Abstract
Alzheimer’s disease (AD) is a neurodegenerative disorder characterized by a progressive loss of memory and cognitive ability and is a serious cause of mortality. Many of the pathological characteristics associated with AD are revealed post-mortem, including amyloid-β plaque deposition, neurofibrillary tangles containing hyperphosphorylated tau proteins and neuronal loss in the hippocampus and cortex. Although several genetic mutations and risk factors have been associated with the disease, the causes remain poorly understood. Study of disease-initiating mechanisms and AD progression in humans is inherently difficult as most available tissue specimens are from late-stages of disease. Therefore, AD researchers rely on in vitro studies and the use of AD animal models where neuroinflammation has been shown to be a major characteristic of AD. Purinergic receptors are a diverse family of proteins consisting of P1 adenosine receptors and P2 nucleotide receptors for ATP, UTP and their metabolites. This family of receptors has been shown to regulate a wide range of physiological and pathophysiological processes, including neuroinflammation, and may contribute to the pathogenesis of neurodegenerative diseases like Parkinson’s disease, multiple sclerosis and AD. Experimental evidence from human AD tissue has suggested that purinergic receptors may play a role in AD progression and studies using selective purinergic receptor agonists and antagonists in vitro and in AD animal models have demonstrated that purinergic receptors represent novel therapeutic targets for the treatment of AD.
Keywords: Alzheimer’s disease, Neuroinflammation, P2Y Receptor, P2X Receptor, P1 receptor
1. Introduction
Alzheimer’s disease (AD) is an age-related neurodegenerative disorder characterized by global cognitive decline, including progressive loss of memory, orientation, reasoning and functional abilities. It is predicted that AD prevalence will grow rapidly in the United States with an estimated 13.8 million AD patients by the year 2050, almost triple the 4.7 million patients in 2010 (Hebert et al., 2013). The associated care required will present unprecedented challenges to the health care system (Hebert et al., 2013). The causes of AD are not well understood and many of the pathological characteristics of the AD brain are revealed postmortem, including β-amyloid plaque deposition, hyperphosphorylated tau tangle formation and degeneration of neurons in the hippocampus and the neocortex (Butterfield and Boyd-Kimball, 2004; Hurtado et al., 2010; Obulesu et al., 2011). A primary hallmark of AD is the production of the amyloid-β1-42 peptide (Aβ) that results from the sequential processing of amyloid precursor protein (APP) by β- and γ-secretases resulting in a buildup of neurotoxic Aβ peptide and eventual plaque deposition and neurodegeneration (O'Brien and Wong, 2011). Several genetic mutations within the APP processing pathway have been shown to cause early-onset AD, namely mutations in APP itself or in the γ-secretase subunits presenilin-1 (PS1) and presenilin-2, although these mutations represent less than 5% of all AD cases (Lemere et al., 1996; Campion et al., 1999; O'Brien and Wong, 2011). The vast majority of AD patients develop late-onset AD with no known genetic cause, although several risk-factor genes have been identified, including the apoE4 allele of apolipoprotein. Individuals that inherit one copy of apoE4 have a 30% lifetime risk of developing AD by the age of 85 while those with 2 copies of apoE4 have a 60% lifetime risk (Genin et al., 2011). How exactly the apoE4 gene allele contributes to the pathogenesis of AD is still not well understood.
Although methods to prevent or reverse AD are not available, it is predicted that a treatment that merely slows the progression of the disease could dramatically reduce the number of AD patients in the coming years (Hebert et al., 2013). Current FDA-approved treatments for AD include two classes of drugs: acetylcholinesterase inhibitors that prevent the breakdown of the neurotransmitter acetylcholine to improve cognition and NMDA receptor antagonists that reduce glutamate-induced neuronal excitotoxicity (Cummings, 2004; Huang and Mucke, 2012). Clinical improvement of AD symptoms using these drugs is modest and neither drug can prevent or reverse disease pathology (Cummings, 2004; Huang and Mucke, 2012). Several novel therapeutic strategies for AD are currently being pursued, including decreasing production or increasing clearance of the amyloidogenic Aβ peptide, preventing tau phosphorylation and aggregation, and administration of anti-inflammatory agents (Cummings, 2004; Cole and Frautschy, 2010; Huang and Mucke, 2012). Unfortunately, these therapeutic strategies have been ineffective and in some cases cause undesirable side effects (Scharf et al., 1999; Aisen et al., 2000; Aisen et al., 2003; Gilman et al., 2005; Green et al., 2009; Salloway et al., 2009; Schor, 2011). The absence of effective treatments to retard the progression of AD strongly encourages attempts to identify initiating mechanisms that could potentially serve as therapeutic targets.
Because the majority of available human AD tissue samples are from deceased patients in the late-stages of disease, investigating the initiating cellular mechanisms and the progression of AD pathologies in humans is difficult. Furthermore, the inability to perform mechanistic studies using live cells and tissues makes most studies of human samples primarily correlative (Kalaria et al., 1990; Ulas et al., 1993; Angulo et al., 2003; Lai et al., 2008). As a result, AD researchers have relied heavily upon the use of transgenic AD-like mouse models and in vitro studies of brain tissue from these mice to investigate the initiating mechanisms of AD. Findings with these approaches have revealed that inflammation is a major characteristic of AD that is manifested by the increased accumulation of cytokines, e.g., IL-1β, TNF-α and IFN-γ (Chakfe et al., 2002; Suzuki et al., 2004), the enhanced activation of microglial cells and astrocytes (Frautschy et al., 1998; Schlachetzki and Hull, 2009; Simpson et al., 2010) and production of neurotoxic forms of Aβ (D'Andrea et al., 2001; Hardy and Selkoe, 2002; Nagele et al., 2003; Butterfield and Boyd-Kimball, 2004; Willuweit et al., 2009). Although it is clear that neuroinflammation is a major component of AD (Heppner et al., 2015), its role in disease pathogenesis is less clear as it could be a response to Aβ accumulation and neurodegeneration, a primary driver of AD progression or an early protective response (Wyss-Coray, 2006). Early in AD progression, inflammation may be neuroprotective, as acute activation of resident immune cells (i.e., microglia) by inflammatory molecules can increase the clearance of neurotoxic Aβ and dead neurons to help maintain brain homeostasis and synapse stability (Koizumi et al., 2007; Kim et al., 2012). Alternatively, chronic inflammation within the brain can enhance reactive gliosis and the production of damaging reactive oxygen species (ROS) that directly contribute to neurodegeneration (Parvathenani et al., 2003; Pekny et al., 2014). Identifying anti-neuroinflammatory drugs and the ideal stage of disease for their administration could lead to novel treatments for AD.
Purinergic signaling through P1 receptors for adenosine and P2 receptors for adenosine 5’-triphosphate (ATP), uridine 5’-triphosphate (UTP) and their metabolites is now recognized as playing a prominent role in the modulation of many physiological processes, including immune cell recruitment, inflammation, neurotransmission, regulation of vascular and muscle tone and perception of pain (Di Virgilio, 1998; Abbracchio and Ceruti, 2007; Trautmann, 2009; Tsuda et al., 2009; Burnstock, 2010; Stagg and Smyth, 2010; Bours et al., 2011). P1 and P2 receptor subtypes are expressed throughout the nervous system where they have been shown to play a role in Parkinson’s disease, Huntington’s disease and multiple sclerosis (Popoli et al., 2002; Chen and Brosnan, 2006; Pinna, 2014; Burnstock, 2015), making these receptors attractive drug targets for novel therapies to treat AD. To date, there have been no human clinical trials undertaken to investigate the therapeutic potential of targeting specific purinergic receptors to treat AD, although a recently-announced Phase II clinical trial will determine the therapeutic effectiveness of intravenous ATP infusion on cerebral metabolism and mental state in patients with moderate to severe AD (ClinicalTrials.gov: NCT02279511). The therapeutic potential of purinergic receptors in other human inflammatory and neurodegenerative diseases has already been recognized, as a number of clinical trials have investigated their role in the treatment of rheumatoid arthritis, inflammatory bowel disease, cancer and Parkinson’s disease (Agteresch et al., 2000; Villalona-Calero et al., 2008; Beijer et al., 2010; Arulkumaran et al., 2011; Hauser et al., 2014). The role of purinergic receptors in neuroinflammation, neurodegeneration and CNS disorders has been extensively reviewed. (Apolloni et al., 2009; Burnstock et al., 2011; Weisman et al., 2012b; Weisman et al., 2012c; Burnstock, 2015). Here, we focus on the relevance of recent studies on purinergic signaling to potential therapies for Alzheimer’s disease, with an emphasis on findings with human samples and studies in which selective agonists and antagonists for P1 and P2 receptor subtypes have been investigated in vitro and in mouse models of AD.
2. P1 receptors in AD
P1 adenosine receptors are a family of seven transmembrane domain G protein-coupled receptors ranging from 318–412 amino acids with 4 subtypes having been identified: A1, A2A, A2B and A3 receptors (Piirainen et al., 2011). These receptors can further be classified according to their preferred G protein interaction, where A1 and A3 receptors primarily couple to Gi protein leading to inhibition of adenylate cyclase and decreased cyclic adenosine 5’-monophosphate (cAMP) levels, whereas A2A and A2B receptors primarily signal through Gs protein leading to activation of adenylate cyclase and increased cAMP production, although coupling of each P1 receptor subtype to other G proteins has been described (Fredholm et al., 2001). Within the central nervous system (CNS), P1 receptors have been shown to play a role in a number of neurodegenerative diseases, including AD, making them potential therapeutic targets (Schwarzschild et al., 2006; Rahman, 2009; Stone et al., 2009; Rivera-Oliver and Diaz-Rios, 2014).
Although there exists sufficient evidence to suggest that A2B and A3 receptors represent therapeutic targets in the CNS (Rosi et al., 2003; Chen et al., 2006; Popoli and Pepponi, 2012; Little et al., 2015), A1 and A2A receptor antagonism has been the most well studied therapeutic approach involving P1 receptors and neurological disorders. In Parkinson’s Disease (PD), A2A receptors are thought to contribute to disease pathologies through modulation of dopaminergic signaling, where A2A receptors decrease the binding affinity of dopamine to D2 receptors (Diaz-Cabiale et al., 2001; Morelli et al., 2010). Furthermore, studies in human PD patients have shown A2A receptors to be upregulated in the brain regions affected by PD and increased A2A receptor expression is an early event in disease progression (Mishina et al., 2011; Villar-Menendez et al., 2014). A2A receptor antagonists, including Istradefylline (KW-6002), Preladenant (SCH420814) and Tozadenant (SYN115), have undergone Phase I-III clinical trials with varying degrees of success in the treatment of Parkinson’s Disease (LeWitt et al., 2008; Hauser et al., 2011; Hauser et al., 2014; Pinna, 2014). Interestingly, the most widely consumed drug in the world, caffeine (Fredholm et al., 1999), is an A1 and A2A receptor antagonist and a Phase III clinical trial is currently underway to assess caffeine administration as an effective therapy for Parkinson’s Disease (ClinicalTrials.gov: NCT01738178). Several epidemiological studies in humans also have demonstrated the beneficial cognitive effects of caffeine consumption that have been associated with a lower risk of developing PD, AD and dementia (Ascherio et al., 2001; Maia and de Mendonca, 2002; Eskelinen and Kivipelto, 2010) and reduced age-related cognitive decline (van Gelder et al., 2007). While the contributions of A1 and A2A receptors to the development and progression of AD in humans are unknown, the expression of these receptors is altered in areas of the brain known to be involved in the progression of the disease. Previous studies using post-mortem brain samples from AD patients and age-matched controls have shown decreased A1 receptor expression in both the dentate gyrus and CA3 regions of the hippocampus of AD patients, focal points for the spread of AD-associated neurofibrillary tangles and neuronal loss (Braak and Braak, 1996), which was attributed to neuronal cell death in these brain regions (Jansen et al., 1990; Kalaria et al., 1990; Ulas et al., 1993). In contrast, another study utilized post-mortem AD frontal cortex samples and showed increased expression of A1 and A2A receptors, as compared to the frontal cortex of age-matched control patients (Albasanz et al., 2008). Yet another study using AD patient samples and age-matched controls found that expression of A1 receptors was increased in degenerating neurons and around Aβ plaques in the AD brain, whereas A2A receptor expression, which was found primarily in neurons of the striatum in control patients, was increased in glial cells of the cortex and hippocampus of AD patients (Angulo et al., 2003). Taken together, these studies provide evidence of P1 receptor contributions to AD-related pathologies and suggest that the expression and distribution of these receptors in the AD brain is region- and cell type-specific.
