-dependent and -independent effects, adenosine has emerged not only as a potent seizure suppressor14 but also as an antiepileptogenic, disease modifying agent.15,16
Epilepsy and Seizures
Epilepsy is the fourth most common neurological disorder, affecting more than 70 million persons worldwide according to the International League Against Epilepsy (ILAE). Epilepsy is a complex group of cerebral disorders characterized by a lasting predisposition for the occurrence of unpredictable spontaneous and recurrent epileptic seizures that are a consequence of anomalous brain activity.17–19 Accordingly, epilepsy exists when a person has an epileptic seizure and their brain “demonstrates a pathologic and enduring tendency to have recurrent seizures,”19 and an epileptic seizure is defined as “a transient occurrence of signs and/or symptoms due to abnormal excessive or synchronous neuronal activity in the brain”19 that lasts from 10 seconds to several minutes.20 Ictogenesis is the generation of a seizure by an epileptic brain.20 It is a progressive process that leads to synchronous high-frequency electroencephalographic activity, which triggers an epileptic seizure.21 Therefore, ictogenesis can occur spontaneously or be objectively related to a stimulus, as in cases of reflex seizures.22–24
Although a large number of ASDs are available for controlling epileptic seizures, they are symptomatic treatments and fail to prevent epilepsy developments.25 In addition, over 35% of persons with epilepsy have intractable epileptic seizures, in which ASDs have no effect on treating the symptoms.2,3 A major risk factor for those with epilepsy is sudden unexpected death in epilepsy (SUDEP).26–29
Epileptogenesis
Epileptogenesis is the process of progressive neurobiological alterations that converts a nonepileptic brain into a brain capable of generating spontaneous and recurrent seizures.30,31
Approximately 20–60% of all epilepsies are caused by acute insults to the brain, such as traumatic brain injury, status epilepticus (SE), cerebrovascular insult, or infections, which trigger a cascade of events, including inflammatory processes, microglial, and astroglial activation, as well as epigenetic changes, which can trigger early seizures within 1 week after the injury.10,32 However, after a primary acute insult to the brain, a “clinically silent” latent period of epileptogenesis is often observed that is characterized by a lack of a clinical phenotype but ongoing remodeling processes that can last from days to years between the insult and the onset of clinically defined epilepsy.10 During this seizure-free period, evidence of pathophysiological alterations in the brain are apparent in both patients and animal models of posttraumatic epilepsy.10,32 These alterations include metabolic impairments, increased inflammatory processes, molecular reorganization, changes in gene and voltage-gated channels and receptor expression, astrogliosis, neurogenesis, aberrant synaptic plasticity, remodeling of neuronal circuits, axonal sprouting, neuronal loss, and epigenetic reprogramming that includes DNA hypermethylation.10
In addition, mesial temporal lobe epilepsy (MTLE), the most common form of acquired epilepsy in adults33 with a high incidence of pharmacoresistence,34 is frequently, but not always, associated with hippocampal sclerosis (HS), which includes hippocampal tissue atrophy and stiffening as a result of intense neuronal loss in CA1 and CA3 subfields and further aberrant astrocyte proliferation, known as reactive astrogliosis35,36 as well as neuronal circuit rebuilding, which includes mossy fiber sprouting and granule cell dispersion.34,37,38 These changes during epileptogenesis result in persistently increased neuronal excitability, which is a determinant for seizures and for epilepsy progression, including interictal discharges, high-frequency oscillations, and for augmented responses to stimuli.30,39
Although for a long time, it was thought that epileptogenesis is a restricted process over a limited time, evidence is recently emerging that epileptogenesis is an extended and continuous process that continues beyond the expression of spontaneous seizures.32,40 Therefore, it is becoming crucial to develop therapies addressing the epileptogenic process rather than only the symptoms of epilepsy.
Role of ARs in Synaptic Function, Synaptic Plasticity, and Synaptotoxicity in Epilepsy
The antiictogenic potential of adenosine has been recognized for the first time 45 years ago by identifying an inhibitory action of adenosine on neuronal activity.41 Since then, experimental studies have shown that maladaptative changes in G-protein coupled ARs (A1 receptor [A1R], A2AR, A2BR and A3R) expression and signaling contribute to the pathophysiology of epilepsy.42
The actions of adenosine in the brain are mostly mediated by A1R and A2AR, whereas the possible central roles of A2BR and A3R are still poorly studied.42
ARs were first classified according to their differential coupling to adenylyl cyclase to regulate cyclic adenosine monophosphate (cAMP) levels. A1Rs and A3ARs are coupled to Gi/o proteins, leading to the inhibition of cAMP production and leading to the inhibition of presynaptic glutamate release. A1R activation may also increase phospholipase C (PLC) activity and directly couple to and activate K+ channels and Q, N, and P type voltage sensitive Ca2+ channels.42,43 In contrast, A2ARs and A2BRs are pleiotropic receptors, mostly coupled to Gs/olf proteins, increasing the production of cAMP, leading to the activation of protein kinase A (PKA), which in turn leads to the recruitement and opening of Ca2+ channels, increasing cell excitability.42
In the brain, A1Rs are widely distributed on neurons, mostly located in excitatory synapses,44–46 astrocytes,47 oligodendrocytes,48 and microglia49 in the cortex, hippocampus, and cerebellum.42 A2ARs have a more restrictive and heterogenous distribution in the brain with higher levels in the nucleus accumbens shell, striatum, and tuberculum olfatorium,50 whereas lower levels were found in hippocampus, cerebral cortex, amygdala, and cerebellum.51 A2ARs are not only most densely located in synapses of neurons52 but also present in microglia53,54 and astrocytes.43,53,55
Detailed in vitro studies of circuits of one of the most afflicted regions in MTLE—the hippocampus—then followed56 and shortly after, Dunwiddie and Worth in 198257 associated the anticonvulsant action of adenosine with A1R activation. Since then, it has been repeatedly reported that endogenous adenosine, as well as systemically administered selective A1R agonists, such as 2-chloro-N6-cyclopentyl-adenosine (CCPA) or N6-cyclohexyladenosine (CHA), possess anticonvulsant actions in a variety of acute and chronic models of epilepsy, which are prevented by A1R antagonists such as 8-cyclopentyl-1,3-dimethylxanthine (DPX) or 8-cyclopentyl-1,3-dipropylxanthine (DPCPX).58
Importantly, so far available information shows that A1R activation is not only capable of preventing seizure occurrence when administered before the trigger but also to abort an already emerging seizure event, thus working as a true anticonvulsant substance.
The hyperpolarizing effect of A1R activation is still considered one of the main mechanisms that contribute to the A1R-dependent suppression of seizures, but other synaptic mechanisms likely coexist. In fact, presynaptic actions of A1Rs control epileptiform activity by altering glutamatergic transmission, in accordance with the predominant presynaptic localization of the A1R,44,59 mainly in glutamatergic terminals.46 This includes reduction of the probability of glutamate release through inhibition of voltage-dependent Ca2+ channels60–62 or reduction of Ca2+-independent spontaneous release.63,64
Postsynaptic actions are mostly related to a control of the recruitment of AMPA and NMDA receptors,65 since both are implicated in the initiation and establishment of epileptic seizures.66–69 In this sense, suppression of A1R tonus in vitro causes sustained synaptically mediated epileptiform activity that has an AMPAR-dependent70,71 and NMDAR-dependent component.72 A1R also directly influence GABA function in several brain areas.73 It is, however, important to stress that A1Rs supress tonic extrasynaptic GABAAR currents in pyramidal cells and in a specific population of postsynaptic GABAergic inhibitory neurons, the CB1R-expressing interneurons,74 not affecting phasic GABAergic inputs to glutamatergic neurons.75–77 These effects may be of particular importance during hyperexcitable conditions to reduce disinhibitoy actions of interneurons to pyramidal neurons.
When evaluating alterations of the adenosine modulation system that may prompt likely therapeutic targets, one concludes that both adenosine metabolism and ARs are modified. Thus, there is an increased activity of adenosine kinase (ADK), the major enzyme for the metabolic clearance of adenosine, which removes adenosine by phosphorylating it to AMP,78 and an expected decreased formation of astrocyte-derived adenosine79 associated with a lower tonic activation of A1R in animal models of epilepsy reviewed in.80 In parallel, there is a decreased density of A1R in excitatory synapses in different animal models of epilepsy,45,46,81–83 as well as in patients with epilepsy.84 This decrease of A1R density results from the repeated activation of A1R, which tend to develop tolerance in the control of excitatory transmission85 and a gradual loss of their anticonvulsant capacity.86
Furthermore, peripheral actions of systemically administered A1R agonists, mostly related to cardiovascular adverse effects such as decrease in arterial blood pressure and heart rate,87,88 have prevented the use of A1R agonists for the treatment of central nervous system diseases.
With regard to the role of A2AR and epilepsy, there is a false impression of inconsistency of results. In fact, major evidences indicate that the tonic activation of A2AR by endogenous adenosine results in proepileptic effects, although the activation with exogenous agonists, such as 2-p-(2-carboxyethyl)phenethylamino-5′-N-ethylcarboxamidoadenosine hydrochloride (CGS21680), of different populations of A2AR89,90 can cause a paradoxical attenuation of seizure activity.
Some animal studies claimed that activation of A2AR with exogenously added agonists 2-hexynyl-5′-N-ethylcarboxamidoadenosine (2HE-NECA)86 or CGS2168091,92 contributed to seizure suppression in acute models of seizure and that its blockade with the antagonist 3,7-dimethyl-1-propargylxanthine (DMPX) had proconvulsant actions93; however, these findings probably result from the activation of A1R rather than A2AR since DMPX has an A2A/A1 selectivity ratio of 3 in rats and 5 in cloned human A1R94 and the anticonvulsant effect of CGS21680, as well as other central effects,95 has been shown to be abrogated by the selective A1R antagonist, DPCPX.96
Most studies using both selective antagonists and genetic manipulations of A2AR have consistently demonstrated proconvulsant effects of endogenously activated A2AR.97–101 Notably, the genetic ablation or pharmacological inhibition of A2AR in animal models of epilepsy mostly prevents proconvulsant epileptogenic changes during evolution of kindling.102–104 These results are indicative of a main role of A2ARs in the cumulative aggravation of convulsions rather than in the onset of seizure activity.
The neuroprotective role of the A2AR antagonist istradefylline (KW-6002) was also documented to dampen long-term deleterious consequences of early-life convulsions105 and tinkering with the ability of A2AR to control the migration of cortical interneurons during brain development in mice results in increased susceptibility to seizures.106 Moreover, it has also been reported that genetic variations in the human adenosine A2AR gene (ADORA2A), associated with increased expression of A2AR and higher levels of cAMP production, is a predisposing factor for childhood encephalopathy following severe febrile seizures.107 Overall, these evidences implicate a deleterious role of endogenous A2AR activation in epilepsy.
A putative gain of function of A2AR during epileptogenesis is best heralded by the reported increased density of A2AR in different animal models of epilepsy,46,104,108,109 as well as in patients with epilepsy,110 which is most evident in excitatory synapses.46,104 Clearly, multiple evidences suggest that both decreased A1R and increased A2AR function contribute to the establishment of epileptic conditions.111 Thus, experimental data support a key role of A1R as a hurdle to the spreading of abnormal neuronal function in animal models of epilepsy.112–114 This prompts the suggestion that a combination of controlling A1R desensitization115 and attenuating A2AR function should be an effective strategy to control epilepsy.