Unlike studies and clinical trials with A1 and A2A receptor antagonists or agonists for the treatment of Parkinson’s disease, investigations of their potential as therapeutic agents in Alzheimer’s disease have been limited to in vitro studies and the use of animal models of AD. In the SH-SY5Y human neuroblastoma cell line, it has been shown that activation of the A1 receptor (A1R) using the selective agonist (R)-N6-(1-methyl-2-phenylethyl)adenosine (R-PIA) can enhance soluble APPα (sAPPα) production through a PKC- and ERK1/2-dependent mechanism suggesting that activation of the A1R is neuroprotective by promoting α-secretase-mediated non-amyloidogenic sAPPα production (Angulo et al., 2003). A1R-mediated sAPPα production was further shown to be attenuated by the selective A1R antagonist dipropylcyclopentylxanthine (DPCPX) (Angulo et al., 2003). Additionally, the neuroprotective effects of caffeine have been demonstrated in cultured rat cerebellar neurons where its application reduced Aβ-induced neurotoxicity primarily through A2A receptor (A2AR) antagonism, as addition of the selective A2AR antagonist ZM241385 mirrored the neuroprotective effects of caffeine whereas the selective A1R antagonist 8-cyclopentyltheophylline (CPT) had no effect (Dall'Igna et al., 2003). Caffeine has also been shown to produce neuroprotective effects in vivo in transgenic AD mouse models expressing mutant forms of APP and/or PS1. Administration of caffeine to transgenic mice expressing the Swedish mutant of APP (APPswe) beginning at 4 months of age through 8 months of age was shown to protect against cognitive decline in a battery of behavioral tests, as well as reduce the levels of neurotoxic Aβ1-42 in the hippocampus (Arendash et al., 2006). Although the exact mechanism of caffeine-mediated neuroprotection was unclear, in vitro release of Aβ1–40 and Aβ1-42 was decreased in a dose-dependent manner following caffeine addition to differentiated N2a neuronal cells, suggesting that antagonism of neuronal A1Rs and/or A2ARs and decreased amyloidogenic Aβ production is neuroprotective (Arendash et al., 2006). In the APPswe and APP+PS1 mouse models of AD, short-term caffeine treatment (< 24 h) reduced plasma Aβ levels in both ~3 month-old mice (before Aβ plaque development in the brain) and 15~20 month-old mice (after Aβ plaque development) (Cao et al., 2009). Furthermore, long-term caffeine treatment (4 weeks) in ~18 month-old APPswe mice was shown to reduce Aβ plaque load in the brain and reverse cognitive declines (Cao et al., 2009). In the intracerebroventricular (ICV) Aβ injection mouse model, which demonstrates Aβ-induced cognitive impairment and neurotoxicity, both caffeine and the selective A2AR antagonist SCH58261 were shown to prevent Aβ-induced cognitive impairments (Dall’Igna et al., 2007) and SCH58261 was further shown to reduce synapse loss and hippocampal neuron toxicity following ICV Aβ administration in rats (Canas et al., 2009). Pharmacological A2AR blockade also attenuated Aβ-induced neuronal death in rat primary cultured neurons, likely through a reduction in A2AR-mediated p38 MAPK activation and preservation of hippocampal synaptosome function (Canas et al., 2009). Interestingly, while these studies have primarily targeted A1 and A2A receptor antagonism in the brain as a therapeutic modality, it has been shown that A1 and A2A receptor agonism using NECA (5’-N-ethylcarboxamidoadenosine; a pan-P1 receptor agonist) or the FDA-approved A2AR agonist Lexiscan (regadenoson) increases the permeability of the blood-brain barrier to macromolecules, including anti-Aβ antibodies, in the APPswe/PS1dE9 mouse model of AD (Carman et al., 2011). This effect was thought to be mediated through adenosine receptor signaling in brain endothelial cells that make up part of the blood-brain barrier. These findings suggest that NECA and Lexiscan could be used as a combination therapy with other drugs to increase brain penetrance and improve the likelihood of drugs reaching their intended target within the brain. The results of these studies further suggest that P1 receptors, specifically A1 and A2A adenosine receptors, may represent novel drug targets for the treatment of AD.
3. P2 receptors in Alzheimer’s disease
3.1. P2X receptors in Alzheimer’s disease
P2X receptors (P2XR) are a family of ATP-gated cation channels ranging in size from 379–595 amino acids in length and consisting of intracellular N- and C-termini, two transmembrane domains and a large extracellular loop that shares ~33% sequence homology between the seven P2X receptor subtypes (P2X1–7) and contains a putative consensus ATP-binding site (Vial et al., 2004; Weisman et al., 2012b). P2X receptor subtypes interact in a number of homotrimeric and heterotrimeric configurations to regulate a variety of physiological and pathophysiological processes in the CNS, including pain sensation, neurotransmitter release and neuroinflammation (Rodrigues et al., 2005; Khakh and North, 2006; Koles et al., 2007; Ulmann et al., 2008). The expression of P2X receptor subtypes in the CNS varies with brain region and cell type, with P2X2, P2X4 and P2X6 receptors being highly expressed in neurons where their activation has been shown to induce both pre- and post-synaptic responses (Tanaka et al., 1996; Kanjhan et al., 1999; Rubio and Soto, 2001; Watano et al., 2004; Rodrigues et al., 2005; Khakh and North, 2006). Although P2X receptor subtypes are widely expressed in the CNS in both neurons and glial cells (Khakh and North, 2006; Illes et al., 2012), the role of most P2X receptor subtypes in CNS pathologies is not clearly defined. However, one prominent functional role for P2X receptors is the regulation of pain sensation, particularly by the P2X3 receptor and P2X2/P2X3 receptor heterotrimers in neurons and P2X4 receptors in microglia (Chizh and Illes, 2001; Tsuda et al., 2003; Ford, 2012). Blockade of P2X3 and P2X2/3 receptors by local application of the selective antagonist A-317491 has been shown to attenuate both chronic inflammatory pain and thermal neuropathic pain in mice, suggesting that P2X3 and P2X2/3 receptor antagonists may represent novel analgesics (Jarvis et al., 2002; McGaraughty et al., 2003). The therapeutic relevance of P2X3 and P2X2/3 receptor blockade, as well as blockade of other P2X receptor subtypes, has not been investigated in the context of Alzheimer’s disease, except in the case of the P2X7 receptor.
Among the P2X receptors, the P2X7 receptor (P2X7R) has garnered the most attention due to its key role in the regulation of inflammatory responses in a number of pathophysiological circumstances, including bacterial infection, lung inflammation, salivary gland inflammation and neuroinflammation (Monif et al., 2009; Lucattelli et al., 2011; Weisman et al., 2012b; Woods et al., 2012; Csoka et al., 2015). High concentrations of extracellular ATP (> 0.1 mM), such as those observed under inflammatory conditions (Cauwels et al., 2014), are required for P2X7R activation, likely because the fully ionized form of ATP (i.e., ATP4−) is the primary agonist (Steinberg et al., 1987). P2X7R activation leads to the opening of non-selective cation channels whose sustained activity induces mitochondrial and plasma membrane depolarization, the formation of plasma membrane pores, plasma membrane blebbing, and production of reactive oxygen species (Weisman et al., 1984; Parvathenani et al., 2003; Verhoef et al., 2003; Lister et al., 2007; Roger et al., 2008; Lee et al., 2011). One of the primary roles of the P2X7R is in immune cells where its activation stimulates the formation and activation of the NLRP3 inflammasome leading to increased caspase-1 activity and subsequent processing and release of IL-1β, a cytokine whose levels are elevated in the AD brain (Di Virgilio, 2007; Simi et al., 2007; Shaftel et al., 2008; Woods et al., 2012). P2X7R activation also has been shown to increase the release of TNF-α, IL-18 and IL-6, as well as induce apoptotic cell death, making the P2X7R an attractive therapeutic target to reduce inflammation and ATP-induced apoptosis through P2X7R antagonism (Hide et al., 2000; Perregaux et al., 2000; Kong et al., 2005; Shieh et al., 2014). In fact, human clinical trials have been undertaken to investigate the therapeutic potential of P2X7R antagonists in inflammatory diseases, including rheumatoid arthritis, Crohn’s disease and chronic obstructive pulmonary disease (Arulkumaran et al., 2011). Although there is clinical interest in testing P2X7R antagonism as a therapy for human CNS disorders (Chrovian et al., 2014), no clinical trials to date have specifically targeted the P2X7R in the CNS. Therefore, evidence for the role of the P2X7R in AD has primarily come from in vitro studies and the use of animal models of AD.
The P2X7R is widely distributed in the CNS and its expression has been reported in neurons, astrocytes and microglia where P2X7R activation contributes to numerous physiological processes, including neurotransmitter release from neurons and intercellular crosstalk via Ca2+ signaling (Collo et al., 1997; Verderio and Matteoli, 2001; Sperlagh et al., 2002; Sperlagh et al., 2006). P2X7R also contributes to CNS pathologies where its antagonism has been shown to be therapeutic in mouse models of neurodegeneration and neuroinflammation, including ischemia, Huntington’s disease, multiple sclerosis and AD (Matute et al., 2007; Diaz-Hernandez et al., 2009; Chu et al., 2012; Diaz-Hernandez et al., 2012). Because microglia are primary drivers of neuroinflammation, antagonism of microglial P2X7Rs may reduce neuroinflammatory responses in the AD brain that have been shown to contribute to disease pathogenesis (Heppner et al., 2015). Exposure of both human and rodent microglia to Aβ has been shown to increase P2X7R expression (McLarnon et al., 2006) and induce the release of proinflammatory cytokines, including TNFα, IL-6 and IL-1β (Yates et al., 2000; Combs et al., 2001; Parajuli et al., 2013), whose levels have been shown to be elevated in the brains of human AD patients and in mouse models of AD (Griffin et al., 1989; Patel et al., 2005; Simi et al., 2007; Sanz et al., 2009). The P2X7R appears to play a prominent role in the regulation of IL-1β levels in the brain, as intrahippocampal Aβ injection causes a large accumulation of IL-1β in the hippocampi of wild type, but not P2X7R−/−, mice (Sanz et al., 2009). The lack of Aβ-induced IL-1β accumulation in P2X7R−/− mouse hippocampus is likely due to the loss of microglial P2X7Rs, as in vitro studies have shown that Aβ increases ATP release, [Ca2+]i, and IL-1β release from primary microglia of wild type, but not P2X7R−/−, mice, suggesting that microglial P2X7Rs are a primary regulator of inflammation in the AD brain (Sanz et al., 2009). In the human AD brain, P2X7R expression has been shown to colocalize with activated microglia surrounding Aβ plaques and, in microglia isolated posthumously from AD patients, P2X7R expression was shown to be significantly increased compared to microglia from non-demented patients (McLarnon et al., 2006). Similarly, the P2X7R was shown to be upregulated in both the Tg2576 and APPswe/PS1dE9 transgenic mouse models of AD and its expression colocalized with microglia surrounding Aβ plaques (Parvathenani et al., 2003; Lee et al., 2011). Furthermore, P2X7R activation by ATP or the high affinity P2X7R agonist BzATP (3'-O-(4-benzoyl)benzoyl-ATP) (Erb et al., 1990) significantly enhanced ROS production in primary rat microglia, and P2X7R–expressing microglia in APPswe/PS1dE9 mice exhibited significant ROS production that colocalized with decreased expression of the synapse protein postsynaptic density-95 (PSD-95), suggesting that P2X7R–mediated ROS production by microglia cells contributes to synaptic loss in this mouse model of AD (Parvathenani et al., 2003; Lee et al., 2011). Microglial P2X7R upregulation was also observed in a rat model where Aβ1-42 was directly injected into the hippocampus, which lead to memory deficits and degeneration of hippocampal neurons within 7 days (Kowall et al., 1991; Malin et al., 2001; McLarnon et al., 2006). Administration of the selective P2X7R antagonist Brilliant Blue G (BBG) (Jiang et al., 2000) following hippocampal Aβ1-42 injection was shown to confer neuroprotection and reduce inflammatory responses, reactive gliosis and neuronal loss in the hippocampus (Ryu and McLarnon, 2008). In a similar mouse model, BBG administration following intrahippocampal Aβ injection significantly decreased Aβ-induced cognitive deficits. However, neuronal P2X7Rs appear to contribute to Aβ-induced neurodegeneration in mice, since in vitro studies using primary hippocampal neurons demonstrated that blockade of neuronal P2X7Rs by BBG attenuated Aβ-induced loss of dendritic spines and filopodia (Chen et al., 2014). Similarly, neuronal P2X7R antagonism was shown to be therapeutic in the J20 hAPP transgenic mouse model of AD where systemic BBG administration prevented the formation of Aβ plaques (Diaz-Hernandez et al., 2012). It was demonstrated that inhibition of the P2X7R by BBG increased α-secretase activity in hippocampal neurons via glycogen synthase kinase 3-beta (GSK-3β), thus reducing the generation of Aβ and subsequent plaques, although decreased IL-1β accumulation in the hippocampus was also observed following BBG administration, suggesting that blockade of microglial P2X7R is also therapeutic in this model (Diaz-Hernandez et al., 2012). Beneficial effects of P2X7R antagonism also may result from blockade of P2X7R-mediated apoptosis, as neuronal P2X7R activation by ATP or BzATP has previously been shown to induce caspase- and FAS-mediated apoptosis (Kong et al., 2005; Gandelman et al., 2013).
Lastly, there is some evidence that the P2X4 receptor (P2X4R) may contribute to neurodegeneration in Alzheimer’s disease. It has been demonstrated that treatment of rat primary hippocampal neurons with the neurotoxic Aβ1-42 peptide increases P2X4R expression and cell death (Varma et al., 2009). Furthermore, increasing P2X4R expression through lentiviral-based transfection led to increased Aβ1-42-induced neuronal death, whereas decreasing P2X4R expression using P2X4R siRNA decreased Aβ-induced neuronal death (Varma et al., 2009). Neuronal death caused by Aβ was postulated to result from dysregulation of P2X4R-mediated Ca2+ influx whereby Aβ enhanced caspase 3-mediated cleavage of the P2X4R C-terminus thus increasing Ca2+ channel closure time and decreasing agonist-dependent receptor internalization from the cell membrane. As previous studies have already demonstrated that Aβ induces the release of the P2XR agonist ATP from microglia (Kim et al., 2007; Kim et al., 2012), it is suggested that increased Aβ levels in the AD brain enhance ATP release from microglia to increase P2X4R activation leading to neuronal cell death. Interestingly, P2X4R expression was shown to be downregulated in the frontal lobe of AD patients with severe cognitive impairment, as compared to non-diseased control patients, where it was hypothesized that Aβ-induced P2X4R expression increased neuronal death (Varma et al., 2009). The observed decrease in P2X4R expression in the AD brain may be indicative of the loss of P2X4R-expressing neurons in late-stage AD. These findings suggest that, in addition to the P2X7R, the P2X4R may represent a novel therapeutic target for the treatment of AD.
3.2. P2Y receptors in Alzheimer’s disease
P2Y receptors (P2YRs) are a family of classical seven transmembrane spanning G protein-coupled receptors with extracellular N-termini containing potential glycosylation sites and structurally diverse intracellular domains that couple with Gq, Gs or Gi proteins (Erb et al., 1995; Flores et al., 2005; Abbracchio et al., 2006; Burnstock, 2015). Eight different P2Y receptor subtypes (P2Y1, P2Y2, P2Y4, P2Y6, P2Y11-14) ranging from 328 to 377 amino acids have been cloned and characterized as having distinct pharmacological profiles in their response to endogenous agonists, including ATP, UTP, adenosine 5’-diphosphate (ADP), uridine 5’-diphosphate (UDP) and UDP-glucose (Abbracchio et al., 2006; Burnstock, 2006). Several positively charged amino acids that are conserved among the P2YR subtypes have been shown to facilitate the binding of fully ionized, negatively charged P2Y2R agonists to the receptor (Lustig et al., 1992; Erb et al., 1995). P2Y1, P2Y2, P2Y4, P2Y6 and P2Y11 receptors are coupled to the heterotrimeric Gq protein whose activation stimulates phospholipase C (PLC) and subsequent inositol 1,4,5-trisphosphate (IP3) and diacylglycerol generation leading to release of Ca2+ from intracellular stores and activation of protein kinase C (PKC), respectively (Abbracchio et al., 2006). The P2Y11 receptor also couples to Gs proteins to stimulate adenylate cyclase activity and increase production of the downstream effector cAMP (Nguyen et al., 2001). Alternatively, P2Y12, P2Y13 and P2Y14 receptors are coupled to Gi/o proteins whose activation inhibits adenylate cyclase and decreases cAMP production (Abbracchio et al., 2006). Activation of P2YRs regulates numerous physiological and pathophysiological processes, including inflammation, cell growth, wound healing and thrombosis (Boeynaems et al., 2005; Burnstock and Verkhratsky, 2010; Gendaszewska-Darmach and Kucharska, 2011; Idzko et al., 2014). Within the CNS, P2Y receptors are expressed in neurons, astrocytes, oligodendrocytes and microglia with physiological roles in neurotransmission, neurogenesis and glial cell communication (Burnstock et al., 1972; Guthrie et al., 1999; Fields and Burnstock, 2006; Mishra et al., 2006; Weisman et al., 2012a; Peterson et al., 2013). As the study of purinergic signaling in the CNS progresses, it has become increasingly clear that P2YRs play important pathophysiological roles where the activation of specific P2YR subtypes generates either neuroprotective or neurodegenerative responses making them novel targets for the treatment of CNS pathologies, including Alzheimer’s disease (Davalos et al., 2005; Sperlagh and Illes, 2007; Burnstock, 2008; Kuboyama et al., 2011; Kim et al., 2012; Weisman et al., 2012a).
Previous investigations of AD brain samples demonstrated that the expression of the P2Y1 receptor (P2Y1R) is primarily localized to neurons in both AD and age-matched control brains. However, in the AD brain P2Y1R expression also colocalized with neurofibrillary tangles and Aβ plaques, suggesting a possible role in AD pathology (Moore et al., 2000). While the role of neuronal P2Y1R expression in AD brain remains unclear, in vitro studies of adult mouse neural stem cell (NSC) cultures have demonstrated that P2Y1R activation by its endogenous agonist ADP induces proliferation of NSCs that is attenuated by the selective P2Y1R antagonist MRS2179 (Mishra et al., 2006). Proliferation of NSCs is a crucial step during the process of neurogenesis in which new neurons are formed from self-renewing adult NSCs in the mammalian hippocampus (van Praag et al., 2002; Mu and Gage, 2011). Functional neurogenesis during adulthood has been demonstrated in both rodents and humans where it has been shown to contribute to hippocampus-dependent memory and cognition (Kuhn et al., 1996; Eriksson et al., 1998; Shors et al., 2001; Deng et al., 2010). Because degeneration of hippocampal neurons is a hallmark of AD, stimulating endogenous NSCs to increase neurogenesis has been suggested to be a potential regenerative therapy in AD to replace damaged neurons and improve cognition (Brinton and Wang, 2006; Mu and Gage, 2011; Fuster-Matanzo et al., 2013). Thus, the ability to increase NSC proliferation through P2Y1R activation could be a novel therapeutic approach to promote regeneration of neurons in the hippocampus of AD patients. The cell-specific contributions of purinergic signaling have been clarified by recent work on the functional role of the P2Y1R in glial cells, particularly astrocytes. In the APPswe/PS1dE9 and APP+PS1 mouse models of AD, multiphoton fluorescence lifetime in vivo imaging (FLIM) was utilized to demonstrate that astrocytic networks in the AD mouse brain have significantly higher resting [Ca2+]i levels than in wild type animals and exhibited increased propagation of intercellular calcium waves near Aβ plaques, which was suggested to contribute to Aβ-induced neuronal damage and synapse loss (Kuchibhotla et al., 2009; Delekate et al., 2014). Furthermore, expression of the P2Y1R in the APP+PS1 mouse brain was shown to be primarily localized to reactive astrocytes near Aβ plaques and blockade of P2Y1R signaling using the selective antagonist MRS2179 abolished the astrocytic hyperactivity and reduced [Ca2+]i and intercellular calcium waves (Delekate et al., 2014), suggesting that P2Y1R inhibition in astrocytes could be therapeutic in the treatment of AD.
Among P2Y receptors, the P2Y2 receptor (P2Y2R) is unique in that, in addition to canonical Gq-coupled activation of the PLC/IP3/PKC pathway, it possesses several structural features allowing for interaction with other molecules to regulate cell migration, proliferation and growth factor release (Erb et al., 2001; Liu et al., 2004; Wang et al., 2005; Peterson et al., 2010; Ratchford et al., 2010). An Arg-Gly-Asp (RGD) motif in the first extracellular loop of the P2Y2R enables interaction with RGD-binding αV/β3/5 integrins and integrin-dependent activation of Go and G12 proteins that regulates the activities of the small GTPases Rho and Rac (Erb et al., 2001; Bagchi et al., 2005; Liao et al., 2007). In this way, activation of the P2Y2R by its endogenous agonists ATP or UTP can stimulate Rho- and Rac-dependent cytoskeletal rearrangements and cell migration (Bagchi et al., 2005; Wang et al., 2005). Additionally, a binding site in the intracellular C-terminus allows P2Y2R interaction with the actin-binding protein filamin A that also regulates P2Y2R-mediated cell migration (Yu et al., 2008). Also within the C-terminus of the P2Y2R, two Src-homology-3 (SH3) binding sites directly interact with and activate the tyrosine kinase Src, enabling P2Y2R-mediated transactivation of growth factor receptors, including the EGFR (Liu et al., 2004). Lastly, the P2Y2R has been shown to activate the α-secretases, ADAM10 and ADAM17, which promotes the release of membrane-bound receptor ligands, including growth factors (Ratchford et al., 2010; El-Sayed et al., 2014), and the cleavage of APP to form non-amyloidogenic sAPPα peptide (Camden et al., 2005; Kong et al., 2009).
Within the CNS, it appears that P2Y2R activation is most relevant in regulating neuroprotective responses during neuroinflammation, as seen in AD. In vitro, P2Y2R expression in rat primary cortical neurons is upregulated in response to IL-1β (Kong et al., 2009), a well-described inflammatory cytokine whose levels are elevated in the AD brain (Shaftel et al., 2008), whereupon activation of the P2Y2R by UTP stimulates neurite extension and non-amyloidogenic APP processing (Camden et al., 2005; Pooler et al., 2005; Kong et al., 2009; Peterson et al., 2013). In mouse primary microglial cells, P2Y2R activation by UTP induces both the uptake and degradation of Aβ1-42 (Kim et al., 2012). These findings suggest multiple mechanisms for P2Y2R-mediated neuroprotection, making it an attractive novel target for the treatment of AD. Consistent with these in vitro observations, global knockdown of P2Y2R expression in the TgCRND8 mouse model of AD (i.e., TgCRND8 × P2Y2R+/− mice) was shown to significantly increase AD-like pathology in the brain, including elevated Aβ1-42 levels, Aβ plaque deposition, leukocyte infiltration and neurological deficits leading to premature death at ~10 weeks of age (Ajit et al., 2014). These observations likely reflect the loss of P2Y2R-mediated neuroprotective responses in both neurons and microglia investigated in vitro, as described above. Furthermore, P2Y2R expression, but not P2Y4 or P2Y6 receptor expression, is selectively decreased in the parietal cortex of AD patients, as compared to age-matched non-AD controls, which correlates with synapse degeneration (Lai et al., 2008), consistent with the hypothesis that loss of P2Y2R-mediated responses contributes to disease pathology in AD.
Similar to the P2Y2R, the P2Y4 receptor (P2Y4R) has been shown to play a role in the uptake of Aβ1-42 by rat microglial cells through a mechanism dependent on autocrine ATP signaling (Li et al., 2013). Application of Aβ 1-42 to primary mouse microglial cells has been shown to induce the release of ATP, which enhances cell migration and Aβ uptake that were inhibited by the addition of apyrase or P2Y2R knockout (Kim et al., 2012). Autocrine ATP-induced Aβ uptake by microglia also was shown to be reduced following knockdown of the P2Y4R in rats (Li et al., 2013), suggesting the presence of redundant regulators of ATP-induced Aβ uptake (i.e., P2Y2R and P2Y4R), which should be exploited therapeutically to increase the uptake and degradation of Aβ1-42 in AD.
In addition to their role in Aβ clearance, microglial cells are the primary cells responsible for phagocytosis and clearance of dead neurons and debris in the AD brain that are necessary to maintain synapses and reduce neuroinflammation (Noda and Suzumura, 2012). The microglial P2Y6 receptor (P2Y6R), whose natural ligand is UDP, has been shown to be a primary regulator of the phagocytosis of damaged neurons in a kainic acid (KA)-induced model of neuronal cell death (Koizumi et al., 2007). Intraperitoneal application of KA, which has been shown to induce excitatory cell death, led to increased neuronal death in the rat hippocampus as well as increased P2Y6R expression in microglial cells surrounding dying neurons. Furthermore, direct injection of KA into the hippocampus increased extracellular uridine nucleotide levels and phagocytic activity of microglia, which were attenuated by the P2Y6R antagonist MRS2578 (Koizumi et al., 2007). However, targeting the P2Y6R in the treatment of AD may be complicated by the recent finding that activated microglia can phagocytose viable neurons, a process termed phagoptosis (Brown and Neher, 2014). Exposure of neuron-microglial cell cocultures to sub-neurotoxic levels of Aβ1-42 (10–250 nM) has been shown to result in significant neuronal death, presumably through phagoptosis (Neniskyte et al., 2011). Further studies of this phenomenon demonstrated that inhibition of the P2Y6R using MRS2578 significantly reduced phagoptosis in Aβ1-42-treated neuron-microglial cell cocultures and that the subsequent addition of UDP was sufficient to restore microglial cell-mediated neuronal death (Neher et al., 2014). Whether P2Y6R agonism or antagonism represents a viable therapeutic approach in AD remains to be explored.
Lastly, microglial P2Y12 and neuronal P2Y13 receptors have been shown to activate neuroprotective responses by inducing microglial migration/filopodial extension towards injurious stimuli and protecting neurons from ROS-induced neurotoxicity, respectively. Microglia have been shown to extend filopodia and migrate towards focal neuron damage and P2Y12 receptor (P2Y12R) deficiency or inhibition abrogates ADP-induced chemotaxis and process extension, suggesting that microglial P2Y12R activation may modulate neurodegeneration in the AD brain (Haynes et al., 2006; Koizumi et al., 2013; Webster et al., 2013). Activation of the P2Y13 receptor (P2Y13R) in rat primary cerebellar neurons using the selective agonist 2-methylthio-ADP has been shown to attenuate ROS-induced neuronal death through activation of Nrf2 (NF-E2-related factor-2) and downstream antioxidant response elements, suggesting that neuronal P2Y13Rs could be targeted to reduce ROS-induced neurotoxicity during neuroinflammation (Espada et al., 2010).