Much less is known about the role of A3R in epilepsy, in line with the current view that the central effects of adenosine mostly involve A1R and A2AR.116 Due to their lower affinity for adenosine, A3R are activated only when relatively high concentrations of extracellular adenosine are reached, as occurs during high frequency neuronal discharges. Knowledge about the endogenous role of A3R to epilepsy is still limited. While there is still conflicting information about whether the activation of A3R shows a pro- or anticonvulsant effect,93,117–121 it is suggested that A3R alter the stability of GABAAR, thereby fine-tuning neuronal excitability.118 A heterologous desensitization of A1R has been described after activation of A3R by high levels of adenosine, which could explain a possible proconvulsant effect of A3R activation.122
The scarce understanding of the role of A3Rs in epilepsy, together with some evidence for their dual role in seizure control, makes it even more difficult to understand their mechanism of action. Not surprisingly then, very few studies addressed the synaptic mechanisms that might be linked to A3R in epilepsy. These effects may be direct or indirect, mediated through A1Rs, influencing glutamatergic transmission presynaptically or postsynaptic K+ channel function,122 or even causing desensitization of GABAARs.123
AR-Independent Effects of Adenosine
Epigenetic changes such as histone modification, DNA methylation, and noncoding RNAs have been recently identified as novel AR-independent roles of adenosine. These epigenetic changes play important roles in controlling gene expression without changing the DNA sequence.16 Thereafter, alterations due to these factors have been linked to several neurological disorders.124 In particular, DNA hypermethylation has been associated with the development of epilepsy giving rise to the methylation hypothesis of epileptogenesis.125–131
In addition to the receptor-mediated effects of adenosine mentioned above, adenosine can act as an endogenous regulator of DNA methyltransferase activity.16 Adenosine is a product of the transmethylation pathway, which ultimately converts S-adenosylmethionine (SAM) into adenosine and homocysteine via methyltransferases and S-adenosylhomocysteine (SAH) hydrolase.132 As a product of the transmethylation pathway, changes in adenosine levels can have a major effect on DNA methylation, which proceeds only if adenosine and homocysteine are effectively metabolized by ADK.132 Therefore, dysregulation of the intracellular metabolism of adenosine during epileptogenesis is directly linked to epigenetic changes thought to drive the development of epilepsy.
Disruption of Adenosine Metabolism in Epilepsy
Astroglial dysfunction in epilepsy
Astrocytes are the most abundant cells in the brain of higher mammalian species.133 Among other functions, astrocytes provide energetic and homeostatic support for neurons134; guide the formation and function of synapses5,135–137; and also perform a crucial role in the blood–brain barrer creating a tight interface in the brain that regulates the exchange of molecules in and out of the brain to maintain homeostasis.138
Astrogliosis, also known as reactive astrogliosis, is a prevalent pathological hallmark of many forms of epilepsy, including MTLE,139 and refers to a broad spectrum of functional and structural alterations in glial cells, particularly astrocytes and microglia.140 In MTLE, an important feature of astrogiosis is HS that is characterized by loss of neurons in the hippocampus, induction of hypertrophy of astroglial cell bodies and processes, upregulation of the expression of several proteins, including intermediate filament proteins such as glial fibrillary acidic protein (GFAP) and vimentin. In addition to cellular proliferation, the formation of a glial scar rich in chondroitin sulfate and proteoglycans is also observed in HS.141,142
Functionally, astrogliosis may be implicated in seizure generation and also, most importantly, in epileptogenesis.5,136 For example, dysregulation of astrocytic activity may increase epileptiform activity through the release of gliotransmitters, including glutamate, D-serine, and ATP, which increases the excitability of neurons.143 Therefore, impairment of the homeostatic control of network excitability can be a consequence of astrocyte activation. This might involve impairment of K+-buffering,136 dysfunctional gap junctions,144 dysfunctional glutamate homeostasis,145 changes in calcium buffering and gliotransmitter release,146 and dysfunctional adenosine homeostasis5,8,147 and modulation.148 On the contrary, astrogliosis may occur as a compensatory mechanism.
Astrocytes can reduce neuronal excitability by facilitating the reuptake of excessive glutamate released during a seizure.143 Moreover, astrogliosis alone seems to be capable of inducing spontaneous recurrent seizures.149
The ADK hypothesis of epileptogenesis
As mentioned before, ADK is the major enzyme for the metabolic clearance of adenosine by phosphorylating it to AMP.78 In the adult brain, ADK is predominantly expressed in astrocytes.150 After acute insults, such as a SE or a stroke, ADK is rapidly downregulated, contributing to a large astrocytic-derived surge in extracellular adenosine, which serves to terminate seizures and to provide neuroprotection.78,151 On the contrary, high amounts of ATP-derived adenosine released into the synaptic cleft can contribute to synaptotoxicity where adenosine activates A2AR on neurons and leads to an increase of intracellular calcium concentrations.71,104,152–154
During the clinically silent latent period of epileptogenesis, the inflammatory responses triggered after the acute insult lead to astrogliosis.6,155 Moreover, experimental data show an important association of astrogliosis with overexpression of ADK, which results in a chronic deficiency in the availability of adenosine.156 Accordingly, results obtained from studies in both animal models of epileptogenesis147 and specimens from human patients with MTLE associated with HS, who underwent surgical resection of their hippocampus as a treatment of epilepsy, showed that ADK is robustly overexpressed in those brain areas involved in epileptogenesis.157–160 Indeed, ADK overexpression by itself is sufficient to trigger seizures.151,160
Evidence described by Li et al. attest that transgenic Adk-tg mice, that intrinsically overexpress ADK, experience spontaneous recurrent seizures limited to astrogliotic hippocampal CA3 that were similar in rate, frequency, and duration to those in a mouse model of intra-amygdaloid kainic acid (KA)-induced focal epilepsy.161 Taken together, these observations lead to the hypothesis that overexpression of ADK is a crucial mechanistic contributor to the development of epilepsy.
Altered purinergic modulation of synaptic function in epilepsy
Despite the evidence supporting a role for ADK modifications during epileptogenesis, the link between the altered ADK activity during epileptogenesis and the alteration of neuronal function to trigger the hyperexcitability and hypersynchronicity characteristics of circuits engaged in epileptic seizures is still poorly understood. A possible involvement of the purinergic system in this astrocyte-to-neuron adaptive process is proposed, based on the role of astrocytic ATP-derived adenosine to activate A1R to control heterosynaptic depression,69,146,162,163 the alteration of the function of A2AR in excitatory synapses upon tinkering with astrocytes,148 with astrocytic ADK164 or ARs.148 However, the exact mechanisms underlying a possible role of purines in the astrocyte-to-neuron adaptive processes underlying epileptogenesis still remain to be experimentally scrutinized.
Irrespective of the trigger(s) of the alterations engaging epileptogenesis, it is clear that epileptic phenotypes are cumulative in the sense that seizures aggravate subsequent seizures in a process designated as seizures beget seizures; conversely, the control of seizure activity is a sine qua non condition to successfully manage epilepsy and hopefully treat these disorders.165,166 This implies that critical factors governing epileptogenesis should be identified upon triggering a convulsant episode in naive animals and these factors might be targeted to develop antiepileptic strategies since epilepsy can be pharmacologically controlled.
As mentioned before, the main brake of excitatory transmission and neuronal excitability, A1Rs, have their density decreased in the epileptic brain. In contrast, increased neuronal activity and seizure-like firing patterns trigger a sustained augmentation of the release of synaptic ATP45,167–170 and of the density and activity of ecto-5′-nucleotidase (ecto-5′-NT).45,110,171,172 Thereafter, ATP and ecto-5′-NT are responsible for ATP-derived adenosine formation and for the selective activation of synaptic A2AR.154,173,174 The A2ARs are the main bolsterer of excitatory transmission and neuronal excitability and have the expression increased in epilepsy, being a critical player in linking increased excitability with neurodegeneration.104 However, it is not yet clear if any of these changes is causative of epilepsy or if they are an adaptive consequence of epilepsy.
Aging—Altered Adenosine Modulation with Increased Incidence of Epilepsy
Epidemiological evidences show that the incidence of seizures and epilepsy is dramatically increased in persons older than 60 years. This may be due to a higher risk of older people to suffer a stroke or a head trauma as a result of a fall, leading to the development of cryptogenic or poststroke, and posttraumatic epilepsy.175
Another hypothesis argues that the prevalence of higher rates of epilepsy in elderly individuals may also be linked to alterations of the adenosine modulation system.176–178 In addition to the alterations of excitatory and inhibitory transmission systems179 as well as numerous neuromodulation systems,180 there is a modification of the adenosine modulation system upon aging that matches the alterations found in epileptic animal models and patients. In fact, there is a decrease of the extracellular levels of adenosine with aging.168,181 which is paralleled by a decreased density and function of inhibitory A1R in aged rodents180,182–186 and humans.187,188 In addition, in aged brain preparations, there is an increased release of ATP from recruited synapses168,189 with an increased density and activity of ecto-5′-NT168,190,191 and an increased density and function of facilitatory A2AR in animal models of aging180,192–195 and in humans.153
In particular, the overfunction of A2AR is deleterious for brain function196–198 and was recently proposed to be a causative factor of an imbalance of excitatory transmission by generating a hippocampal long-term depression to long-term potentiation (LTD-to-LTP) shift accompanied by increased NMDA receptor gating and enhanced Ca2+ influx dependent of mGluR5153 precipitating synaptic alterations that increase frailty of neuronal networks199 and accelerating neuronal dysfunction in animal models of brain diseases.200 This striking parallel supports the contention that a synaptic dysfunction of the adenosine modulation system might be a triggering factor for the increased susceptibility to epilepsy in the elderly.
Sudden Unexpected Death in Epilepsy
SUDEP is defined as “sudden, unexpected, witnessed or unwitnessed, non-traumatic and non-drowning death”201 and is a significant comorbidity for persons with epilepsy. In most witnessed or recorded cases, sudden deaths were immediately preceded by a seizure.202 SUDEP can affect individuals of any age, but is most common in young adults (aged 20–45 years) with an incidence of 1.2 per 1000 person-years.203
The biggest risk factor for SUDEP are generalized tonic-clonic seizures,203 although other risk factors have been discussed, such as cardiac problems and respiratory arrest. Accordingly, respiratory problems were observed in most cases of SUDEP.27,204 Indeed, epileptic seizures can induce significant hypoventilation.205,206 However, apnea and O2 desaturation are more common than previously realized after generalized partial or convulsant seizures,26 and recent evidence supports a significant role of apnea and hypoventilation in SUDEP.207
Respiration is controlled mainly by two neuronal groups located at the brainstem: (1) the parafacial respiratory group (pFRG) and (2) the pre-Bötzinger complex (inspiratory pacemaker population).208 The brainstem also contains nuclei responsible by other vital functions, such as the vagal nuclei, which significantly participates in the central control of cardiac activity.209 Molecularly, the control of respiration depends on the availability of a number of neuroactive substances, such as serotonin (5-HT), adenosine, and endogenous opioid peptides, that are released during seizures and whose concentrations are altered during the phase of postictal depression.26
Experimental evidences show that mice that had 5-HT2c receptor genetically deleted presented spontaneous generalized seizures that developed to a tonic extension phase and rapidly died.210 In addition, according to the “adenosine hypothesis of SUDEP,” seizure-induced adenosine release with impaired metabolic clearance by astrocytes, specifically in the brainstem may trigger lethal apnea or cardiac arrest. Accordingly, AR antagonists, such as caffeine or theophylline might prevent SUDEP.211 Therefore, serotonin and ARs, or other components of those pathways, may be promising targets for pharmacological SUDEP prevention therapy.26,27,212
Adenosine Augmentation Therapies for Epilepsy
Pharmacological approaches
Although seizure activity causes a robust elevation of adenosine levels in afflicted brain regions,213,214 the global augmentation of extracellular adenosine still emerges as a promising strategy to limit the spreading of seizure activity to still naive brain areas reviewed in.111 ADK inhibition is an ideal strategy for adenosine augmentation for several reasons. First, ADK is the major metabolizing enzyme of adenosine.78 In tissue studies, the Km for adenosine is 1–2 orders of magnitude lower than that of adenosine deaminase (ADA). Thereby, a minor change in ADK activity can result in a major change in adenosine concentration.78,215 In agreement with this, ADK inhibition, but not ADA inhibition, increased endogenous adenosine and neuronal inhibition in hippocampal slices216 and supressed bicuculine-induced seizures in rats.217
Furthermore, there is a pathological upregulation of ADK expression at an astrocyte enriched seizure focus in the epileptic brain in animal models and in tissue resected from humans with temporal lobe epilepsy.157 Consequently, pharmacological inhibition of ADK with 5-iodotubercidin was able to suppress seizures in the KA mouse model of pharmacoresistant epilepsy.218 Past drug discovery efforts have focused on finding a clinically useful ADK inhibitor219,220 and both effective nonnucleoside and nucleoside inhibitors have been identified.221–224 Although highly efficient in suppressing seizures, even those resistant to common ASDs,14 the clinical use of ADK inhibitors has been hampered by cardiovascular and sedative side effects of systemic adenosine augmentation.78,224
Cell-based adenosine delivery
To avoid side effects of systemic adenosine augmentation, local adenosine delivery approaches have been developed with the goal to increase adenosine concentrations locally in the brain in the vicinity of a seizure producing focus.225
In a pioneering proof of principle study, it was shown that encapsulated cell grafts of baby hamster kidney fibroblasts engineered to release adenosine based on the disruption of ADK expression, effectively suppressed seizures after implantation into the brain ventricles of rats submitted to electrical kindling.226 This strategy of local adenosine augmentation demonstrated that the focal paracrine release of adenosine was not only sufficient to suppress seizures227 but also to avoid sedative side effects associated with systemic ADK inhibition or A1R activation.228
Cell-based adenosine release for seizure suppression in epilepsy was further refined by engineering mouse embryonic stem cells to lack both alleles of the Adk gene. Those Adk−/− embryonic stem cells were differentiated into adenosine-releasing neural precursor (NP) cells and transplanted into the infrahippocampal cleft of rats 1 week before the onset of hippocampal electrical kindling.229 The Adk−/− NP cell recipient rats showed reduction of epileptogenesis induced by electrical kindling, and furthermore, 48 hours after kindling stimulations, these rats did not develop generalized seizures, suggesting for the first time a novel antiepileptogenic effect of adenosine.229
In a different study, the same Adk−/− NP cells were transplanted into the infrahippocampal cleft of mice 24 hours after receiving an intra-amygdaloid injection of KA. Mice, which received the implant of Adk−/− NP cells did not develop any spontaneous seizures, and when analyzed after 3 weeks of the KA injection, it was seen that astrogliosis was significantly reduced and ADK expression levels were close to normal when compared to control or sham animals.230 These latter two studies for the first time suggested a hitherto unknown antiepileptogenic potential of adenosine releasing cellular brain implants.
In an attempt to generate a cell line compatible for future clinical applications, Ren et al. used a model of ADK ablation in human mesenchymal stem cells based on the lentiviral expression of a microRNA (miRNA) directed against ADK, achieving around 80% reduction of ADK expression in these cells.231,232 Transplantation of these cells into mice before the injection of KA significantly reduced the acute brain injury and seizures in these mice.232
Gene therapy
Gene therapy focuses on the attenuation of the endogenous ADK gene. The logic is as follows: by reducing the activity of ADK, which is overexpressed in an epileptic brain, adenosine levels will increase and ultimately exert their anticonvulsant effects. Gene therapy is a logical avenue of treatment as it corrects the pathology, being the overexpression of ADK, at the genetic level rather than just suppressing symptomology, which is typically the case in pharmacological treatments.
Promising results were seen in a study that utilized an adeno-associated virus (AAV) vector that expressed ADK-complementary DNA (cDNA) in antisense orientation under the control of gfaABC1D promotor was injected unilaterally into the CA3 region of the hippocampus to knock down ADK in astrocytes in adult male Adk-tg mice.233 The mice showed a significant suppression of local seizure activity ipsilateral to the injection side with 0.6 ± 0.6 seizures/h compared to 5.8 ± 0.5 seizures/h in the noninjection contralateral side.233 These results show that antisense-mediated knockdown of ADK in astrocytes is an effective strategy for the suppression of seizure activity.233
Dietary interventions
An additional strategy to augment adenosine levels is through a ketogenic diet (KD). A KD, which consists of a high-fat, low-carbohydrate intake, produces an increase in ATP levels within the brain, ultimately increasing adenosine concentration as one of its beneficial mechanisms.234
When carbohydrate intake is limited, as in a KD, blood glucose levels decline causing the liver to convert fatty acids to ketone bodies, which enable a more efficient ATP production. Since a small increase in ATP translates into a disproportional larger increase in adenosine concentration reviewed in,235 the KD becomes an attractive method of augmenting adenosine as a therapy for epilepsy. Accordingly, an in vitro study showed that a mimic of KD afforded a hyperpolarizing effect in hippocampal CA3 pyramidal cells (a cell type and location involved in seizures), through a combined increased activation of A1R and opening of ATP-sensitive potassium channels in response to reduced glucose levels.158
Through the augmentation of adenosine, KD offers not only anticonvulsant effects, but antiepileptogenic effects as well. In fact, mice fed a KD were significantly less susceptible to chemical kindling epileptogenesis than mice fed a control diet, indicating that the KD delayed the development of epilepsy.236
To determine the longevity of the antiepileptogenic effects of the KD, the KD-fed mice and PTZ-kindled mice were administered a single injection of glucose, which is known to terminate acute impacts of the KD and model a diet reversal. After 4 days, the mean Racine's scale seizure score of the two KD groups were alike and significantly lower than the control group, demonstrating a lasting antiepileptogenic impact.236
In a long-term study performed in the pilocarpine model of epilepsy in rats, it was shown that in KD-treated rats a lasting reduction of spontaneous recurrent seizure activity was achieved even 6–8 weeks after diet reversal, whereas control diet exposed rats experienced robust prolonged seizures with varying patterns and intensity.236 The KD-fed rats had reduced levels of DNA methylation compared with the control diet fed rats even after diet reversal, which supports that the reduction in seizures is linked to an antiepileptogenic disease-modifying mechanism induced by epigenetic changes.236 This study warrants further research of the antiepileptogenic effects of a KD, specifically in controlled clinical studies. Indeed, the few available clinical studies indicate that a KD has significant disease-modifying impacts, leaving some patients seizure-free even after discontinuation of the KD.237,238
Notably, the KD has been most successful in mediating seizures in pediatric epilepsy. In a randomized controlled clinical trial, the impacts of a 3-month long trial of a KD was studied in children between the ages of 2 and 16 years, diagnosed with either generalized or focal epilepsy who had at least a seizure per day.239 The results showed that 38% of patients displayed a >50% reduction, with some reaching as high as >90% reduction.239
Another pediatric study, which focused on children with pharmacoresistant epilepsy, showed after introducing a modified Atkins diet (a less restrictive variation of the KD), 30% of children experienced 90% seizure reduction and 52% of children experienced 50% seizure reduction after 3 months.238,240 In an adult model, 32% of adult patients with pharmacoresistant epilepsy showed >50% seizure reduction using a KD.237,238
While the therapeutic impact of adenosine augmentation is the focus of this section, it is important to note that the KD offers seizure control through other mechanisms as well. There is evidence that the presence of ketone bodies, free fatty acids, and overall glucose restriction has anticonvulsant properties and can lead to seizure protection.241 In addition, the KD offers a variety of other physiological benefits, especially for adult patients, including reduction of comorbidities, enhancing adenosine's role as a neuroprotective molecule, aiding in sleep, and a significant role in pain reduction.234
Strategies for Epilepsy Prevention
Epilepsy prevention is the Holy Grail of epilepsy research and therapy development. Aproximately 15% of all diagnosed epilepsies are caused by an acute acquired brain insult, such as traumatic brain injury, stroke, or encephalitis.10,242 The latent period following a potentially epilepsy triggering event presents a unique opportunity for preventive interventions. Several lines of evidence show that the transient therapeutic augmentation of adenosine has lasting antiepileptogenic and disease-modifying effects.
The first indication that adenosine has antiepileptogenic effects was provided in the cell transplantation studies discussed above. Adenosine-releasing hippocampal cell grafts suppressed hippocampal electrical kindling development in rats229 and epilepsy development in the mouse intraamygdaloid KA model.225
To understand the mechanisms behind those antiepileptogenic effects, the biomaterial silk was engineered to release a defined dose of adenosine for a restricted time span of 10 days.243 As expected, adenosine releasing silk implanted into the lateral brain ventricles of rats suppressed electrically induced seizures in rats submitted to electrical kindling.243
The same implants were introduced into the brain ventricles of rats 9 weeks after the systemic injection of KA. At this timepoint, all rats had experienced at least four to six spontaneous convulsant seizures, had overexpressed ADK, and increased DNA methylation levels in the hippocampus. The acute release of adenosine (during 10 days) suppressed DNA methyltransferase activity and reduced DNA methylation levels to baseline levels seen in control animals. Importantly, normalization of DNA methylation status was maintained even after cessation of the adenosine release. While seizure rate (seizures per week) and intensity (duration) continuously increased in all sham or control silk-treated animals, the development of epilepsy was completely stalled for at least 3 months in animals that received the adenosine-releasing brain implants.
In parallel to the suppression of seziure development further, sprouting of mossy fibers was blocked. In line with suppression of epilepsy progression, DNA methylation levels were found to be normalized. This study identified a novel adenosine-dependent epigenetic mechanism responsible for the prevention of epilepsy progression.16
Adenosine-releasing silk might not be the most practical approach for clinical translation. Therefore, ADK inhibitors might provide a better alternative for epilepsy prevention. A recent study demonstrated that a transient dose of the ADK inhibitor 5-iodotubercidin given systemically from 3 to 8 days after the intrahippocampal injection of KA prevented epilepsy development in 50% of all of tested mice.15 This is an important finding, because the transient use of an ADK inhibitor would be acceptable in light of side effects associated with chronic inhibition of ADK.
Apart from these strategies based on the augmentation of adenosine, there is also recent evidence showing that the pharmacological blockade of A2AR can also interupt the casacde of events converting abnormal brain activity into neurodegeneration,111 a key process in the development of circuit remodeling and establishment of epilepsy.165 In fact, a recent study identified a key role of A2AR linking increased excitability with neurodegeneration in a kainate model of temporal lobe epilepsy,104 in accordance with the ability of A2AR blockade to dampen the proconvulsant epileptogenic changes during evolution of kindling.102–104
This neuroprotective role of A2AR antagonists was also documented in animal models for long-term deleterious consequences of early-life convulsions105 and also in clinical findings with the report that genetic variation in human adenosine A2AR gene (ADORA2A) associated with increased expression of A2AR and higher levels of cAMP production is a predisposing factor for childhood encephalopathy following severe febrile seizures.107 It is worth mentioning that the interest of considering the use of A2AR antagonist as coadjuvants to manage the evolution of epilepsy is particularly attractive in view of the ability of these A2AR antagonists to control the most common comorbibities with epilepsy, namely memory impairment,244–248 mood dysfunction,249–251 migrane,252,253 and sleep disorders.254,255
Conclusions and Outlook
There is now solid evidence that the adenosine system is dysregulated on several different levels in epilepsy. In particular, maladaptive changes in adenosine metabolism can explain the generation of seizures (ictogenesis), as well as comorbidities of epilepsy including SUDEP, whereas A2AR overfunction seems paramount for the emergence of neurodegenerative consequences of seizure activity and circuit remodeling to implement an epileptic condition. Consequently, adenosine-based therapies are based on a solid rationale to directly affect multiple mechanisms involved in the expression of epilepsy.
Novel findings document the therapeutic potential of A2AR antagonists as well as AR-independent roles of adenosine as an epigenetic regulator, which form the basis for the development of antiepileptogenic therapies. The development of novel compounds acting on the nuclear form of ADK might capitalize on the epigenetic effects of adenosine without causing AR-mediated effects that still remain to be tested in conjunction with A2AR antagonists to maximize the therapeutic possibilities of targeting the adenosione system to manage epilepsy. Future work needs to determine whether those strategies are safe and also effective in clinically realistic models of posttraumatic epilepsy.