4. Conclusion
The studies presented in this review highlight the research that has been done investigating the therapeutic potential of targeting purinergic receptors to treat Alzheimer’s disease and the findings are summarized in Figure 1. Previous studies have identified the A2AR, P2X4R and P2X7R as novel therapeutic drug targets whose antagonism or inhibition can reduce neurodegeneration. Targeting the P2Y2R also may have therapeutic value, particularly in the early stages of disease where neuroinflammatory responses may play a role in tissue repair, as P2Y2R activation in both neurons and microglia has been shown to promote neuroprotective responses. The P2Y1R also may be relevant to AD pharmacotherapies, since P2Y1R activation in neural stem cells enhances neurogenesis, whereas P2Y1R activation in astrocytes may contribute to neurodegeneration. The P2Y6R in microglial cells has been shown to regulate phagocytosis of neuronal debris that should reduce neuroinflammation in the AD brain, but the same pathway can promote phagocytosis of viable neurons (i.e., phagoptosis) that could contribute to neurodegeneration and, therefore, further studies are needed to evaluate the therapeutic potential of several P2YRs. Of major importance will be the continued development of potent and highly-specific P1 and P2 receptor agonists and antagonists that can be utilized in studies with animal models of AD and ultimately in human clinical trials. Nonetheless, current research strongly supports the relevance of P1 and P2 receptors as important targets in the treatment of AD and other neurodegenerative diseases, and clinical interest in these receptors is apparent with the announcement of a Phase II clinical trial to investigate the therapeutic use of intravenous ATP infusion to improve cerebral metabolism and mental state in AD patients (ClinicalTrials.gov: NCT02279511).
Figure 1. Purinergic receptors as novel drug targets to treat Alzheimer’s disease.
Numerous studies have utilized selective agonists and antagonists for purinergic receptors in vitro and in rodent models of Alzheimer’s disease to demonstrate their therapeutic potential to treat AD. Neuronal P1 receptors (A1R and A2AR) have primarily been targeted to promote non-amyloidogenic amyloid precursor protein (APP) processing and prevent amyloid β-induced neurotoxicity. P2X and P2Y receptors have been targeted in neurons, microglia and astrocytes to enhance numerous therapeutic responses, including reducing neuroinflammation and neurotoxicity, enhancing non-amyloidogenic APP processing, promoting Aβ uptake and degradation and stimulating neurogenesis. This figure summarizes only the findings presented within this review paper and is not intended to be comprehensive.
Highlights.
Alzheimer’s disease is a neurodegenerative disorder resulting in loss of cognition
Neuroinflammation is a major component of Alzheimer’s disease
Purinergic receptors can regulate neurodegeneration and neuroinflammation
Purinergic receptors are novel drug targets to treat Alzheimer’s disease
Acknowledgements
This work was supported by NIH grant DE023342, BrightFocus Foundation grant A2013171S and a grant from the Spinal Cord Injuries Research Program from the University of Missouri.
Abbreviations
- Aβ
amyloid beta
- ADAM10/17
a disintegrin and metalloproteinase domain 10/17
- AD
Alzheimer’s disease
- APP
amyloid precursor protein
- APPswe
mutant APP bearing the Swedish mutation
- BBG
Brilliant Blue G
- ICV
intracerebroventricular
- IP3
inositol 1, 4, 5-trisphosphate
- NLRP3
nucleotide-binding domain and leucine-rich repeat containing family, pyrin domain, containing 3
- P2XR
P2X receptors
- P2YR
P2Y receptors
- PLC
phospholipase C
- PKC
protein kinase C
- PS1
presenilin 1
- PS1dE9
mutant presenilin 1 bearing the deltaE9 mutation
- sAPPα
soluble amyloid precursor protein α
Footnotes
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
References
- Abbracchio MP, Burnstock G, Boeynaems JM, Barnard EA, Boyer JL, Kennedy C, Knight GE, Fumagalli M, Gachet C, Jacobson KA, Weisman GA. International Union of Pharmacology LVIII: update on the P2Y G protein-coupled nucleotide receptors: from molecular mechanisms and pathophysiology to therapy. Pharmacol Rev. 2006;58:281–341. doi: 10.1124/pr.58.3.3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Abbracchio MP, Ceruti S. P1 receptors and cytokine secretion. Purinergic Signal. 2007;3:13–25. doi: 10.1007/s11302-006-9033-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Agteresch HJ, Dagnelie PC, van der Gaast A, Stijnen T, Wilson JH. Randomized clinical trial of adenosine 5'-triphosphate in patients with advanced non-small-cell lung cancer. J Natl Cancer Inst. 2000;92:321–328. doi: 10.1093/jnci/92.4.321. [DOI] [PubMed] [Google Scholar]
- Aisen PS, Davis KL, Berg JD, Schafer K, Campbell K, Thomas RG, Weiner MF, Farlow MR, Sano M, Grundman M, Thal LJ. A randomized controlled trial of prednisone in Alzheimer’s disease. Alzheimer’s Disease Cooperative Study. Neurology. 2000;54:588–593. doi: 10.1212/wnl.54.3.588. [DOI] [PubMed] [Google Scholar]
- Aisen PS, Schafer KA, Grundman M, Pfeiffer E, Sano M, Davis KL, Farlow MR, Jin S, Thomas RG, Thal LJ. Effects of rofecoxib or naproxen vs placebo on Alzheimer disease progression: a randomized controlled trial. JAMA. 2003;289:2819–2826. doi: 10.1001/jama.289.21.2819. [DOI] [PubMed] [Google Scholar]
- Ajit D, Woods LT, Camden JM, Thebeau CN, El-Sayed FG, Greeson GW, Erb L, Petris MJ, Miller DC, Sun GY, Weisman GA. Loss of P2Y2 nucleotide receptors enhances early pathology in the TgCRND8 mouse model of Alzheimer's disease. Mol Neurobiol. 2014;49:1031–1042. doi: 10.1007/s12035-013-8577-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Albasanz JL, Perez S, Barrachina M, Ferrer I, Martin M. Up-regulation of adenosine receptors in the frontal cortex in Alzheimer's disease. Brain Pathol. 2008;18:211–219. doi: 10.1111/j.1750-3639.2007.00112.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Angulo E, Casado V, Mallol J, Canela EI, Vinals F, Ferrer I, Lluis C, Franco R. A1 adenosine receptors accumulate in neurodegenerative structures in Alzheimer disease and mediate both amyloid precursor protein processing and tau phosphorylation and translocation. Brain Pathol. 2003;13:440–451. doi: 10.1111/j.1750-3639.2003.tb00475.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Apolloni S, Montilli C, Finocchi P, Amadio S. Membrane compartments and purinergic signalling: P2X receptors in neurodegenerative and neuroinflammatory events. The FEBS journal. 2009;276:354–364. doi: 10.1111/j.1742-4658.2008.06796.x. [DOI] [PubMed] [Google Scholar]
- Arendash GW, Schleif W, Rezai-Zadeh K, Jackson EK, Zacharia LC, Cracchiolo JR, Shippy D, Tan J. Caffeine protects Alzheimer's mice against cognitive impairment and reduces brain β-amyloid production. Neuroscience. 2006;142:941–952. doi: 10.1016/j.neuroscience.2006.07.021. [DOI] [PubMed] [Google Scholar]
- Arulkumaran N, Unwin RJ, Tam FW. A potential therapeutic role for P2X7 receptor (P2X7R) antagonists in the treatment of inflammatory diseases. Expert Opin Investig Drugs. 2011;20:897–915. doi: 10.1517/13543784.2011.578068. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ascherio A, Zhang SM, Hernan MA, Kawachi I, Colditz GA, Speizer FE, Willett WC. Prospective study of caffeine consumption and risk of Parkinson's disease in men and women. Ann Neurol. 2001;50:56–63. doi: 10.1002/ana.1052. [DOI] [PubMed] [Google Scholar]
- Bagchi S, Liao Z, Gonzalez FA, Chorna NE, Seye CI, Weisman GA, Erb L. The P2Y2 nucleotide receptor interacts with αv integrins to activate Go and induce cell migration. J Biol Chem. 2005;280:39050–39057. doi: 10.1074/jbc.M504819200. [DOI] [PubMed] [Google Scholar]
- Beijer S, Hupperets PS, van den Borne BE, Wijckmans NE, Spreeuwenberg C, van den Brandt PA, Dagnelie PC. Randomized clinical trial on the effects of adenosine 5'-triphosphate infusions on quality of life, functional status, and fatigue in preterminal cancer patients. J Pain Symptom Manage. 2010;40:520–530. doi: 10.1016/j.jpainsymman.2010.01.023. [DOI] [PubMed] [Google Scholar]
- Boeynaems JM, van Giezen H, Savi P, Herbert JM. P2Y receptor antagonists in thrombosis. Curr Opin Investig Drugs. 2005;6:275–282. [PubMed] [Google Scholar]
- Bours MJ, Dagnelie PC, Giuliani AL, Wesselius A, Di Virgilio F. P2 receptors and extracellular ATP: a novel homeostatic pathway in inflammation. Front Biosci (Schol Ed) 2011;3:1443–1456. doi: 10.2741/235. [DOI] [PubMed] [Google Scholar]
- Braak H, Braak E. Evolution of the neuropathology of Alzheimer's disease. Acta Neurol Scand. 1996;(Suppl 165):3–12. doi: 10.1111/j.1600-0404.1996.tb05866.x. [DOI] [PubMed] [Google Scholar]
- Brinton RD, Wang JM. Therapeutic potential of neurogenesis for prevention and recovery from Alzheimer's disease: allopregnanolone as a proof of concept neurogenic agent. Curr Alzheimer Res. 2006;3:185–190. doi: 10.2174/156720506777632817. [DOI] [PubMed] [Google Scholar]
- Brown GC, Neher JJ. Microglial phagocytosis of live neurons. Nat Rev Neurosci. 2014;15:209–216. doi: 10.1038/nrn3710. [DOI] [PubMed] [Google Scholar]
- Burnstock G. Purinergic signalling. Br J Pharmacol. 2006;147(Suppl 1):S172–S181. doi: 10.1038/sj.bjp.0706429. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Burnstock G. Purinergic signalling and disorders of the central nervous system. Nat Rev. Drug Discov. 2008;7:575–590. doi: 10.1038/nrd2605. [DOI] [PubMed] [Google Scholar]
- Burnstock G. Control of vascular tone by purines and pyrimidines. Br J Pharmacol. 2010;161:527–529. doi: 10.1111/j.1476-5381.2010.00937.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Burnstock G. An introduction to the roles of purinergic signalling in neurodegeneration, neuroprotection and neuroregeneration. Neuropharmacology. 2015 doi: 10.1016/j.neuropharm.2015.05.031. [Epub ahead of print]. [DOI] [PubMed] [Google Scholar]
- Burnstock G, Dumsday B, Smythe A. Atropine resistant excitation of the urinary bladder: the possibility of transmission via nerves releasing a purine nucleotide. Br J Pharmacol. 1972;44:451–461. doi: 10.1111/j.1476-5381.1972.tb07283.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Burnstock G, Krugel U, Abbracchio MP, Illes P. Purinergic signalling: from normal behaviour to pathological brain function. Prog Neurobiol. 2011;95:229–274. doi: 10.1016/j.pneurobio.2011.08.006. [DOI] [PubMed] [Google Scholar]
- Burnstock G, Verkhratsky A. Long-term (trophic) purinergic signalling: purinoceptors control cell proliferation, differentiation and death. Cell Death Dis. 2010;1:e9. doi: 10.1038/cddis.2009.11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Butterfield DA, Boyd-Kimball D. Amyloid β-peptide1-42 contributes to the oxidative stress and neurodegeneration found in Alzheimer disease brain. Brain Pathol. 2004;14:426–432. doi: 10.1111/j.1750-3639.2004.tb00087.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Camden JM, Schrader AM, Camden RE, Gonzalez FA, Erb L, Seye CI, Weisman GA. P2Y2 nucleotide receptors enhance α-secretase-dependent amyloid precursor protein processing. J Biol Chem. 2005;280:18696–18702. doi: 10.1074/jbc.M500219200. [DOI] [PubMed] [Google Scholar]
- Campion D, Dumanchin C, Hannequin D, Dubois B, Belliard S, Puel M, Thomas-Anterion C, Michon A, Martin C, Charbonnier F, Raux G, Camuzat A, Penet C, Mesnage V, Martinez M, Clerget-Darpoux F, Brice A, Frebourg T. Early-onset autosomal dominant Alzheimer disease: prevalence, genetic heterogeneity, and mutation spectrum. Am J Hum Genet. 1999;65:664–670. doi: 10.1086/302553. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Canas PM, Porciuncula LO, Cunha GM, Silva CG, Machado NJ, Oliveira JM, Oliveira CR, Cunha RA. Adenosine A2A receptor blockade prevents synaptotoxicity and memory dysfunction caused by β-amyloid peptides via p38 mitogen-activated protein kinase pathway. J Neurosci. 2009;29:14741–14751. doi: 10.1523/JNEUROSCI.3728-09.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cao C, Cirrito JR, Lin X, Wang L, Verges DK, Dickson A, Mamcarz M, Zhang C, Mori T, Arendash GW, Holtzman DM, Potter H. Caffeine suppresses amyloid-β levels in plasma and brain of Alzheimer's disease transgenic mice. J Alzheimers Dis. 2009;17:681–697. doi: 10.3233/JAD-2009-1071. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Carman AJ, Mills JH, Krenz A, Kim DG, Bynoe MS. Adenosine receptor signaling modulates permeability of the blood-brain barrier. J Neurosci. 2011;31:13272–13280. doi: 10.1523/JNEUROSCI.3337-11.2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cauwels A, Rogge E, Vandendriessche B, Shiva S, Brouckaert P. Extracellular ATP drives systemic inflammation, tissue damage and mortality. Cell Death Dis. 2014;5:e1102. doi: 10.1038/cddis.2014.70. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chakfe Y, Seguin R, Antel JP, Morissette C, Malo D, Henderson D, Seguela P. ADP and AMP induce interleukin-1β release from microglial cells through activation of ATP-primed P2X7 receptor channels. J Neurosci. 2002;22:3061–3069. doi: 10.1523/JNEUROSCI.22-08-03061.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chen GJ, Harvey BK, Shen H, Chou J, Victor A, Wang Y. Activation of adenosine A3 receptors reduces ischemic brain injury in rodents. J Neurosci Res. 2006;84:1848–1855. doi: 10.1002/jnr.21071. [DOI] [PubMed] [Google Scholar]
- Chen L, Brosnan CF. Exacerbation of experimental autoimmune encephalomyelitis in P2X7R−/− mice: evidence for loss of apoptotic activity in lymphocytes. J Immunol. 2006;176:3115–3126. doi: 10.4049/jimmunol.176.5.3115. [DOI] [PubMed] [Google Scholar]
- Chen X, Hu J, Jiang L, Xu S, Zheng B, Wang C, Zhang J, Wei X, Chang L, Wang Q. Brilliant Blue G improves cognition in an animal model of Alzheimer's disease and inhibits amyloid-β-induced loss of filopodia and dendrite spines in hippocampal neurons. Neuroscience. 2014;279:94–101. doi: 10.1016/j.neuroscience.2014.08.036. [DOI] [PubMed] [Google Scholar]
- Chizh BA, Illes P. P2X receptors and nociception. Pharmacol Rev. 2001;53:553–568. [PubMed] [Google Scholar]
- Chrovian CC, Rech JC, Bhattacharya A, Letavic MA. P2X7 antagonists as potential therapeutic agents for the treatment of CNS disorders. Prog Med Chem. 2014;53:65–100. doi: 10.1016/B978-0-444-63380-4.00002-0. [DOI] [PubMed] [Google Scholar]
- Chu K, Yin B, Wang J, Peng G, Liang H, Xu Z, Du Y, Fang M, Xia Q, Luo B. Inhibition of P2X7 receptor ameliorates transient global cerebral ischemia/reperfusion injury via modulating inflammatory responses in the rat hippocampus. J Neuroinflammation. 2012;9:69. doi: 10.1186/1742-2094-9-69. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cole GM, Frautschy SA. Mechanisms of action of non-steroidal anti-inflammatory drugs for the prevention of Alzheimer's disease. CNS Neurol Disord Drug Targets. 2010;9:140–148. doi: 10.2174/187152710791011991. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Collo G, Neidhart S, Kawashima E, Kosco-Vilbois M, North RA, Buell G. Tissue distribution of the P2X7 receptor. Neuropharmacology. 1997;36:1277–1283. doi: 10.1016/s0028-3908(97)00140-8. [DOI] [PubMed] [Google Scholar]
- Combs CK, Karlo JC, Kao SC, Landreth GE. β-Amyloid stimulation of microglia and monocytes results in TNFα-dependent expression of inducible nitric oxide synthase and neuronal apoptosis. J Neurosci. 2001;21:1179–1188. doi: 10.1523/JNEUROSCI.21-04-01179.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Csoka B, Nemeth ZH, Toro G, Idzko M, Zech A, Koscso B, Spolarics Z, Antonioli L, Cseri K, Erdelyi K, Pacher P, Hasko G. Extracellular ATP protects against sepsis through macrophage P2X7 purinergic receptors by enhancing intracellular bacterial killing. FASEB J. 2015 doi: 10.1096/fj.15-272450. [Epub ahead of print]. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cummings JL. Alzheimer's disease. New Engl J Med. 2004;351:56–67. doi: 10.1056/NEJMra040223. [DOI] [PubMed] [Google Scholar]
- D'Andrea MR, Nagele RG, Wang HY, Peterson PA, Lee DH. Evidence that neurones accumulating amyloid can undergo lysis to form amyloid plaques in Alzheimer’s disease. Histopathology. 2001;38:120–134. doi: 10.1046/j.1365-2559.2001.01082.x. [DOI] [PubMed] [Google Scholar]
- Dall'Igna OP, Fett P, Gomes MW, Souza DO, Cunha RA, Lara DR. Caffeine and adenosine A2A receptor antagonists prevent β-amyloid25-35-induced cognitive deficits in mice. Exp Neurol. 2007;203:241–245. doi: 10.1016/j.expneurol.2006.08.008. [DOI] [PubMed] [Google Scholar]
- Dall'Igna OP, Porciuncula LO, Souza DO, Cunha RA, Lara DR. Neuroprotection by caffeine and adenosine A2A receptor blockade of β-amyloid neurotoxicity. Br J Pharmacol. 2003;138:1207–1209. doi: 10.1038/sj.bjp.0705185. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Davalos D, Grutzendler J, Yang G, Kim JV, Zuo Y, Jung S, Littman DR, Dustin ML, Gan WB. ATP mediates rapid microglial response to local brain injury in vivo. Nat Neurosci. 2005;8:752–758. doi: 10.1038/nn1472. [DOI] [PubMed] [Google Scholar]
- Delekate A, Fuchtemeier M, Schumacher T, Ulbrich C, Foddis M, Petzold GC. Metabotropic P2Y1 receptor signalling mediates astrocytic hyperactivity in vivo in an Alzheimer's disease mouse model. Nat Comm. 2014;5:5422. doi: 10.1038/ncomms6422. [DOI] [PubMed] [Google Scholar]
- Deng W, Aimone JB, Gage FH. New neurons and new memories: how does adult hippocampal neurogenesis affect learning and memory? Nat Rev Neurosci. 2010;11:339–350. doi: 10.1038/nrn2822. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Di Virgilio F. ATP as a death factor. Biofactors. 1998;8:301–303. doi: 10.1002/biof.5520080318. [DOI] [PubMed] [Google Scholar]
- Di Virgilio F. Liaisons dangereuses: P2X7 and the inflammasome. Trends Pharmacol Sci. 2007;28:465–472. doi: 10.1016/j.tips.2007.07.002. [DOI] [PubMed] [Google Scholar]
- Diaz-Cabiale Z, Hurd Y, Guidolin D, Finnman UB, Zoli M, Agnati LF, Vanderhaeghen JJ, Fuxe K, Ferre S. Adenosine A2A agonist CGS 21680 decreases the affinity of dopamine D2 receptors for dopamine in human striatum. Neuroreport. 2001;12:1831–1834. doi: 10.1097/00001756-200107030-00014. [DOI] [PubMed] [Google Scholar]
- Diaz-Hernandez JI, Gomez-Villafuertes R, Leon-Otegui M, Hontecillas-Prieto L, Del Puerto A, Trejo JL, Lucas JJ, Garrido JJ, Gualix J, Miras-Portugal MT, Diaz-Hernandez M. In vivo P2X7 inhibition reduces amyloid plaques in Alzheimer's disease through GSK3β and secretases. Neurobiol Aging. 2012;33:1816–1828. doi: 10.1016/j.neurobiolaging.2011.09.040. [DOI] [PubMed] [Google Scholar]
- Diaz-Hernandez M, Diez-Zaera M, Sanchez-Nogueiro J, Gomez-Villafuertes R, Canals JM, Alberch J, Miras-Portugal MT, Lucas JJ. Altered P2X7-receptor level and function in mouse models of Huntington's disease and therapeutic efficacy of antagonist administration. FASEB J. 2009;23:1893–1906. doi: 10.1096/fj.08-122275. [DOI] [PubMed] [Google Scholar]
- El-Sayed FG, Camden JM, Woods LT, Khalafalla MG, Petris MJ, Erb L, Weisman GA. P2Y2 nucleotide receptor activation enhances the aggregation and self-organization of dispersed salivary epithelial cells. Am J Physiol Cell Physiol. 2014;307:C83–C96. doi: 10.1152/ajpcell.00380.2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Erb L, Garrad R, Wang Y, Quinn T, Turner JT, Weisman GA. Site-directed mutagenesis of P2U purinoceptors. Positively charged amino acids in transmembrane helices 6 and 7 affect agonist potency and specificity. J Biol Chem. 1995;270:4185–4188. doi: 10.1074/jbc.270.9.4185. [DOI] [PubMed] [Google Scholar]
- Erb L, Liu J, Ockerhausen J, Kong Q, Garrad RC, Griffin K, Neal C, Krugh B, Santiago-Perez LI, Gonzalez FA, Gresham HD, Turner JT, Weisman GA. An RGD sequence in the P2Y2 receptor interacts with αvβ3 integrins and is required for Go-mediated signal transduction. J Cell Biol. 2001;153:491–501. doi: 10.1083/jcb.153.3.491. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Erb L, Lustig KD, Ahmed AH, Gonzalez FA, Weisman GA. Covalent incorporation of 3'-O-(4-benzoyl)benzoyl-ATP into a P2 purinoceptor in transformed mouse fibroblasts. J Biol Chem. 1990;265:7424–7431. [PubMed] [Google Scholar]
- Eriksson PS, Perfilieva E, Bjork-Eriksson T, Alborn AM, Nordborg C, Peterson DA, Gage FH. Neurogenesis in the adult human hippocampus. Nat Med. 1998;4:1313–1317. doi: 10.1038/3305. [DOI] [PubMed] [Google Scholar]
- Eskelinen MH, Kivipelto M. Caffeine as a protective factor in dementia and Alzheimer’s disease. J Alzheimers Dis. 2010;1(20 Suppl):S167–S174. doi: 10.3233/JAD-2010-1404. [DOI] [PubMed] [Google Scholar]
- Espada S, Ortega F, Molina-Jijon E, Rojo AI, Perez-Sen R, Pedraza-Chaverri J, Miras-Portugal MT, Cuadrado A. The purinergic P2Y13 receptor activates the Nrf2/HO-1 axis and protects against oxidative stress-induced neuronal death. Free Radic Biol Med. 2010;49:416–426. doi: 10.1016/j.freeradbiomed.2010.04.031. [DOI] [PubMed] [Google Scholar]
- Fields RD, Burnstock G. Purinergic signalling in neuron-glia interactions. Nat Rev Neurosci. 2006;7:423–436. doi: 10.1038/nrn1928. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Flores RV, Hernandez-Perez MG, Aquino E, Garrad RC, Weisman GA, Gonzalez FA. Agonist-induced phosphorylation and desensitization of the P2Y2 nucleotide receptor. Mol Cell Biochem. 2005;280:35–45. doi: 10.1007/s11010-005-8050-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ford AP. In pursuit of P2X3 antagonists: novel therapeutics for chronic pain and afferent sensitization. Purinergic Signal. 2012;8:3–26. doi: 10.1007/s11302-011-9271-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Frautschy SA, Yang F, Irrizarry M, Hyman B, Saido TC, Hsiao K, Cole GM. Microglial response to amyloid plaques in APPsw transgenic mice. Am J Pathol. 1998;152:307–317. [PMC free article] [PubMed] [Google Scholar]
- Fredholm BB, AP IJ, Jacobson KA, Klotz KN, Linden J. International Union of Pharmacology. XXV. Nomenclature and classification of adenosine receptors. Pharmacol Rev. 2001;53:527–552. [PMC free article] [PubMed] [Google Scholar]
- Fredholm BB, Battig K, Holmen J, Nehlig A, Zvartau EE. Actions of caffeine in the brain with special reference to factors that contribute to its widespread use. Pharmacol Rev. 1999;51:83–133. [PubMed] [Google Scholar]