Author Disclosure Statement
No competing financial interests exist.
Funding Information
The authors acknowledge grant support from the National Institutes of Health to D.B. (NINDS: NS103740, NS065957); support to A.M.S. by LISBOA-01-0145-FEDER-007391, project cofunded by Fundo Europeu Para o Desenvolvimento Regional through POR Lisboa 2020 (Programa Operacional Regional de Lisboa) from PORTUGAL 2020 and Fundação para a Ciência e Tecnologia (FCT), by an FCT project (PTDC/MED-FAR/30933/2017) and by Twinning action (SynaNet) from the EU H2020 programme (project number: 692340); and support to R.A.C. and A.R.T. by Fundacion LaCaixa (LCF/PR/HP17/52190001), Centro 2020 (CENTRO-01-0145-FEDER-000008: BrainHealth 2020 and CENTRO-01-0246-FEDER-000010) and FCT (POCI-01-0145-FEDER-03127).
References
- 1. White HS, Smith MD, Wilcox KS. Mechanisms of action of antiepileptic drugs. Int Rev Neurobiol. 2007;81:85–110 [DOI] [PubMed] [Google Scholar]
- 2. Pitkanen A, Nehlig A, Brooks-Kayal AR, et al. . Issues related to development of antiepileptogenic therapies. Epilepsia. 2013;54(Suppl 4):35–43 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3. Loscher W, Schmidt D. Modern antiepileptic drug development has failed to deliver: ways out of the current dilemma. Epilepsia. 2011;52:657–678 [DOI] [PubMed] [Google Scholar]
- 4. Boison D. The biochemistry and epigenetics of epilepsy: focus on adenosine and glycine. Front Mol Neurosci. 2016;9:26. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5. Boison D, Steinhauser C. Epilepsy and astrocyte energy metabolism. Glia. 2018;66:1235–1243 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6. Devinsky O, Vezzani A, Najjar S, et al. . Glia and epilepsy: excitability and inflammation. Trends Neurosci. 2013;36:174–184 [DOI] [PubMed] [Google Scholar]
- 7. Vezzani A, Aronica E, Mazarati A, et al. . Epilepsy and brain inflammation. Exp Neurol. 2013;244:11–21 [DOI] [PubMed] [Google Scholar]
- 8. Boison D. Astrogliosis and adenosine kinase: a glial basis of epilepsy. Future Neurol. 2008;3:221–224 [Google Scholar]
- 9. Steinhauser C, Grunnet M, Carmignoto G. Crucial role of astrocytes in temporal lobe epilepsy. Neuroscience. 2016;323:157–169 [DOI] [PubMed] [Google Scholar]
- 10. Klein P, Dingledine R, Aronica E, et al. . Commonalities in epileptogenic processes from different acute brain insults: do they translate? Epilepsia. 2018;59:37–66 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11. Welzel L, Twele F, Schidlitzki A, et al. . Network pharmacology for antiepileptogenesis: tolerability and neuroprotective effects of novel multitargeted combination treatments in nonepileptic vs. post-status epilepticus mice. Epilepsy Res. 2019;151:48–66 [DOI] [PubMed] [Google Scholar]
- 12. Loscher W. The holy grail of epilepsy prevention: preclinical approaches to antiepileptogenic treatments. Neuropharmacology. In Press [DOI] [PubMed] [Google Scholar]
- 13. Cunha RA. Signaling by adenosine receptors-Homeostatic or allostatic control? PLoS Biol. 2019;17:e3000213. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14. Gouder N, Fritschy JM, Boison D. Seizure suppression by adenosine A1 receptor activation in a mouse model of pharmacoresistant epilepsy. Epilepsia. 2003;44:877–885 [DOI] [PubMed] [Google Scholar]
- 15. Sandau US, Yahya M, Bigej R, et al. . Transient use of a systemic adenosine kinase inhibitor attenuates epilepsy development in mice. Epilepsia. 2019;60:615–625 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16. Williams-Karnesky RL, Sandau US, Lusardi TA, et al. . Epigenetic changes induced by adenosine augmentation therapy prevent epileptogenesis. J Clin Invest. 2013;123:3552–3563 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17. Turski L, Ikonomidou C, Turski WA, et al. . Review: cholinergic mechanisms and epileptogenesis. The seizures induced by pilocarpine: a novel experimental model of intractable epilepsy. Synapse. 1989;3:154–171 [DOI] [PubMed] [Google Scholar]
- 18. Fisher RS, Acevedo C, Arzimanoglou A, et al. . ILAE official report: a practical clinical definition of epilepsy. Epilepsia. 2014;55:475–482 [DOI] [PubMed] [Google Scholar]
- 19. Falco-Walter JJ, Scheffer IE, Fisher RS. The new definition and classification of seizures and epilepsy. Epilepsy Res. 2018;139:73–79 [DOI] [PubMed] [Google Scholar]
- 20. Blauwblomme T, Jiruska P, Huberfeld G. Mechanisms of ictogenesis. Int Rev Neurobiol. 2014;114:155–185 [DOI] [PubMed] [Google Scholar]
- 21. Cook MJ, O'Brien TJ, Berkovic SF, et al. . Prediction of seizure likelihood with a long-term, implanted seizure advisory system in patients with drug-resistant epilepsy: a first-in-man study. Lancet Neurol. 2013;12:563–571 [DOI] [PubMed] [Google Scholar]
- 22. Irmen F, Wehner T, Lemieux L. Do reflex seizures and spontaneous seizures form a continuum? - triggering factors and possible common mechanisms. Seizure. 2015;25:72–79 [DOI] [PubMed] [Google Scholar]
- 23. Lowrie M, Bessant C, Harvey RJ, et al. . Audiogenic reflex seizures in cats. J Feline Med Surg. 2016;18:328–336 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24. Gelisse P, Genton P, Coubes P, et al. . Can emotional stress trigger the onset of epilepsy? Epilepsy Behav. 2015;48:15–20 [DOI] [PubMed] [Google Scholar]
- 25. Klitgaard H, Pitkanen A. Antiepileptogenesis, neuroprotection, and disease modification in the treatment of epilepsy: focus on levetiracetam. Epileptic Disord. 2003;5(Suppl 1):S9–S16 [PubMed] [Google Scholar]
- 26. Richerson GB, Boison D, Faingold CL, et al. . From unwitnessed fatality to witnessed rescue: pharmacologic intervention in sudden unexpected death in epilepsy. Epilepsia. 2016;57(Suppl 1):35–45 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27. Boison D. A breather for SUDEP. Epilepsy Curr. 2012;12:111–112 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28. Hauser WA. Sudden unexpected death in patients with epilepsy: issues for further study. Epilepsia. 1997;38:S26–S29 [DOI] [PubMed] [Google Scholar]
- 29. Hughes JR. A review of sudden unexpected death in epilepsy: prediction of patients at risk. Epilepsy Behav. 2009;14:280–287 [DOI] [PubMed] [Google Scholar]
- 30. Sloviter RS, Bumanglag AV. Defining “epileptogenesis” and identifying “antiepileptogenic targets” in animal models of acquired temporal lobe epilepsy is not as simple as it might seem. Neuropharmacology. 2013;69:3–15 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31. Pitkanen A, Lukasiuk K, Dudek FE, et al. . Epileptogenesis. Cold Spring Harb Perspect Med. 2015;5:a022822. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32. Clossen BL, Reddy DS. Novel therapeutic approaches for disease-modification of epileptogenesis for curing epilepsy. Biochim Biophys Acta Mol Basis Dis. 2017;1863:1519–1538 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33. Pohlen MS, Jin J, Tobias RS, et al. . Pharmacoresistance with newer anti-epileptic drugs in mesial temporal lobe epilepsy with hippocampal sclerosis. Epilepsy Res. 2017;137:56–60 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34. Venceslas D, Corinne R. A mesiotemporal lobe epilepsy mouse model. Neurochem Res. 2017;42:1919–1925 [DOI] [PubMed] [Google Scholar]
- 35. Babb TL, Brown WJ, Pretorius J, et al. . Temporal lobe volumetric cell densities in temporal lobe epilepsy. Epilepsia. 1984;25:729–740 [DOI] [PubMed] [Google Scholar]
- 36. Thijs RD, Surges R, O'Brien TJ, et al. . Epilepsy in adults. Lancet. 2019;393:689–701 [DOI] [PubMed] [Google Scholar]
- 37. Houser CR. Granule cell dispersion in the dentate gyrus of humans with temporal lobe epilepsy. Brain Res. 1990;535:195–204 [DOI] [PubMed] [Google Scholar]
- 38. Mathern GW, Leite JP, Babb TL, et al. . Aberrant hippocampal mossy fiber sprouting correlates with greater NMDAR2 receptor staining. Neuroreport. 1996;7:1029–1035 [DOI] [PubMed] [Google Scholar]
- 39. Sloviter RS, Sollas AL, Barbaro NM, et al. . Calcium-binding protein (Calbindin-D28K) and parvalbumin immunocytochemistry in the normal and epileptic human hippocampus. J Comp Neurol. 1991;308:381–396 [DOI] [PubMed] [Google Scholar]
- 40. Dudek FE, Staley KJ. The time course of acquired epilepsy: implications for therapeutic intervention to suppress epileptogenesis. Neurosci Lett. 2011;497:240–246 [DOI] [PubMed] [Google Scholar]
- 41. Phillis JW, Kostopoulos GK, Limacher JJ. Depression of corticospinal cells by various purines and pyrimidines. Can J Physiol Pharmacol. 1974;52:1226–1229 [DOI] [PubMed] [Google Scholar]
- 42. Fredholm BB, Ijzerman A, Jacobson KA, et al. . International Union of Basic and Clinical Pharmacology. LXXXI. Nomenclature and classification of adenosine receptors—an update. Pharmacol Rev. 2011;63:1–34 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43. Sheth S, Brito R, Mukherjea D, et al. . Adenosine receptors: expression, function and regulation. Int J Mol Sci. 2014;15:2024–2052 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44. Tetzlaff W, Schubert P, Kreutzberg GW. Synaptic and extrasynaptic localization of adenosine binding sites in the rat hippocampus. Neuroscience. 1987;21:869–875 [DOI] [PubMed] [Google Scholar]
- 45. Rebola N, Coelho JE, Costenla AR, et al. . Decrease of adenosine A1 receptor density and of adenosine neuromodulation in the hippocampus of kindled rats. Eur J Neurosci. 2003;18:820–828 [DOI] [PubMed] [Google Scholar]
- 46. Rebola N, Porciúncula LO, Lopes LV, et al. . Long-term effect of convulsive behavior on the density of adenosine A1 and A 2A receptors in the rat cerebral cortex. Epilepsia. 2005;46(Suppl 5):159–165 [DOI] [PubMed] [Google Scholar]
- 47. Biber K, Klotz KN, Berger M, et al. . Adenosine A1 receptor-mediated activation of phospholipase C in cultured astrocytes depends on the level of receptor expression. J Neurosci. 1997;17:4956–4964 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48. Othman T, Yan H, Rivkees SA. Oligodendrocytes express functional A1 adenosine receptors that stimulate cellular migration. Glia. 2003;44:166–172 [DOI] [PubMed] [Google Scholar]
- 49. Gebicke-Haerter PJ, Christoffel F, Timmer J, et al. . Both adenosine A1- and A2-receptors are required to stimulate microglial proliferation. Neurochem Int. 1996;29:37–42 [PubMed] [Google Scholar]
- 50. de Lera Ruiz M, Lim YH, Zheng J. Adenosine A2A receptor as a drug discovery target. J Med Chem. 2014;57:3623–3650 [DOI] [PubMed] [Google Scholar]
- 51. Cunha RA, Johansson B, van der Ploeg I, et al. . Evidence for functionally important adenosine A2a receptors in the rat hippocampus. Brain Res. 1994;649:208–216 [DOI] [PubMed] [Google Scholar]
- 52. Rebola N, Canas PM, Oliveira CR, et al. . Different synaptic and subsynaptic localization of adenosine A2A receptors in the hippocampus and striatum of the rat. Neuroscience. 2005;132:893–903 [DOI] [PubMed] [Google Scholar]
- 53. Orr AG, Orr AL, Li XJ, et al. . Adenosine A(2A) receptor mediates microglial process retraction. Nat Neurosci. 2009;12:872–878 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54. Rebola N, Simões AP, Canas PM, et al. . Adenosine A2A receptors control neuroinflammation and consequent hippocampal neuronal dysfunction. J Neurochem. 2011;117:100–111 [DOI] [PubMed] [Google Scholar]
- 55. Orr AG, Hsiao EC, Wang MM, et al. . Astrocytic adenosine receptor A2A and Gs-coupled signaling regulate memory. Nat Neurosci. 2015;18:423–434 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56. Dunwiddie TV. Endogenously released adenosine regulates excitability in the in vitro hippocampus. Epilepsia. 1980;21:541–548 [DOI] [PubMed] [Google Scholar]
- 57. Dunwiddie TV, Worth T. Sedative and anticonvulsant effects of adenosine analogs in mouse and rat. J Pharmacol Exp Ther. 1982;220:70–76 [PubMed] [Google Scholar]
- 58. Rombo DM, Ribeiro JA, Sebastião AM. Role of adenosine receptors in epileptic seizures. In: The Adenosine Receptors. The Receptors Borea P.A., Varani P.A., Gessi K., et al. (Eds). Cham: Humana Press; 2018: pp. 309–350 [Google Scholar]
- 59. Rebola N, Pinheiro PC, Oliveira CR, et al. . Subcellular localization of adenosine A(1) receptors in nerve terminals and synapses of the rat hippocampus. Brain Res. 2003;987:49–58 [DOI] [PubMed] [Google Scholar]
- 60. MacDonald RL, Skerritt JH, Werz MA. Adenosine agonists reduce voltage-dependent calcium conductance of mouse sensory neurones in cell culture. J Physiol. 1986;370:75–90 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61. Schubert P, Heinemann U, Kolb R. Differential effect of adenosine on pre- and postsynaptic calcium fluxes. Brain Res. 1986;376:382–386 [DOI] [PubMed] [Google Scholar]
- 62. Wu LG, Saggau P. Adenosine inhibits evoked synaptic transmission primarily by reducing presynaptic calcium influx in area CA1 of hippocampus. Neuron. 1994;12:1139–1148 [DOI] [PubMed] [Google Scholar]
- 63. Scanziani M, Capogna M, Gahwiler BH, et al. . Presynaptic inhibition of miniature excitatory synaptic currents by baclofen and adenosine in the hippocampus. Neuron. 1992;9:919–927 [DOI] [PubMed] [Google Scholar]
- 64. Scholz KP, Miller RJ. Inhibition of quantal transmitter release in the absence of calcium influx by a G protein-linked adenosine receptor at hippocampal synapses. Neuron. 1992;8:1139–1150 [DOI] [PubMed] [Google Scholar]
- 65. de Mendonça A, Sebastião AM, Ribeiro JA. Inhibition of NMDA receptor-mediated currents in isolated rat hippocampal neurones by adenosine A1 receptor activation. Neuroreport. 1995;6:1097–1100 [DOI] [PubMed] [Google Scholar]
- 66. Kharazia VN, Prince DA. Changes of alpha-amino-3-hydroxy-5-methyl-4-isoxazole-propionate receptors in layer V of epileptogenic, chronically isolated rat neocortex. Neuroscience. 2001;102:23–34 [DOI] [PubMed] [Google Scholar]
- 67. Kohl BK, Dannhardt G. The NMDA receptor complex: a promising target for novel antiepileptic strategies. Curr Med Chem. 2001;8:1275–1289 [DOI] [PubMed] [Google Scholar]
- 68. Rice AC, DeLorenzo RJ. NMDA receptor activation during status epilepticus is required for the development of epilepsy. Brain Res. 1998;782:240–247 [DOI] [PubMed] [Google Scholar]
- 69. Zhang JM, Wang HK, Ye CQ, et al. . ATP released by astrocytes mediates glutamatergic activity-dependent heterosynaptic suppression. Neuron. 2003;40:971–982 [DOI] [PubMed] [Google Scholar]
- 70. Alzheimer C, Sutor B, ten Bruggencate G. Disinhibition of hippocampal CA3 neurons induced by suppression of an adenosine A1 receptor-mediated inhibitory tonus: pre- and postsynaptic components. Neuroscience. 1993;57:565–575 [DOI] [PubMed] [Google Scholar]
- 71. Moschovos C, Kostopoulos G, Papatheodoropoulos C. Endogenous adenosine induces NMDA receptor-independent persistent epileptiform discharges in dorsal and ventral hippocampus via activation of A2 receptors. Epilepsy Res. 2012;100:157–167 [DOI] [PubMed] [Google Scholar]
- 72. Thummler S, Dunwiddie TV. Adenosine receptor antagonists induce persistent bursting in the rat hippocampal CA3 region via an NMDA receptor-dependent mechanism. J Neurophysiol. 2000;83:1787–1795 [DOI] [PubMed] [Google Scholar]
- 73. Rombo DM, Ribeiro JA, Sebastião AM. Hippocampal GABAergic transmission: a new target for adenosine control of excitability. J Neurochem. 2016;139:1056–1070 [DOI] [PubMed] [Google Scholar]
- 74. Rombo DM, Dias RB, Duarte ST, et al. . Adenosine A1 receptor suppresses tonic GABAA receptor currents in hippocampal pyramidal cells and in a defined subpopulation of interneurons. Cereb Cortex. 2016;26:1081–1095 [DOI] [PubMed] [Google Scholar]
- 75. Lambert NA, Teyler TJ. Adenosine depresses excitatory but not fast inhibitory synaptic transmission in area CA1 of the rat hippocampus. Neurosci Lett. 1991;122:50–52 [DOI] [PubMed] [Google Scholar]
- 76. Prince DA, Stevens CF. Adenosine decreases neurotransmitter release at central synapses. Proc Natl Acad Sci U S A. 1992;89:8586–8590 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77. Yoon KW, Rothman SM. Adenosine inhibits excitatory but not inhibitory synaptic transmission in the hippocampus. J Neurosci. 1991;11:1375–1380 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78. Boison D. Adenosine kinase: exploitation for therapeutic gain. Pharmacol Rev. 2013;65:906–943 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79. Etherington LA, Patterson GE, Meechan L, et al. . Astrocytic adenosine kinase regulates basal synaptic adenosine levels and seizure activity but not activity-dependent adenosine release in the hippocampus. Neuropharmacology. 2009;56:429–437 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80. Boison D. Adenosinergic signaling in epilepsy. Neuropharmacology. 2016;104:131–139 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81. Ekonomou A, Sperk G, Kostopoulos G, et al. . Reduction of A1 adenosine receptors in rat hippocampus after kainic acid-induced limbic seizures. Neurosci Lett. 2000;284:49–52 [DOI] [PubMed] [Google Scholar]
- 82. Hamil NE, Cock HR, Walker MC. Acute down-regulation of adenosine A(1) receptor activity in status epilepticus. Epilepsia. 2012;53:177–188 [DOI] [PubMed] [Google Scholar]
- 83. Ochiishi T, Takita M, Ikemoto M, et al. . Immunohistochemical analysis on the role of adenosine A1 receptors in epilepsy. Neuroreport. 1999;10:3535–3541 [DOI] [PubMed] [Google Scholar]
- 84. Glass M, Faull RL, Bullock JY, et al. . Loss of A1 adenosine receptors in human temporal lobe epilepsy. Brain Res. 1996;710:56–68 [DOI] [PubMed] [Google Scholar]
- 85. Coelho JE, Rebola N, Fragata I, et al. . Hypoxia-induced desensitization and internalization of adenosine A1 receptors in the rat hippocampus. Neuroscience. 2006;138:1195–1203 [DOI] [PubMed] [Google Scholar]
- 86. Adami M, Bertorelli R, Ferri N, et al. . Effects of repeated administration of selective adenosine A1 and A2A receptor agonists on pentylenetetrazole-induced convulsions in the rat. Eur J Pharmacol. 1995;294:383–389 [DOI] [PubMed] [Google Scholar]
- 87. Stella L, Berrino L, Maione S, et al. . Cardiovascular effects of adenosine and its analogs in anaesthetized rats. Life Sci. 1993;53:755–763 [DOI] [PubMed] [Google Scholar]
- 88. Schindler CW, Karcz-Kubicha M, Thorndike EB, et al. . Role of central and peripheral adenosine receptors in the cardiovascular responses to intraperitoneal injections of adenosine A1 and A2A subtype receptor agonists. Br J Pharmacol. 2005;144:642–650 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89. Shen HY, Coelho JE, Ohtsuka N, et al. . A critical role of the adenosine A2A receptor in extrastriatal neurons in modulating psychomotor activity as revealed by opposite phenotypes of striatum and forebrain A2A receptor knock-outs. J Neurosci. 2008;28:2970–2975 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90. Shen HY, Canas PM, Garcia-Sanz P, et al. . Adenosine A(2)A receptors in striatal glutamatergic terminals and GABAergic neurons oppositely modulate psychostimulant action and DARPP-32 phosphorylation. PLoS One. 2013;8:e80902. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91. Jones PA, Smith RA, Stone TW. Protection against kainate-induced excitotoxicity by adenosine A2A receptor agonists and antagonists. Neuroscience. 1998;85:229–237 [DOI] [PubMed] [Google Scholar]
- 92. De Sarro G, De Sarro A, Di Paola ED, et al. . Effects of adenosine receptor agonists and antagonists on audiogenic seizure-sensible DBA/2 mice. Eur J Pharmacol. 1999;371:137–145 [DOI] [PubMed] [Google Scholar]
- 93. Vianna EP, Ferreira AT, Dona F, et al. . Modulation of seizures and synaptic plasticity by adenosinergic receptors in an experimental model of temporal lobe epilepsy induced by pilocarpine in rats. Epilepsia. 2005;46(Suppl 5):166–173 [DOI] [PubMed] [Google Scholar]
- 94. Yang M, Soohoo D, Soelaiman S, et al. . Characterization of the potency, selectivity, and pharmacokinetic profile for six adenosine A2A receptor antagonists. Naunyn Schmiedebergs Arch Pharmacol. 2007;375:133–144 [DOI] [PubMed] [Google Scholar]
- 95. Yu L, Shen HY, Coelho JE, et al. . Adenosine A2A receptor antagonists exert motor and neuroprotective effects by distinct cellular mechanisms. Ann Neurol. 2008;63:338–346 [DOI] [PubMed] [Google Scholar]
- 96. Zhang G, Franklin PH, Murray TF. Activation of adenosine A1 receptors underlies anticonvulsant effect of CGS21680. Eur J Pharmacol. 1994;255:239–243 [DOI] [PubMed] [Google Scholar]
- 97. El Yacoubi M, Ledent C, Parmentier M, et al. . Absence of the adenosine A(2A) receptor or its chronic blockade decrease ethanol withdrawal-induced seizures in mice. Neuropharmacology. 2001;40:424–432 [DOI] [PubMed] [Google Scholar]
- 98. El Yacoubi M, Ledent C, Parmentier M, et al. . 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. 2008;55:35–40 [DOI] [PubMed] [Google Scholar]
- 99. Hosseinmardi N, Mirnajafi-Zadeh J, Fathollahi Y, et al. . The role of adenosine A1 and A2A receptors of entorhinal cortex on piriform cortex kindled seizures in rats. Pharmacol Res. 2007;56:110–117 [DOI] [PubMed] [Google Scholar]
- 100. Zeraati M, Mirnajafi-Zadeh J, Fathollahi Y, et al. . Adenosine A1 and A2A receptors of hippocampal CA1 region have opposite effects on piriform cortex kindled seizures in rats. Seizure. 2006;15:41–48 [DOI] [PubMed] [Google Scholar]
- 101. Li X, Kang H, Liu X, et al. . Effect of adenosine A2A receptor antagonist ZM241385 on amygdala-kindled seizures and progression of amygdala kindling. J Huazhong Univ Sci Technolog Med Sci. 2012;32:257–264 [DOI] [PubMed] [Google Scholar]
- 102. El Yacoubi M, Ledent C, Parmentier M, et al. . Adenosine A2A receptor deficient mice are partially resistant to limbic seizures. Naunyn Schmiedebergs Arch Pharmacol. 2009;380:223–232 [DOI] [PubMed] [Google Scholar]