- Fuster-Matanzo A, Llorens-Martin M, Hernandez F, Avila J. Role of neuroinflammation in adult neurogenesis and Alzheimer disease: therapeutic approaches. Mediators Inflamm. 2013:260925. doi: 10.1155/2013/260925. Epub 2013 Apr 3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gandelman M, Levy M, Cassina P, Barbeito L, Beckman JS. P2X7 receptor-induced death of motor neurons by a peroxynitrite/FAS-dependent pathway. J Neurochem. 2013;126:382–388. doi: 10.1111/jnc.12286. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gendaszewska-Darmach E, Kucharska M. Nucleotide receptors as targets in the pharmacological enhancement of dermal wound healing. Purinergic Signal. 2011;7:193–206. doi: 10.1007/s11302-011-9233-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Genin E, Hannequin D, Wallon D, Sleegers K, Hiltunen M, Combarros O, Bullido MJ, Engelborghs S, De Deyn P, Berr C, Pasquier F, Dubois B, Tognoni G, Fievet N, Brouwers N, Bettens K, Arosio B, Coto E, Del Zompo M, Mateo I, Epelbaum J, Frank-Garcia A, Helisalmi S, Porcellini E, Pilotto A, Forti P, Ferri R, Scarpini E, Siciliano G, Solfrizzi V, Sorbi S, Spalletta G, Valdivieso F, Vepsalainen S, Alvarez V, Bosco P, Mancuso M, Panza F, Nacmias B, Bossu P, Hanon O, Piccardi P, Annoni G, Seripa D, Galimberti D, Licastro F, Soininen H, Dartigues JF, Kamboh MI, Van Broeckhoven C, Lambert JC, Amouyel P, Campion D. APOE and Alzheimer disease: a major gene with semi-dominant inheritance. Mol Psychiatry. 2011;16:903–907. doi: 10.1038/mp.2011.52. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gilman S, Koller M, Black RS, Jenkins L, Griffith SG, Fox NC, Eisner L, Kirby L, Rovira MB, Forette F, Orgogozo JM. Clinical effects of Aβ immunization (AN1792) in patients with AD in an interrupted trial. Neurology. 2005;64:1553–1562. doi: 10.1212/01.WNL.0000159740.16984.3C. [DOI] [PubMed] [Google Scholar]
- Green RC, Schneider LS, Amato DA, Beelen AP, Wilcock G, Swabb EA, Zavitz KH. Effect of tarenflurbil on cognitive decline and activities of daily living in patients with mild Alzheimer disease: a randomized controlled trial. JAMA. 2009;302:2557–2564. doi: 10.1001/jama.2009.1866. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Griffin WS, Stanley LC, Ling C, White L, MacLeod V, Perrot LJ, White CL, 3rd, Araoz C. Brain interleukin 1 and S-100 immunoreactivity are elevated in Down syndrome and Alzheimer disease. Proc Natl Acad Sci USA. 1989;86:7611–7615. doi: 10.1073/pnas.86.19.7611. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Guthrie PB, Knappenberger J, Segal M, Bennett MV, Charles AC, Kater SB. ATP released from astrocytes mediates glial calcium waves. J Neurosci. 1999;19:520–528. doi: 10.1523/JNEUROSCI.19-02-00520.1999. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hardy J, Selkoe DJ. The amyloid hypothesis of Alzheimer’s disease: progress and problems on the road to therapeutics. Science. 2002;297:353–356. doi: 10.1126/science.1072994. [DOI] [PubMed] [Google Scholar]
- Hauser RA, Cantillon M, Pourcher E, Micheli F, Mok V, Onofrj M, Huyck S, Wolski K. Preladenant in patients with Parkinson’s disease and motor fluctuations: a phase 2, double-blind, randomised trial. Lancet Neurol. 2011;10:221–229. doi: 10.1016/S1474-4422(11)70012-6. [DOI] [PubMed] [Google Scholar]
- Hauser RA, Olanow CW, Kieburtz KD, Pourcher E, Docu-Axelerad A, Lew M, Kozyolkin O, Neale A, Resburg C, Meya U, Kenney C, Bandak S. Tozadenant (SYN115) in patients with Parkinson’s disease who have motor fluctuations on levodopa: a phase 2b, double-blind, randomised trial. Lancet Neurol. 2014;13:767–776. doi: 10.1016/S1474-4422(14)70148-6. [DOI] [PubMed] [Google Scholar]
- Haynes SE, Hollopeter G, Yang G, Kurpius D, Dailey ME, Gan WB, Julius D. The P2Y12 receptor regulates microglial activation by extracellular nucleotides. Nat Neurosci. 2006;9:1512–1519. doi: 10.1038/nn1805. [DOI] [PubMed] [Google Scholar]
- Hebert LE, Weuve J, Scherr PA, Evans DA. Alzheimer disease in the United States (2010–2050) estimated using the 2010 census. Neurology. 2013;80:1778–1783. doi: 10.1212/WNL.0b013e31828726f5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Heppner FL, Ransohoff RM, Becher B. Immune attack: the role of inflammation in Alzheimer disease. Nat Rev Neurosci. 2015;16:358–372. doi: 10.1038/nrn3880. [DOI] [PubMed] [Google Scholar]
- Hide I, Tanaka M, Inoue A, Nakajima K, Kohsaka S, Inoue K, Nakata Y. Extracellular ATP triggers tumor necrosis factor-α release from rat microglia. J Neurochem. 2000;75:965–972. doi: 10.1046/j.1471-4159.2000.0750965.x. [DOI] [PubMed] [Google Scholar]
- Huang Y, Mucke L. Alzheimer mechanisms and therapeutic strategies. Cell. 2012;148:1204–1222. doi: 10.1016/j.cell.2012.02.040. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hurtado DE, Molina-Porcel L, Iba M, Aboagye AK, Paul SM, Trojanowski JQ, Lee VMY. Aβ Accelerates the Spatiotemporal Progression of Tau Pathology and Augments Tau Amyloidosis in an Alzheimer Mouse Model. Amer J Pathol. 2010;177:1977–1988. doi: 10.2353/ajpath.2010.100346. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Idzko M, Ferrari D, Eltzschig HK. Nucleotide signalling during inflammation. Nature. 2014;509:310–317. doi: 10.1038/nature13085. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Illes P, Verkhratsky A, Burnstock G, Franke H. P2X receptors and their roles in astroglia in the central and peripheral nervous system. Neuroscientist. 2012;18:422–438. doi: 10.1177/1073858411418524. [DOI] [PubMed] [Google Scholar]
- Jansen KL, Faull RL, Dragunow M, Synek BL. Alzheimer’s disease: changes in hippocampal N-methyl-D-aspartate, quisqualate, neurotensin, adenosine, benzodiazepine, serotonin and opioid receptors--an autoradiographic study. Neuroscience. 1990;39:613–627. doi: 10.1016/0306-4522(90)90246-z. [DOI] [PubMed] [Google Scholar]
- Jarvis MF, Burgard EC, McGaraughty S, Honore P, Lynch K, Brennan TJ, Subieta A, Van Biesen T, Cartmell J, Bianchi B, Niforatos W, Kage K, Yu H, Mikusa J, Wismer CT, Zhu CZ, Chu K, Lee CH, Stewart AO, Polakowski J, Cox BF, Kowaluk E, Williams M, Sullivan J, Faltynek C. A-317491, a novel potent and selective non-nucleotide antagonist of P2X3 and P2X2/3 receptors, reduces chronic inflammatory and neuropathic pain in the rat. Proc Natl Acad Sci USA. 2002;99:17179–17184. doi: 10.1073/pnas.252537299. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jiang LH, Mackenzie AB, North RA, Surprenant A. Brilliant blue G selectively blocks ATP-gated rat P2X7 receptors. Mol Pharmacol. 2000;58:82–88. [PubMed] [Google Scholar]
- Kalaria RN, Sromek S, Wilcox BJ, Unnerstall JR. Hippocampal adenosine A1 receptors are decreased in Alzheimer’s disease. Neurosci Lett. 1990;118:257–260. doi: 10.1016/0304-3940(90)90641-l. [DOI] [PubMed] [Google Scholar]
- Kanjhan R, Housley GD, Burton LD, Christie DL, Kippenberger A, Thorne PR, Luo L, Ryan AF. Distribution of the P2X2 receptor subunit of the ATP-gated ion channels in the rat central nervous system. J Comp Neurol. 1999;407:11–32. [PubMed] [Google Scholar]
- Khakh BS, North RA. P2X receptors as cell-surface ATP sensors in health and disease. Nature. 2006;442:527–532. doi: 10.1038/nature04886. [DOI] [PubMed] [Google Scholar]
- Kim HJ, Ajit D, Peterson TS, Wang Y, Camden JM, Gibson Wood W, Sun GY, Erb L, Petris M, Weisman GA. Nucleotides released from Aβ1-42-treated microglial cells increase cell migration and Aβ1-42 uptake through P2Y2 receptor activation. J Neurochem. 2012;121:228–238. doi: 10.1111/j.1471-4159.2012.07700.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kim SY, Moon JH, Lee HG, Kim SU, Lee YB. ATP released from β-amyloid-stimulated microglia induces reactive oxygen species production in an autocrine fashion. Exp Mol Med. 2007;39:820–827. doi: 10.1038/emm.2007.89. [DOI] [PubMed] [Google Scholar]
- Koizumi S, Ohsawa K, Inoue K, Kohsaka S. Purinergic receptors in microglia: functional modal shifts of microglia mediated by P2 and P1 receptors. Glia. 2013;61:47–54. doi: 10.1002/glia.22358. [DOI] [PubMed] [Google Scholar]
- Koizumi S, Shigemoto-Mogami Y, Nasu-Tada K, Shinozaki Y, Ohsawa K, Tsuda M, Joshi BV, Jacobson KA, Kohsaka S, Inoue K. UDP acting at P2Y6 receptors is a mediator of microglial phagocytosis. Nature. 2007;446:1091–1095. doi: 10.1038/nature05704. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Koles L, Furst S, Illes P. Purine ionotropic (P2X) receptors. Curr Pharm Des. 2007:2368–2384. doi: 10.2174/138161207781368747. [DOI] [PubMed] [Google Scholar]
- Kong Q, Peterson TS, Baker O, Stanley E, Camden J, Seye CI, Erb L, Simonyi A, Wood WG, Sun GY, Weisman GA. Interleukin-1β enhances nucleotide-induced and α-secretase-dependent amyloid precursor protein processing in rat primary cortical neurons via up-regulation of the P2Y2 receptor. J Neurochem. 2009;109:1300–1310. doi: 10.1111/j.1471-4159.2009.06048.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kong Q, Wang M, Liao Z, Camden JM, Yu S, Simonyi A, Sun GY, Gonzalez FA, Erb L, Seye CI, Weisman GA. P2X7 nucleotide receptors mediate caspase-8/9/3-dependent apoptosis in rat primary cortical neurons. Purinergic Signal. 2005;1:337–347. doi: 10.1007/s11302-005-7145-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kowall NW, Beal MF, Busciglio J, Duffy LK, Yankner BA. An in vivo model for the neurodegenerative effects of β-amyloid and protection by substance P. Proc Natl Acad Sci USA. 1991;88:7247–7251. doi: 10.1073/pnas.88.16.7247. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kuboyama K, Harada H, Tozaki-Saitoh H, Tsuda M, Ushijima K, Inoue K. Astrocytic P2Y1 receptor is involved in the regulation of cytokine/chemokine transcription and cerebral damage in a rat model of cerebral ischemia. J Cereb Blood Flow Metab. 2011;31:1930–1941. doi: 10.1038/jcbfm.2011.49. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kuchibhotla KV, Lattarulo CR, Hyman BT, Bacskai BJ. Synchronous hyperactivity and intercellular calcium waves in astrocytes in Alzheimer mice. Science. 2009;323:1211–1215. doi: 10.1126/science.1169096. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kuhn HG, Dickinson-Anson H, Gage FH. Neurogenesis in the dentate gyrus of the adult rat: age-related decrease of neuronal progenitor proliferation. J Neurosci. 1996;16:2027–2033. doi: 10.1523/JNEUROSCI.16-06-02027.1996. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lai MK, Tan MG, Kirvell S, Hobbs C, Lee J, Esiri MM, Chen CP, Francis PT. Selective loss of P2Y2 nucleotide receptor immunoreactivity is associated with Alzheimer’s disease neuropathology. J Neural Transm. 2008;115:1165–1172. doi: 10.1007/s00702-008-0067-y. [DOI] [PubMed] [Google Scholar]
- Lee HG, Won SM, Gwag BJ, Lee YB. Microglial P2X7 receptor expression is accompanied by neuronal damage in the cerebral cortex of the APPswe/PS1dE9 mouse model of Alzheimer’s disease. Exp Mol Med. 2011;43:7–14. doi: 10.3858/emm.2011.43.1.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lemere CA, Lopera F, Kosik KS, Lendon CL, Ossa J, Saido TC, Yamaguchi H, Ruiz A, Martinez A, Madrigal L, Hincapie L, Arango JC, Anthony DC, Koo EH, Goate AM, Selkoe DJ. The E280A presenilin 1 Alzheimer mutation produces increased Aβ 42 deposition and severe cerebellar pathology. Nat Med. 1996;2:1146–1150. doi: 10.1038/nm1096-1146. [DOI] [PubMed] [Google Scholar]
- LeWitt PA, Guttman M, Tetrud JW, Tuite PJ, Mori A, Chaikin P, Sussman NM. Adenosine A2A receptor antagonist istradefylline (KW-6002) reduces "off" time in Parkinson's disease: a double-blind, randomized, multicenter clinical trial (6002-US-005) Ann Neurol. 2008;63:295–302. doi: 10.1002/ana.21315. [DOI] [PubMed] [Google Scholar]
- Li HQ, Chen C, Dou Y, Wu HJ, Liu YJ, Lou HF, Zhang JM, Li XM, Wang H, Duan S. P2Y4 receptor-mediated pinocytosis contributes to amyloid β-induced self-uptake by microglia. Mol Cell Biol. 2013;33:4282–4293. doi: 10.1128/MCB.00544-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Liao Z, Seye CI, Weisman GA, Erb L. The P2Y2 nucleotide receptor requires interaction with αv integrins to access and activate G12. J Cell Sci. 2007;120:1654–1662. doi: 10.1242/jcs.03441. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lister MF, Sharkey J, Sawatzky DA, Hodgkiss JP, Davidson DJ, Rossi AG, Finlayson K. The role of the purinergic P2X7 receptor in inflammation. J Inflamm (Lond) 2007;4:5. doi: 10.1186/1476-9255-4-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Little JW, Ford A, Symons-Liguori AM, Chen Z, Janes K, Doyle T, Xie J, Luongo L, Tosh DK, Maione S, Bannister K, Dickenson AH, Vanderah TW, Porreca F, Jacobson KA, Salvemini D. Endogenous adenosine A3 receptor activation selectively alleviates persistent pain states. Brain. 2015;138:28–35. doi: 10.1093/brain/awu330. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Liu J, Liao Z, Camden J, Griffin KD, Garrad RC, Santiago-Perez LI, Gonzalez FA, Seye CI, Weisman GA, Erb L. Src homology 3 binding sites in the P2Y2 nucleotide receptor interact with Src and regulate activities of Src, proline-rich tyrosine kinase 2, and growth factor receptors. J Biol Chem. 2004;279:8212–8218. doi: 10.1074/jbc.M312230200. [DOI] [PubMed] [Google Scholar]
- Lucattelli M, Cicko S, Muller T, Lommatzsch M, De Cunto G, Cardini S, Sundas W, Grimm M, Zeiser R, Durk T, Zissel G, Sorichter S, Ferrari D, Di Virgilio F, Virchow JC, Lungarella G, Idzko M. P2X7 receptor signaling in the pathogenesis of smoke-induced lung inflammation and emphysema. Am J Respir Cell Mol Biol. 2011;44:423–429. doi: 10.1165/rcmb.2010-0038OC. [DOI] [PubMed] [Google Scholar]
- Lustig KD, Sportiello MG, Erb L, Weisman GA. A nucleotide receptor in vascular endothelial cells is specifically activated by the fully ionized forms of ATP and UTP. Biochem J. 1992;284(Pt 3):733–739. doi: 10.1042/bj2840733. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Maia L, de Mendonca A. Does caffeine intake protect from Alzheimer’s disease? Eur J Neurol Societies. 2002;9:377–382. doi: 10.1046/j.1468-1331.2002.00421.x. [DOI] [PubMed] [Google Scholar]
- Malin DH, Crothers MK, Lake JR, Goyarzu P, Plotner RE, Garcia SA, Spell SH, Tomsic BJ, Giordano T, Kowall NW. Hippocampal injections of amyloid β-peptide 1–40 impair subsequent one-trial/day reward learning. Neurobiol Learn Mem. 2001;76:125–137. doi: 10.1006/nlme.2000.3991. [DOI] [PubMed] [Google Scholar]
- Matute C, Torre I, Perez-Cerda F, Perez-Samartin A, Alberdi E, Etxebarria E, Arranz AM, Ravid R, Rodriguez-Antiguedad A, Sanchez-Gomez M, Domercq M. P2X7 receptor blockade prevents ATP excitotoxicity in oligodendrocytes and ameliorates experimental autoimmune encephalomyelitis. J Neurosci. 2007;27:9525–9533. doi: 10.1523/JNEUROSCI.0579-07.2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- McGaraughty S, Wismer CT, Zhu CZ, Mikusa J, Honore P, Chu KL, Lee CH, Faltynek CR, Jarvis MF. Effects of A-317491, a novel and selective P2X3/P2X2/3 receptor antagonist, on neuropathic, inflammatory and chemogenic nociception following intrathecal and intraplantar administration. Br J Pharmacol. 2003;140:1381–1388. doi: 10.1038/sj.bjp.0705574. [DOI] [PMC free article] [PubMed] [Google Scholar]
- McLarnon JG, Ryu JK, Walker DG, Choi HB. Upregulated expression of purinergic P2X7 receptor in Alzheimer disease and amyloid-β peptide-treated microglia and in peptide-injected rat hippocampus. J Neuropathol Exp Neurol. 2006;65:1090–1097. doi: 10.1097/01.jnen.0000240470.97295.d3. [DOI] [PubMed] [Google Scholar]
- Mishina M, Ishiwata K, Naganawa M, Kimura Y, Kitamura S, Suzuki M, Hashimoto M, Ishibashi K, Oda K, Sakata M, Hamamoto M, Kobayashi S, Katayama Y, Ishii K. Adenosine A2A receptors measured with [C]TMSX PET in the striata of Parkinson’s disease patients. PLoS One. 2011;6:e17338. doi: 10.1371/journal.pone.0017338. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mishra SK, Braun N, Shukla V, Fullgrabe M, Schomerus C, Korf HW, Gachet C, Ikehara Y, Sevigny J, Robson SC, Zimmermann H. Extracellular nucleotide signaling in adult neural stem cells: synergism with growth factor-mediated cellular proliferation. Development. 2006;133:675–684. doi: 10.1242/dev.02233. [DOI] [PubMed] [Google Scholar]
- Monif M, Reid CA, Powell KL, Smart ML, Williams DA. The P2X7 receptor drives microglial activation and proliferation: a trophic role for P2X7R pore. J Neurosci. 2009;29:3781–3791. doi: 10.1523/JNEUROSCI.5512-08.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Moore D, Iritani S, Chambers J, Emson P. Immunohistochemical localization of the P2Y1 purinergic receptor in Alzheimer’s disease. Neuroreport. 2000;11:3799–3803. doi: 10.1097/00001756-200011270-00041. [DOI] [PubMed] [Google Scholar]
- Morelli M, Carta AR, Kachroo A, Schwarzschild MA. Pathophysiological roles for purines: adenosine, caffeine and urate. Prog Brain Res. 2010;183:183–208. doi: 10.1016/S0079-6123(10)83010-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mu Y, Gage FH. Adult hippocampal neurogenesis and its role in Alzheimer’s disease. Mol Neurodegener. 2011;6:85. doi: 10.1186/1750-1326-6-85. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nagele RG, D’Andrea MR, Lee H, Venkataraman V, Wang HY. Astrocytes accumulate Aβ 42 and give rise to astrocytic amyloid plaques in Alzheimer disease brains. Brain Res. 2003;971:197–209. doi: 10.1016/s0006-8993(03)02361-8. [DOI] [PubMed] [Google Scholar]
- Neher JJ, Neniskyte U, Hornik T, Brown GC. Inhibition of UDP/P2Y6 purinergic signaling prevents phagocytosis of viable neurons by activated microglia in vitro and in vivo. Glia. 2014;62:1463–1475. doi: 10.1002/glia.22693. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Neniskyte U, Neher JJ, Brown GC. Neuronal death induced by nanomolar amyloid β is mediated by primary phagocytosis of neurons by microglia. J Biol Chem. 2011;286:39904–39913. doi: 10.1074/jbc.M111.267583. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nguyen TD, Meichle S, Kim US, Wong T, Moody MW. P2Y11, a purinergic receptor acting via cAMP, mediates secretion by pancreatic duct epithelial cells. Am J Physiol Gastrointest Liver Physiol. 2001;280:G795–G804. doi: 10.1152/ajpgi.2001.280.5.G795. [DOI] [PubMed] [Google Scholar]
- Noda M, Suzumura A. Sweepers in the CNS: Microglial Migration and Phagocytosis in the Alzheimer Disease Pathogenesis. Int J Alzheimers Dis. 2012;2012:891087. doi: 10.1155/2012/891087. [DOI] [PMC free article] [PubMed] [Google Scholar]
- O’Brien RJ, Wong PC. Amyloid precursor protein processing and Alzheimer’s disease. Annu Rev Neurosci. 2011;34:185–204. doi: 10.1146/annurev-neuro-061010-113613. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Obulesu M, Venu R, Somashekhar R. Tau Mediated Neurodegeneration: An Insight into Alzheimer’s Disease Pathology. Neurochem Res. 2011;36:1329–1335. doi: 10.1007/s11064-011-0475-5. [DOI] [PubMed] [Google Scholar]
- Parajuli B, Sonobe Y, Horiuchi H, Takeuchi H, Mizuno T, Suzumura A. Oligomeric amyloid β induces IL-1β processing via production of ROS: implication in Alzheimer’s disease. Cell Death Dis. 2013;4:e975. doi: 10.1038/cddis.2013.503. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Parvathenani LK, Tertyshnikova S, Greco CR, Roberts SB, Robertson B, Posmantur R. P2X7 mediates superoxide production in primary microglia and is up-regulated in a transgenic mouse model of Alzheimer’s disease. J Biol Chem. 2003;278:13309–13317. doi: 10.1074/jbc.M209478200. [DOI] [PubMed] [Google Scholar]
- Patel NS, Paris D, Mathura V, Quadros AN, Crawford FC, Mullan MJ. Inflammatory cytokine levels correlate with amyloid load in transgenic mouse models of Alzheimer’s disease. J Neuroinflammation. 2005;2:9. doi: 10.1186/1742-2094-2-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pekny M, Wilhelmsson U, Pekna M. The dual role of astrocyte activation and reactive gliosis. Neurosci Lett. 2014;565:30–38. doi: 10.1016/j.neulet.2013.12.071. [DOI] [PubMed] [Google Scholar]
- Perregaux DG, McNiff P, Laliberte R, Conklyn M, Gabel CA. ATP acts as an agonist to promote stimulus-induced secretion of IL-1β and IL-18 in human blood. J Immunol. 2000;165:4615–4623. doi: 10.4049/jimmunol.165.8.4615. [DOI] [PubMed] [Google Scholar]
- Peterson TS, Camden JM, Wang Y, Seye CI, Wood WG, Sun GY, Erb L, Petris MJ, Weisman GA. P2Y2 nucleotide receptor-mediated responses in brain cells. Mol Neurobiol. 2010;41:356–366. doi: 10.1007/s12035-010-8115-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Peterson TS, Thebeau CN, Ajit D, Camden JM, Woods LT, Wood WG, Petris MJ, Sun GY, Erb L, Weisman GA. Up-regulation and activation of the P2Y2 nucleotide receptor mediate neurite extension in IL-1β-treated mouse primary cortical neurons. J Neurochem. 2013;125:885–896. doi: 10.1111/jnc.12252. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Piirainen H, Ashok Y, Nanekar RT, Jaakola VP. Structural features of adenosine receptors: from crystal to function. Biochim Biophys Acta. 2011;1808:1233–1244. doi: 10.1016/j.bbamem.2010.05.021. [DOI] [PubMed] [Google Scholar]
- Pinna A. Adenosine A2A receptor antagonists in Parkinson’s disease: progress in clinical trials from the newly approved istradefylline to drugs in early development and those already discontinued. CNS Drugs. 2014;28:455–474. doi: 10.1007/s40263-014-0161-7. [DOI] [PubMed] [Google Scholar]
- Pooler AM, Guez DH, Benedictus R, Wurtman RJ. Uridine enhances neurite outgrowth in nerve growth factor-differentiated PC12 [corrected] Neuroscience. 2005;134:207–214. doi: 10.1016/j.neuroscience.2005.03.050. [DOI] [PubMed] [Google Scholar]
- Popoli P, Pepponi R. Potential therapeutic relevance of adenosine A2B and A2A receptors in the central nervous system. CNS Neurol Disord Drug Targets. 2012;11:664–674. doi: 10.2174/187152712803581100. [DOI] [PubMed] [Google Scholar]
- Popoli P, Pintor A, Domenici MR, Frank C, Tebano MT, Pezzola A, Scarchilli L, Quarta D, Reggio R, Malchiodi-Albedi F, Falchi M, Massotti M. Blockade of striatal adenosine A2A receptor reduces, through a presynaptic mechanism, quinolinic acid-induced excitotoxicity: possible relevance to neuroprotective interventions in neurodegenerative diseases of the striatum. J Neurosci. 2002;22:1967–1975. doi: 10.1523/JNEUROSCI.22-05-01967.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rahman A. The role of adenosine in Alzheimer’s disease. Curr Neuropharmacol. 2009;7:207–216. doi: 10.2174/157015909789152119. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ratchford AM, Baker OJ, Camden JM, Rikka S, Petris MJ, Seye CI, Erb L, Weisman GA. P2Y2 nucleotide receptors mediate metalloprotease-dependent phosphorylation of epidermal growth factor receptor and ErbB3 in human salivary gland cells. J Biol Chem. 2010;285:7545–7555. doi: 10.1074/jbc.M109.078170. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rivera-Oliver M, Diaz-Rios M. Using caffeine and other adenosine receptor antagonists and agonists as therapeutic tools against neurodegenerative diseases: a review. Life Sci. 2014;101:1–9. doi: 10.1016/j.lfs.2014.01.083. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rodrigues RJ, Almeida T, Richardson PJ, Oliveira CR, Cunha RA. Dual presynaptic control by ATP of glutamate release via facilitatory P2X1, P2X2/3, and P2X3 and inhibitory P2Y1, P2Y2, and/or P2Y4 receptors in the rat hippocampus. J Neurosci. 2005;25:6286–6295. doi: 10.1523/JNEUROSCI.0628-05.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Roger S, Pelegrin P, Surprenant A. Facilitation of P2X7 receptor currents and membrane blebbing via constitutive and dynamic calmodulin binding. J Neurosci. 2008;28:6393–6401. doi: 10.1523/JNEUROSCI.0696-08.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rosi S, McGann K, Hauss-Wegrzyniak B, Wenk GL. The influence of brain inflammation upon neuronal adenosine A2B receptors. J Neurochem. 2003;86:220–227. doi: 10.1046/j.1471-4159.2003.01825.x. [DOI] [PubMed] [Google Scholar]