- 103. Lee HK, Choi SS, Han KJ, et al. . Roles of adenosine receptors in the regulation of kainic acid-induced neurotoxic responses in mice. Brain Res Mol Brain Res. 2004;125:76–85 [DOI] [PubMed] [Google Scholar]
- 104. Canas PM, Porciúncula LO, Simões AP, et al. . Neuronal adenosine A2A receptors are critical mediators of neurodegeneration triggered by convulsions. eNeuro. 2018;5:e0385-18 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105. Cognato GP, Agostinho PM, Hockemeyer J, et al. . Caffeine and an adenosine A(2A) receptor antagonist prevent memory impairment and synaptotoxicity in adult rats triggered by a convulsive episode in early life. J Neurochem. 2010;112:453–462 [DOI] [PubMed] [Google Scholar]
- 106. Silva CG, Métin C, Fazeli W, et al. . Adenosine receptor antagonists including caffeine alter fetal brain development in mice. Sci Transl Med. 2013;5:197ra104. [DOI] [PubMed] [Google Scholar]
- 107. Shinohara M, Saitoh M, Nishizawa D, et al. . ADORA2A polymorphism predisposes children to encephalopathy with febrile status epilepticus. Neurology. 2013;80:1571–1576 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108. Crespo M, Leon-Navarro DA, Martin M. Early-life hyperthermic seizures upregulate adenosine A2A receptors in the cortex and promote depressive-like behavior in adult rats. Epilepsy Behav. 2018;86:173–178 [DOI] [PubMed] [Google Scholar]
- 109. D'Alimonte I, D'Auro M, Citraro R, et al. . Altered distribution and function of A2A adenosine receptors in the brain of WAG/Rij rats with genetic absence epilepsy, before and after appearance of the disease. Eur J Neurosci. 2009;30:1023–1035 [DOI] [PubMed] [Google Scholar]
- 110. Barros-Barbosa AR, Ferreirinha F, Oliveira A, et al. . Adenosine A2A receptor and ecto-5′-nucleotidase/CD73 are upregulated in hippocampal astrocytes of human patients with mesial temporal lobe epilepsy (MTLE). Purinergic Signal. 2016;12:719–734 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111. Cunha RA. How does adenosine control neuronal dysfunction and neurodegeneration? J Neurochem. 2016;139:1019–1055 [DOI] [PubMed] [Google Scholar]
- 112. Avsar E, Empson RM. Adenosine acting via A1 receptors, controls the transition to status epilepticus-like behaviour in an in vitro model of epilepsy. Neuropharmacology. 2004;47:427–437 [DOI] [PubMed] [Google Scholar]
- 113. Fedele DE, Li T, Lan JQ, et al. . Adenosine A1 receptors are crucial in keeping an epileptic focus localized. Exp Neurol. 2006;200:184–190 [DOI] [PubMed] [Google Scholar]
- 114. Kochanek PM, Vagni VA, Janesko KL, et al. . Adenosine A1 receptor knockout mice develop lethal status epilepticus after experimental traumatic brain injury. J Cereb Blood Flow Metab. 2006;26:565–575 [DOI] [PubMed] [Google Scholar]
- 115. Stockwell J, Jakova E, Cayabyab FS. Adenosine A1 and A2A receptors in the brain: current research and their role in neurodegeneration. Molecules. 2017;22:676. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116. Fredholm BB, Chen JF, Cunha RA, et al. . Adenosine and brain function. Int Rev Neurobiol. 2005;63:191–270 [DOI] [PubMed] [Google Scholar]
- 117. Laudadio MA, Psarropoulou C. The A3 adenosine receptor agonist 2-Cl-IB-MECA facilitates epileptiform discharges in the CA3 area of immature rat hippocampal slices. Epilepsy Res. 2004;59:83–94 [DOI] [PubMed] [Google Scholar]
- 118. Roseti C, Martinello K, Fucile S, et al. . Adenosine receptor antagonists alter the stability of human epileptic GABAA receptors. Proc Natl Acad Sci U S A. 2008;105:15118–15123 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119. Von Lubitz DK, Carter MF, Deutsch SI, et al. . The effects of adenosine A3 receptor stimulation on seizures in mice. Eur J Pharmacol. 1995;275:23–29 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120. Etherington LA, Frenguelli BG. Endogenous adenosine modulates epileptiform activity in rat hippocampus in a receptor subtype-dependent manner. Eur J Neurosci. 2004;19:2539–2550 [DOI] [PubMed] [Google Scholar]
- 121. Borowicz KK, Swiader M, Wielosz M, et al. . Influence of the combined treatment of LY 300164 (an AMPA/kainate receptor antagonist) with adenosine receptor agonists on the electroconvulsive threshold in mice. Eur Neuropsychopharmacol. 2004;14:407–412 [DOI] [PubMed] [Google Scholar]
- 122. Dunwiddie TV, Diao L, Kim HO, et al. . Activation of hippocampal adenosine A3 receptors produces a desensitization of A1 receptor-mediated responses in rat hippocampus. J Neurosci. 1997;17:607–614 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123. Roseti C, Palma E, Martinello K, et al. . Blockage of A2A and A3 adenosine receptors decreases the desensitization of human GABA(A) receptors microtransplanted to Xenopus oocytes. Proc Natl Acad Sci U S A. 2009;106:15927–15931 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124. Abel T, Poplawski S. Epigenetic advances in clinical neuroscience. Dialogues Clin Neurosci. 2014;16:273–275 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125. Kobow K, Blumcke I. The methylation hypothesis: do epigenetic chromatin modifications play a role in epileptogenesis? Epilepsia. 2011;52(Suppl 4):15–19 [DOI] [PubMed] [Google Scholar]
- 126. Kobow K, Kaspi A, Harikrishnan KN, et al. . Deep sequencing reveals increased DNA methylation in chronic rat epilepsy. Acta Neuropathol. 2013;126:741–756 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127. Boison D, Rho JM. Epigenetics and epilepsy prevention: the therapeutic potential of adenosine and metabolic therapies. Neuropharmacology. In Press [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128. Hauser RM, Henshall DC, Lubin FD. The epigenetics of epilepsy and its progression. Neuroscientist. 2018;24:186–200 [DOI] [PubMed] [Google Scholar]
- 129. Kobow K, Blumcke I. The emerging role of DNA methylation in epileptogenesis. Epilepsia. 2012;53 Suppl 9:11–20 [DOI] [PubMed] [Google Scholar]
- 130. Miller-Delaney SF, Das S, Sano T, et al. . Differential DNA methylation patterns define status epilepticus and epileptic tolerance. J Neurosci. 2012;32:1577–1588 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131. Kobow K, Jeske I, Hildebrandt M, et al. . Increased reelin promoter methylation is associated with granule cell dispersion in human temporal lobe epilepsy. J Neuropathol Exp Neurol. 2009;68:356–364 [DOI] [PubMed] [Google Scholar]
- 132. Boison D, Scheurer L, Zumsteg V, et al. . Neonatal hepatic steatosis by disruption of the adenosine kinase gene. Proc Natl Acad Sci USA. 2002;99:6985–6990 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133. Chen Y, Swanson RA. Astrocytes and brain injury. J Cereb Blood Flow Metab. 2003;23:137–149 [DOI] [PubMed] [Google Scholar]
- 134. Pellerin L, Magistretti PJ. Sweet sixteen for ANLS. J Cereb Blood Flow Metab. 2012;32:1152–1166 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135. Liddelow SA, Guttenplan KA, Clarke LE, et al. . Neurotoxic reactive astrocytes are induced by activated microglia. Nature. 2017;541:481–487 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136. Coulter DA, Steinhauser C. Role of astrocytes in epilepsy. Cold Spring Harb Perspect Med. 2015;5:a022434. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137. Halassa MM, Fellin T, Haydon PG. The tripartite synapse: roles for gliotransmission in health and disease. Trends Mol Med. 2007;13:54–63 [DOI] [PubMed] [Google Scholar]
- 138. Cabezas R, Avila M, Gonzalez J, et al. . Astrocytic modulation of blood brain barrier: perspectives on Parkinson's disease. Front Cell Neurosci. 2014;8:211. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139. Losi G, Cammarota M, Carmignoto G. The role of astroglia in the epileptic brain. Front Pharmacol. 2012;3:1–13 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140. Patel DC, Tewari BP, Chaunsali L, et al. . Neuron-glia interactions in the pathophysiology of epilepsy. Nat Rev Neurosci. 2019;20:282–297 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141. Silver J, Miller JH. Regeneration beyond the glial scar. Nat Rev Neurosci. 2004;5:146–156 [DOI] [PubMed] [Google Scholar]
- 142. Babb TL, Mathern GW, Pretorius JK, et al. . Astrocytes may contribute to the latent period in progressive neuron loss, axon sprouting, and chronic seizures in rat kainate hippocampal epilepsy. Epilepsy Res Suppl. 1996;12:343–354 [PubMed] [Google Scholar]
- 143. Vargas-Sanchez K, Mogilevskaya M, Rodriguez-Perez J, et al. . Astroglial role in the pathophysiology of status epilepticus: an overview. Oncotarget. 2018;9:26954–26976 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144. Rouach N, Koulakoff A, Abudara V, et al. . Astroglial metabolic networks sustain hippocampal synaptic transmission. Science. 2008;322:1551–1555 [DOI] [PubMed] [Google Scholar]
- 145. Eid T, Williamson A, Lee TS, et al. . Glutamate and astrocytes—key players in human mesial temporal lobe epilepsy? Epilepsia. 2008;49(Suppl 2):42–52 [DOI] [PubMed] [Google Scholar]
- 146. Pascual O, Casper KB, Kubera C, et al. . Astrocytic purinergic signaling coordinates synaptic networks. Science. 2005;310:113–116 [DOI] [PubMed] [Google Scholar]
- 147. Li T, Lytle N, Lan J-Q, et al. . Local disruption of glial adenosine homeostasis in mice associates with focal electrographic seizures: a first step in epileptogenesis? Glia. 2012;60:83–95 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148. Matos M, Shen HY, Augusto E, et al. . Deletion of adenosine A2A receptors from astrocytes disrupts glutamate homeostasis leading to psychomotor and cognitive impairment: relevance to schizophrenia. Biol Psychiatry. 2015;78:763–774 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149. Robel S, Buckingham SC, Boni JL, et al. . Reactive astrogliosis causes the development of spontaneous seizures. J Neurosci. 2015;35:3330–3345 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150. Studer FE, Fedele DE, Marowsky A, et al. . Shift of adenosine kinase expression from neurons to astrocytes during postnatal development suggests dual functionality of the enzyme. Neuroscience. 2006;142:125–137 [DOI] [PubMed] [Google Scholar]
- 151. Gouder N. Overexpression of adenosine kinase in epileptic hippocampus contributes to epileptogenesis. J Neurosci. 2004;24:692–701 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152. Gonçalves ML, Cunha RA, Ribeiro JA. Adenosine A2A receptors facilitate 45Ca2+ uptake through class A calcium channels in rat hippocampal CA3 but not CA1 synaptosomes. Neurosci Lett. 1997;238:73–77 [DOI] [PubMed] [Google Scholar]
- 153. Temido-Ferreira M, Ferreira DG, Batalha VL, et al. . Age-related shift in LTD is dependent on neuronal adenosine A2A receptors interplay with mGluR5 and NMDA receptors. Mol Psychiatry. 2018. [Epub ahead of print]; DOI: 10.1038/s41380-018-0110-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 154. Gonçalves FQ, Lopes JP, Silva HB, et al. . Synaptic and memory dysfunction in a beta-amyloid model of early Alzheimer's disease depends on increased formation of ATP-derived extracellular adenosine. Neurobiol Dis. 2019;132:104570. [DOI] [PubMed] [Google Scholar]
- 155. Vezzani A, Friedman A. Brain inflammation as a biomarker in epilepsy. Biomark Med. 2011;5:607–614 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156. Boison D, Aronica E. Comorbidities in neurology: is adenosine the common link? Neuropharmacology. 2015;97:18–34 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 157. Aronica E, Zurolo E, Iyer A, et al. . Upregulation of adenosine kinase in astrocytes in experimental and human temporal lobe epilepsy. Epilepsia. 2011;52:1645–1655 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 158. Masino SA, Kawamura M Jr., Ruskin DN, et al. . Purines and neuronal excitability: links to the ketogenic diet. Epilepsy Res. 2012;100:229–238 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 159. Masino SA, Li T, Theofilas P, et al. . A ketogenic diet suppresses seizures in mice through adenosine A 1 receptors. J Clin Invest. 2011;121:2679–2683 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 160. Boison D. Adenosine dysfunction in epilepsy. Glia. 2012;60:1234–1243 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 161. Li T, Lan JQ, Boison D. Uncoupling of astrogliosis from epileptogenesis in adenosine kinase (ADK) transgenic mice. Neuron Glia Biol. 2008;4:91–99 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 162. Bannon NM, Chistiakova M, Chen JY, et al. . Adenosine shifts plasticity regimes between associative and homeostatic by modulating heterosynaptic changes. J Neurosci. 2017;37:1439–1452 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 163. Serrano A, Haddjeri N, Lacaille JC, et al. . GABAergic network activation of glial cells underlies hippocampal heterosynaptic depression. J Neurosci. 2006;26:5370–5382 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 164. Sandau US, Colino-Oliveira M, Jones A, et al. . Adenosine kinase deficiency in the brain results in maladaptive synaptic plasticity. J Neurosci. 2016;36:12117–12128 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 165. Loscher W, Brandt C. Prevention or modification of epileptogenesis after brain insults: experimental approaches and translational research. Pharmacol Rev. 2010;62:668–700 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 166. McIntosh AM, Berkovic SF. Treatment of new-onset epilepsy: seizures beget discussion. Lancet. 2005;365:1985–1986 [DOI] [PubMed] [Google Scholar]
- 167. Beamer E, Conte G, Engel T. ATP release during seizures—a critical evaluation of the evidence. Brain Res Bull. 2019;151:65–73 [DOI] [PubMed] [Google Scholar]
- 168. Cunha RA, Almeida T, Ribeiro JA. Parallel modification of adenosine extracellular metabolism and modulatory action in the hippocampus of aged rats. J Neurochem. 2001;76:372–382 [DOI] [PubMed] [Google Scholar]
- 169. Dona F, Conceicao IM, Ulrich H, et al. . Variations of ATP and its metabolites in the hippocampus of rats subjected to pilocarpine-induced temporal lobe epilepsy. Purinergic Signal. 2016;12:295–302 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 170. Wieraszko A, Seyfried TN. Increased amount of extracellular ATP in stimulated hippocampal slices of seizure prone mice. Neurosci Lett. 1989;106:287–293 [DOI] [PubMed] [Google Scholar]
- 171. Bonan CD, Walz R, Pereira GS, et al. . Changes in synaptosomal ectonucleotidase activities in two rat models of temporal lobe epilepsy. Epilepsy Res. 2000;39:229–238 [DOI] [PubMed] [Google Scholar]
- 172. Schoen SW, Ebert U, Loscher W. 5′-Nucleotidase activity of mossy fibers in the dentate gyrus of normal and epileptic rats. Neuroscience. 1999;93:519–526 [DOI] [PubMed] [Google Scholar]
- 173. Augusto E, Matos M, Sevigny J, et al. . Ecto-5'-nucleotidase (CD73)-mediated formation of adenosine is critical for the striatal adenosine A2A receptor functions. J Neurosci. 2013;33:11390–11399 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 174. Carmo M, Gonçalves FQ, Canas PM, et al. . Enhanced ATP release and CD73-mediated adenosine formation sustain adenosine A2A receptor over-activation in a rat model of Parkinson's disease. Br J Pharmacol. 2019;176:3666–3680 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 175. Lee SK. Epilepsy in the elderly: treatment and consideration of comorbid diseases. J Epilepsy Res. 2019;9:27–35 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 176. Cloyd J, Hauser W, Towne A, et al. . Epidemiological and medical aspects of epilepsy in the elderly. Epilepsy Res. 2006;68(Suppl 1):S39–S48 [DOI] [PubMed] [Google Scholar]
- 177. Johnson EL, Krauss GL, Lee AK, et al. . Association between midlife risk factors and late-onset epilepsy: results from the atherosclerosis risk in communities study. JAMA Neurol. 2018;75:1375–1382 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 178. Sarkis RA, Willment KC, Pennell PB, et al. . Late-onset unexplained epilepsy: what are we missing? Epilepsy Behav. 2019:106478. [DOI] [PubMed] [Google Scholar]
- 179. Burke SN, Barnes CA. Neural plasticity in the ageing brain. Nat Rev Neurosci. 2006;7:30–40 [DOI] [PubMed] [Google Scholar]
- 180. Rodrigues RJ, Canas PM, Lopes LV, et al. . Modification of adenosine modulation of acetylcholine release in the hippocampus of aged rats. Neurobiol Aging. 2008;29:1597–1601 [DOI] [PubMed] [Google Scholar]
- 181. Pazzagli M, Corsi C, Fratti S, et al. . Regulation of extracellular adenosine levels in the striatum of aging rats. Brain Res. 1995;684:103–106 [DOI] [PubMed] [Google Scholar]
- 182. Pagonopoulou O, Angelatou F. Reduction of A1 adenosine receptors in cortex, hippocampus and cerebellum in ageing mouse brain. Neuroreport. 1992;3:735–737 [DOI] [PubMed] [Google Scholar]
- 183. Cunha RA, Constantino MC, Sebastião AM, et al. . Modification of A1 and A2a adenosine receptor binding in aged striatum, hippocampus and cortex of the rat. Neuroreport. 1995;6:1583–1588 [DOI] [PubMed] [Google Scholar]
- 184. Sebastião AM, Cunha RA, de Mendonça A, et al. . Modification of adenosine modulation of synaptic transmission in the hippocampus of aged rats. Br J Pharmacol. 2000;131:1629–1634 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 185. Meerlo P, Roman V, Farkas E, et al. . Ageing-related decline in adenosine A1 receptor binding in the rat brain: an autoradiographic study. J Neurosci Res. 2004;78:742–748 [DOI] [PubMed] [Google Scholar]
- 186. Costenla AR, Diógenes MJ, Canas PM, et al. . Enhanced role of adenosine A(2A) receptors in the modulation of LTP in the rat hippocampus upon ageing. Eur J Neurosci. 2011;34:12–21 [DOI] [PubMed] [Google Scholar]
- 187. Meyer PT, Elmenhorst D, Boy C, et al. . Effect of aging on cerebral A1 adenosine receptors: a [18F]CPFPX PET study in humans. Neurobiol Aging. 2007;28:1914–1924 [DOI] [PubMed] [Google Scholar]
- 188. Mishina M, Ishiwata K, Naganawa M, et al. . Adenosine A(2A) receptors measured with [C]TMSX PET in the striata of Parkinson's disease patients. PLoS One. 2011;6:e17338. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 189. Lalo U, Bogdanov A, Pankratov Y. Age- and experience-related plasticity of ATP-mediated signaling in the neocortex. Front Cell Neurosci. 2019;13:242. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 190. Fuchs JL. 5′-Nucleotidase activity increases in aging rat brain. Neurobiol Aging. 1991;12:523–530 [DOI] [PubMed] [Google Scholar]
- 191. Mackiewicz M, Nikonova EV, Zimmermann JE, et al. . Age-related changes in adenosine metabolic enzymes in sleep/wake regulatory areas of the brain. Neurobiol Aging. 2006;27:351–360 [DOI] [PubMed] [Google Scholar]
- 192. Canas PM, Duarte JM, Rodrigues RJ, et al. . Modification upon aging of the density of presynaptic modulation systems in the hippocampus. Neurobiol Aging. 2009;30:1877–1884 [DOI] [PubMed] [Google Scholar]
- 193. Garcez ML, Damiani AP, Pacheco R, et al. . Caffeine neuroprotection decreases A2A adenosine receptor content in aged mice. Neurochem Res. 2019;44:787–795 [DOI] [PubMed] [Google Scholar]
- 194. Lopes LV, Cunha RA, Ribeiro JA. Increase in the number, G protein coupling, and efficiency of facilitatory adenosine A2A receptors in the limbic cortex, but not striatum, of aged rats. J Neurochem. 1999;73:1733–1738 [DOI] [PubMed] [Google Scholar]
- 195. Rebola N, Sebastião AM, de Mendonça A, et al. . Enhanced adenosine A2A receptor facilitation of synaptic transmission in the hippocampus of aged rats. J Neurophysiol. 2003;90:1295–1303 [DOI] [PubMed] [Google Scholar]
- 196. Coelho JE, Alves P, Canas PM, et al. . Overexpression of adenosine A2A receptors in rats: effects on depression, locomotion, and anxiety. Front Psychiatry. 2014;5:67. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 197. Li P, Rial D, Canas PM, et al. . Optogenetic activation of intracellular adenosine A2A receptor signaling in the hippocampus is sufficient to trigger CREB phosphorylation and impair memory. Mol Psychiatry. 2015;20:1481. [DOI] [PubMed] [Google Scholar]
- 198. Pagnussat N, Almeida AS, Marques DM, et al. . Adenosine A(2A) receptors are necessary and sufficient to trigger memory impairment in adult mice. Br J Pharmacol. 2015;172:3831–3845 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 199. Temido-Ferreira M, Coelho JE, Pousinha PA, et al. . Novel players in the aging synapse: impact on cognition. J Caffeine Adenosine Res. 2019;9:104–127 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 200. Carvalho K, Faivre E, Pietrowski M, et al. . Exacerbation of C1q dysregulation, synaptic loss and memory deficits in tau pathology linked to neuronal adenosine A2A receptors. Brain. 2019;142:3636–3654 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 201. Bagnall RD, Crompton DE, Semsarian C. Genetic basis of sudden unexpected death in epilepsy. Front Neurol. 2017;8:348. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 202. Devinsky O, Hesdorffer DC, Thurman DJ, et al. . Sudden unexpected death in epilepsy: epidemiology, mechanisms, and prevention. Lancet Neurol. 2016;15:1075–1088 [DOI] [PubMed] [Google Scholar]
- 203. DeGiorgio CM, Curtis A, Hertling D, et al. . Sudden unexpected death in epilepsy: risk factors, biomarkers, and prevention. Acta Neurol Scand. 2018;139:220–230 [DOI] [PubMed] [Google Scholar]
- 204. Langan Y, Nashef L, Sander JW. Sudden unexpected death in epilepsy: a series of witnessed deaths. J Neurol Neurosurg Psychiatry. 2000;68:211–213 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 205. Nashef L, Walker F, Allen P, et al. . Apnoea and bradycardia during epileptic seizures: relation to sudden death in epilepsy. J Neurol Neurosurg Psychiatry. 1996;60:297–300 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 206. Sainju RK, Dragon DN, Winnike HB, et al. . Ventilatory response to CO2 in patients with epilepsy. Epilepsia. 2019;60:508–517 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 207. Ryvlin P, Nashef L, Lhatoo SD, et al. . Incidence and mechanisms of cardiorespiratory arrests in epilepsy monitoring units (MORTEMUS): a retrospective study. Lancet Neurol. 2013;12:966–977 [DOI] [PubMed] [Google Scholar]
- 208. Ikeda K, Kawakami K, Onimaru H, et al. . The respiratory control mechanisms in the brainstem and spinal cord: integrative views of the neuroanatomy and neurophysiology. J Physiol Sci. 2017;67:45–62 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 209. Baker E, Lui F. Neuroanatomy, Vagal Nerve Nuclei (Nucleus Vagus). Treasure Island, FL: StatPearls; 2019 [PubMed] [Google Scholar]
- 210. Brennan TJ, Seeley WW, Kilgard M, et al. . Sound-induced seizures in serotonin 5-HT2c receptor mutant mice. Nat Genet. 1997;16:387–390 [DOI] [PubMed] [Google Scholar]
- 211. Shen HY, Li T, Boison D. A novel mouse model for sudden unexpected death in epilepsy (SUDEP): role of impaired adenosine clearance. Epilepsia. 2010;51:465–468 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 212. Faingold CL, Randall M, Tupal S. DBA/1 mice exhibit chronic susceptibility to audiogenic seizures followed by sudden death associated with respiratory arrest. Epilepsy Behav. 2010;17:436–440 [DOI] [PubMed] [Google Scholar]
- 213. Berman RF, Fredholm BB, Aden U, et al. . Evidence for increased dorsal hippocampal adenosine release and metabolism during pharmacologically induced seizures in rats. Brain Res. 2000;872:44–53 [DOI] [PubMed] [Google Scholar]
- 214. During MJ, Spencer DD. Adenosine: a potential mediator of seizure arrest and postictal refractoriness. Ann Neurol. 1992;32:618–624 [DOI] [PubMed] [Google Scholar]
- 215. Arch JR, Newsholme EA. Activities and some properties of 5'-nucleotidase, adenosine kinase and adenosine deaminase in tissues from vertebrates and invertebrates in relation to the control of the concentration and the physiological role of adenosine. Biochem J. 1978;174:965–977 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 216. Pak MA, Haas HL, Decking UK, et al. . Inhibition of adenosine kinase increases endogenous adenosine and depresses neuronal activity in hippocampal slices. Neuropharmacology. 1994;33:1049–1053 [DOI] [PubMed] [Google Scholar]
- 217. Zhang G, Franklin PH, Murray TF. Manipulation of endogenous adenosine in the rat prepiriform cortex modulates seizure susceptibility. J Pharmacol Exp Ther. 1993;264:1415–1424 [PubMed] [Google Scholar]
- 218. Gouder N, Scheurer L, Fritschy JM, et al. . Overexpression of adenosine kinase in epileptic hippocampus contributes to epileptogenesis. J Neurosci. 2004;24:692–701 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 219. Ugarkar BG, Castellino AJ, DaRe JM, et al. . Adenosine kinase inhibitors. 2. Synthesis, enzyme inhibition, and antiseizure activity of diaryltubercidin analogues. J Med Chem. 2000;43:2894–2905 [DOI] [PubMed] [Google Scholar]
- 220. Ugarkar BG, DaRe JM, Kopcho JJ, et al. . Adenosine kinase inhibitors. 1. Synthesis, enzyme inhibition, and antiseizure activity of 5-iodotubercidin analogues. J Med Chem. 2000;43:2883–2893 [DOI] [PubMed] [Google Scholar]
- 221. Kowaluk EA, Bhagwat SS, Jarvis MF. Adenosine kinase inhibitors. Curr Pharm Des. 1998;4:403–416 [PubMed] [Google Scholar]
- 222. Kowaluk EA, Jarvis MF. Therapeutic potential of adenosine kinase inhibitors. Expert Opin Investig Drugs. 2000;9:551–564 [DOI] [PubMed] [Google Scholar]
- 223. Zheng GZ, Lee C, Pratt JK, et al. . Pyridopyrimidine analogues as novel adenosine kinase inhibitors. Bioorg Med Chem Lett. 2001;11:2071–2074 [DOI] [PubMed] [Google Scholar]
- 224. McGaraughty S, Cowart M, Jarvis MF, et al. . Anticonvulsant and antinociceptive actions of novel adenosine kinase inhibitors. Curr Top Med Chem. 2005;5:43–58 [DOI] [PubMed] [Google Scholar]
- 225. Boison D. Engineered adenosine-releasing cells for epilepsy therapy: human mesenchymal stem cells and human embryonic stem cells. Neurotherapeutics. 2009;6:278–283 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 226. Huber A, Padrun V, Deglon N, et al. . Grafts of adenosine-releasing cells suppress seizures in kindling epilepsy. Proc Natl Acad Sci U S A. 2001;98:7611–7616 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 227. Guttinger M, Fedele D, Koch P, et al. . Suppression of kindled seizures by paracrine adenosine release from stem cell-derived brain implants. Epilepsia. 2005;46:1162–1169 [DOI] [PubMed] [Google Scholar]
- 228. Guttinger M, Padrun V, Pralong WF, et al. . Seizure suppression and lack of adenosine A1 receptor desensitization after focal long-term delivery of adenosine by encapsulated myoblasts. Exp Neurol. 2005;193:53–64 [DOI] [PubMed] [Google Scholar]
- 229. Li T, Steinbeck JA, Lusardi T, et al. . Suppression of kindling epileptogenesis by adenosine releasing stem cell-derived brain implants. Brain. 2007;130:1276–1288 [DOI] [PubMed] [Google Scholar]
- 230. Li T, Ren G, Lusardi T, et al. . Adenosine kinase is a target for the prediction and prevention of epileptogenesis in mice. J Clin Invest. 2008;118:571–582 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 231. Ren G, Boison D. Engineering human mesenchymal stem cells to release adenosine using miRNA technology. Methods Mol Biol. 2017;1622:225–239 [DOI] [PubMed] [Google Scholar]
- 232. Ren G, Li T, Lan JQ, et al. . Lentiviral RNAi-induced downregulation of adenosine kinase in human mesenchymal stem cell grafts: a novel perspective for seizure control. Exp Neurol. 2007;208:26–37 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 233. Theofilas P, Brar S, Stewart KA, et al. . Adenosine kinase as a target for therapeutic antisense strategies in epilepsy. Epilepsia. 2011;52:589–601 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 234. Masino SA, Kawamura M, Wasser CA, et al. . Adenosine, ketogenic diet and epilepsy: the emerging therapeutic relationship between metabolism and brain activity. Curr Neuropharmacol. 2009;7:257–268 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 235. Cunha RA. Adenosine as a neuromodulator and as a homeostatic regulator in the nervous system: different roles, different sources and different receptors. Neurochem Int. 2001;38:107–125 [DOI] [PubMed] [Google Scholar]
- 236. Lusardi TA, Akula KK, Coffman SQ, et al. . Ketogenic diet prevents epileptogenesis and disease progression in adult mice and rats. Neuropharmacology. 2015;99:500–509 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 237. Caraballo R, Darra F, Reyes G, et al. . The ketogenic diet in patients with myoclonic status in non-progressive encephalopathy. Seizure. 2017;51:1–5 [DOI] [PubMed] [Google Scholar]
- 238. Chen F, He X, Luan G, et al. . Role of DNA methylation and adenosine in ketogenic diet for pharmacoresistant epilepsy: focus on epileptogenesis and associated comorbidities. Front Neurol. 2019;10:119. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 239. Neal EG, Chaffe H, Schwartz RH, et al. . The ketogenic diet for the treatment of childhood epilepsy: a randomised controlled trial. Lancet Neurol. 2008;7:500–506 [DOI] [PubMed] [Google Scholar]
- 240. Sharma S, Jain P. The modified atkins diet in refractory epilepsy. Epilepsy Res Treat. 2014;2014:404202. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 241. Bough KJ, Rho JM. Anticonvulsant mechanisms of the ketogenic diet. Epilepsia. 2007;48:43–58 [DOI] [PubMed] [Google Scholar]
- 242. Klein P, Tyrlikova I. Prevention of epilepsy: should we be avoiding clinical trials? Epilepsy Behav. 2017;72:188–194 [DOI] [PubMed] [Google Scholar]
- 243. Szybala C, Pritchard EM, Wilz A, et al. . Antiepileptic effects of silk-polymer based adenosine release in kindled rats. Exp Neurol. 2009;219:126–135 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 244. Canas PM, Porciúncula LO, Cunha GM, et al. . Adenosine A2A receptor blockade prevents synaptotoxicity and memory dysfunction caused by beta-amyloid peptides via p38 mitogen-activated protein kinase pathway. J Neurosci. 2009;29:14741–14751 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 245. Laurent C, Burnouf S, Ferry B, et al. . A2A adenosine receptor deletion is protective in a mouse model of Tauopathy. Mol Psychiatry. 2016;21:97–107 [DOI] [PubMed] [Google Scholar]
- 246. Viana da Silva S, Haberl MG, Zhang P, et al. . Early synaptic deficits in the APP/PS1 mouse model of Alzheimer's disease involve neuronal adenosine A2A receptors. Nat Commun. 2016;7:11915. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 247. Faivre E, Coelho JE, Zornbach K, et al. . Beneficial effect of a selective adenosine A2A receptor antagonist in the APPswe/PS1dE9 mouse model of Alzheimer's disease. Front Mol Neurosci. 2018;11:235. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 248. Silva AC, Lemos C, Gonçalves FQ, et al. . Blockade of adenosine A2A receptors recovers early deficits of memory and plasticity in the triple transgenic mouse model of Alzheimer's disease. Neurobiol Dis. 2018;117:72–81 [DOI] [PubMed] [Google Scholar]
- 249. Kaster MP, Machado NJ, Silva HB, et al. . Caffeine acts through neuronal adenosine A2A receptors to prevent mood and memory dysfunction triggered by chronic stress. Proc Natl Acad Sci U S A. 2015;112:7833–7838 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 250. López-Cruz L, Carbó-Gas M, Pardo M, et al. . Adenosine A2A receptor deletion affects social behaviors and anxiety in mice: involvement of anterior cingulate cortex and amygdala. Behav Brain Res. 2017;321:8–17 [DOI] [PubMed] [Google Scholar]
- 251. Padilla KM, Quintanar-Setephano A, Lopez-Vallejo F, et al. . Behavioral changes induced through adenosine A2A receptor ligands in a rat depression model induced by olfactory bulbectomy. Brain Behav. 2018;8:e00952. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 252. Hohoff C, Marziniak M, Lesch KP, et al. . An adenosine A2A receptor gene haplotype is associated with migraine with aura. Cephalalgia. 2007;27:177–181 [DOI] [PubMed] [Google Scholar]
- 253. Haanes KA, Labastida-Ramirez A, Chan KY, et al. . Characterization of the trigeminovascular actions of several adenosine A2A receptor antagonists in an in vivo rat model of migraine. J Headache Pain. 2018;19:41. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 254. Korkutata M, Saitoh T, Cherasse Y, et al. . Enhancing endogenous adenosine A2A receptor signaling induces slow-wave sleep without affecting body temperature and cardiovascular function. Neuropharmacology. 2019;144:122–132 [DOI] [PubMed] [Google Scholar]
- 255. Lazarus M, Chen JF, Huang ZL, et al. . Adenosine and sleep. Handb Exp Pharmacol. 2019;253:359–381 [DOI] [PubMed] [Google Scholar]