- Rubio ME, Soto F. Distinct Localization of P2X receptors at excitatory postsynaptic specializations. J Neurosci. 2001;21:641–653. doi: 10.1523/JNEUROSCI.21-02-00641.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ryu JK, McLarnon JG. Block of purinergic P2X7 receptor is neuroprotective in an animal model of Alzheimer’s disease. Neuroreport. 2008;19:1715–1719. doi: 10.1097/WNR.0b013e3283179333. [DOI] [PubMed] [Google Scholar]
- Salloway S, Sperling R, Gilman S, Fox NC, Blennow K, Raskind M, Sabbagh M, Honig LS, Doody R, van Dyck CH, Mulnard R, Barakos J, Gregg KM, Liu E, Lieberburg I, Schenk D, Black R, Grundman M. A phase 2 multiple ascending dose trial of bapineuzumab in mild to moderate Alzheimer disease. Neurology. 2009;73:2061–2070. doi: 10.1212/WNL.0b013e3181c67808. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sanz JM, Chiozzi P, Ferrari D, Colaianna M, Idzko M, Falzoni S, Fellin R, Trabace L, Di Virgilio F. Activation of microglia by amyloid β requires P2X7 receptor expression. J Immunol. 2009;182:4378–4385. doi: 10.4049/jimmunol.0803612. [DOI] [PubMed] [Google Scholar]
- Scharf S, Mander A, Ugoni A, Vajda F, Christophidis N. A double-blind, placebo-controlled trial of diclofenac/misoprostol in Alzheimer’s disease. Neurology. 1999;53:197–201. doi: 10.1212/wnl.53.1.197. [DOI] [PubMed] [Google Scholar]
- Schlachetzki JC, Hull M. Microglial activation in Alzheimer’s disease. Curr Alzheimer Res. 2009;6:554–563. doi: 10.2174/156720509790147179. [DOI] [PubMed] [Google Scholar]
- Schor NF. What the halted phase III γ-secretase inhibitor trial may (or may not) be telling us. Ann Neurol. 2011;69:237–239. doi: 10.1002/ana.22365. [DOI] [PubMed] [Google Scholar]
- Schwarzschild MA, Agnati L, Fuxe K, Chen JF, Morelli M. Targeting adenosine A2A receptors in Parkinson’s disease. Trends Neurosci. 2006;29:647–654. doi: 10.1016/j.tins.2006.09.004. [DOI] [PubMed] [Google Scholar]
- Shaftel SS, Griffin WS, O’Banion MK. The role of interleukin-1 in neuroinflammation and Alzheimer disease: an evolving perspective. J Neuroinflammation. 2008;5:7. doi: 10.1186/1742-2094-5-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Shieh CH, Heinrich A, Serchov T, van Calker D, Biber K. P2X7-dependent, but differentially regulated release of IL-6, CCL2, and TNF-α in cultured mouse microglia. Glia. 2014;62:592–607. doi: 10.1002/glia.22628. [DOI] [PubMed] [Google Scholar]
- Shors TJ, Miesegaes G, Beylin A, Zhao M, Rydel T, Gould E. Neurogenesis in the adult is involved in the formation of trace memories. Nature. 2001;410:372–376. doi: 10.1038/35066584. [DOI] [PubMed] [Google Scholar]
- Simi A, Lerouet D, Pinteaux E, Brough D. Mechanisms of regulation for interleukin-1β in neurodegenerative disease. Neuropharmacology. 2007;52:1563–1569. doi: 10.1016/j.neuropharm.2007.02.011. [DOI] [PubMed] [Google Scholar]
- Simpson JE, Ince PG, Lace G, Forster G, Shaw PJ, Matthews F, Savva G, Brayne C, Wharton SB. Astrocyte phenotype in relation to Alzheimer-type pathology in the ageing brain. Neurobiol Aging. 2010;31:578–590. doi: 10.1016/j.neurobiolaging.2008.05.015. [DOI] [PubMed] [Google Scholar]
- Sperlagh B, Illes P. Purinergic modulation of microglial cell activation. Purinergic Signal. 2007;3:117–127. doi: 10.1007/s11302-006-9043-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sperlagh B, Kofalvi A, Deuchars J, Atkinson L, Milligan CJ, Buckley NJ, Vizi ES. Involvement of P2X7 receptors in the regulation of neurotransmitter release in the rat hippocampus. J Neurochem. 2002;81:1196–1211. doi: 10.1046/j.1471-4159.2002.00920.x. [DOI] [PubMed] [Google Scholar]
- Sperlagh B, Vizi ES, Wirkner K, Illes P. P2X7 receptors in the nervous system. Prog Neurobiol. 2006;78:327–346. doi: 10.1016/j.pneurobio.2006.03.007. [DOI] [PubMed] [Google Scholar]
- Stagg J, Smyth MJ. Extracellular adenosine triphosphate and adenosine in cancer. Oncogene. 2010;29:5346–5358. doi: 10.1038/onc.2010.292. [DOI] [PubMed] [Google Scholar]
- Steinberg TH, Silverstein SC. Extracellular ATP4− promotes cation fluxes in the J774 mouse macrophage cell line. J Biol Chem. 1987;262:3118–3122. [PubMed] [Google Scholar]
- Stone TW, Ceruti S, Abbracchio MP. Adenosine receptors and neurological disease: neuroprotection and neurodegeneration. Handb Exp Pharmacol. 2009;193:535–587. doi: 10.1007/978-3-540-89615-9_17. [DOI] [PubMed] [Google Scholar]
- Suzuki T, Hide I, Ido K, Kohsaka S, Inoue K, Nakata Y. Production and release of neuroprotective tumor necrosis factor by P2X7 receptor-activated microglia. J Neurosci. 2004;24:1–7. doi: 10.1523/JNEUROSCI.3792-03.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tanaka J, Murate M, Wang CZ, Seino S, Iwanaga T. Cellular distribution of the P2X4 ATP receptor mRNA in the brain and non-neuronal organs of rats. Arch Histol Cytol. 1996;59:485–490. doi: 10.1679/aohc.59.485. [DOI] [PubMed] [Google Scholar]
- Trautmann A. Extracellular ATP in the immune system: more than just a "danger signal". Sci Signal. 2009;2:pe6. doi: 10.1126/scisignal.256pe6. [DOI] [PubMed] [Google Scholar]
- Tsuda M, Shigemoto-Mogami Y, Koizumi S, Mizokoshi A, Kohsaka S, Salter MW, Inoue K. P2X4 receptors induced in spinal microglia gate tactile allodynia after nerve injury. Nature. 2003;424:778–783. doi: 10.1038/nature01786. [DOI] [PubMed] [Google Scholar]
- Tsuda M, Tozaki-Saitoh H, Inoue K. Pain and purinergic signaling. Brain Res Rev. 2009;63:222–232. doi: 10.1016/j.brainresrev.2009.11.003. [DOI] [PubMed] [Google Scholar]
- Ulas J, Brunner LC, Nguyen L, Cotman CW. Reduced density of adenosine A1 receptors and preserved coupling of adenosine A1 receptors to G proteins in Alzheimer hippocampus: a quantitative autoradiographic study. Neuroscience. 1993;52:843–854. doi: 10.1016/0306-4522(93)90533-l. [DOI] [PubMed] [Google Scholar]
- Ulmann L, Hatcher JP, Hughes JP, Chaumont S, Green PJ, Conquet F, Buell GN, Reeve AJ, Chessell IP, Rassendren F. Up-regulation of P2X4 receptors in spinal microglia after peripheral nerve injury mediates BDNF release and neuropathic pain. J Neurosci. 2008;28:11263–11268. doi: 10.1523/JNEUROSCI.2308-08.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- van Gelder BM, Buijsse B, Tijhuis M, Kalmijn S, Giampaoli S, Nissinen A, Kromhout D. Coffee consumption is inversely associated with cognitive decline in elderly European men: the FINE Study. Eur J Clin Nutr. 2007;61:226–232. doi: 10.1038/sj.ejcn.1602495. [DOI] [PubMed] [Google Scholar]
- van Praag H, Schinder AF, Christie BR, Toni N, Palmer TD, Gage FH. Functional neurogenesis in the adult hippocampus. Nature. 2002;415:1030–1034. doi: 10.1038/4151030a. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Varma R, Chai Y, Troncoso J, Gu J, Xing H, Stojilkovic SS, Mattson MP, Haughey NJ. Amyloid-β induces a caspase-mediated cleavage of P2X4 to promote purinotoxicity. Neuromolecular Med. 2009;11:63–75. doi: 10.1007/s12017-009-8073-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Verderio C, Matteoli M. ATP mediates calcium signaling between astrocytes and microglial cells: modulation by IFN-γ. J Immunol. 2001;166:6383–6391. doi: 10.4049/jimmunol.166.10.6383. [DOI] [PubMed] [Google Scholar]
- Verhoef PA, Estacion M, Schilling W, Dubyak GR. P2X7 receptor-dependent blebbing and the activation of Rho-effector kinases, caspases, and IL-1β release. J Immunol. 2003;170:5728–5738. doi: 10.4049/jimmunol.170.11.5728. [DOI] [PubMed] [Google Scholar]
- Vial C, Roberts JA, Evans RJ. Molecular properties of ATP-gated P2X receptor ion channels. Trends Pharmacol Sci. 2004;25:487–493. doi: 10.1016/j.tips.2004.07.008. [DOI] [PubMed] [Google Scholar]
- Villalona-Calero MA, Otterson GA, Wientjes MG, Weber F, Bekaii-Saab T, Young D, Murgo AJ, Jensen R, Yeh TK, Wei Y, Zhang Y, Eng C, Grever M, Au JL. Noncytotoxic suramin as a chemosensitizer in patients with advanced non-small-cell lung cancer: a phase II study. Ann Oncol. 2008;19:1903–1909. doi: 10.1093/annonc/mdn412. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Villar-Menendez I, Porta S, Buira SP, Pereira-Veiga T, Diaz-Sanchez S, Albasanz JL, Ferrer I, Martin M, Barrachina M. Increased striatal adenosine A2A receptor levels is an early event in Parkinson’s disease-related pathology and it is potentially regulated by miR-34b. Neurobiol Dis. 2014;69:206–214. doi: 10.1016/j.nbd.2014.05.030. [DOI] [PubMed] [Google Scholar]
- Wang M, Kong Q, Gonzalez FA, Sun G, Erb L, Seye C, Weisman GA. P2Y nucleotide receptor interaction with α integrin mediates astrocyte migration. J Neurochem. 2005;95:630–640. doi: 10.1111/j.1471-4159.2005.03408.x. [DOI] [PubMed] [Google Scholar]
- Watano T, Calvert JA, Vial C, Forsythe ID, Evans RJ. P2X receptor subtype-specific modulation of excitatory and inhibitory synaptic inputs in the rat brainstem. J Physiol. 2004;558:745–757. doi: 10.1113/jphysiol.2004.066845. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Webster CM, Hokari M, McManus A, Tang XN, Ma H, Kacimi R, Yenari MA. Microglial P2Y12 deficiency/inhibition protects against brain ischemia. PLoS One [Electronic Resource] 2013;8:e70927. doi: 10.1371/journal.pone.0070927. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Weisman GA, Ajit D, Garrad R, Peterson TS, Woods LT, Thebeau C, Camden JM, Erb L. Neuroprotective roles of the P2Y2 receptor. Purinergic Signal. 2012a;8:559–578. doi: 10.1007/s11302-012-9307-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Weisman GA, Camden JM, Peterson TS, Ajit D, Woods LT, Erb L. P2 receptors for extracellular nucleotides in the central nervous system: role of P2X7 and P2Y2 receptor interactions in neuroinflammation. Mol Neurobiol. 2012b;46:96–113. doi: 10.1007/s12035-012-8263-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Weisman GA, De BK, Friedberg I, Pritchard RS, Heppel LA. Cellular responses to external ATP which precede an increase in nucleotide permeability in transformed cells. J Cell Physiol. 1984;119:211–219. doi: 10.1002/jcp.1041190211. [DOI] [PubMed] [Google Scholar]
- Weisman GA, Woods LT, Erb L, Seye CI. P2Y receptors in the mammalian nervous system: pharmacology, ligands and therapeutic potential. CNS Neurol Disord Drug Targets. 2012c;11:722–738. doi: 10.2174/187152712803581047. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Willuweit A, Velden J, Godemann R, Manook A, Jetzek F, Tintrup H, Kauselmann G, Zevnik B, Henriksen G, Drzezga A, Pohlner J, Schoor M, Kemp JA, von der Kammer H. Early-onset and robust amyloid pathology in a new homozygous mouse model of Alzheimer’s disease. PLoS ONE [Electronic Resource] 2009;4:e7931. doi: 10.1371/journal.pone.0007931. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Woods LT, Camden JM, Batek JM, Petris MJ, Erb L, Weisman GA. P2X7 receptor activation induces inflammatory responses in salivary gland epithelium. Am J Physiol Cell Physiol. 2012;303:C790–C801. doi: 10.1152/ajpcell.00072.2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wyss-Coray T. Inflammation in Alzheimer disease: driving force, bystander or beneficial response? Nat Med. 2006;12:1005–1015. doi: 10.1038/nm1484. [DOI] [PubMed] [Google Scholar]
- Yates SL, Burgess LH, Kocsis-Angle J, Antal JM, Dority MD, Embury PB, Piotrkowski AM, Brunden KR. Amyloid β and amylin fibrils induce increases in proinflammatory cytokine and chemokine production by THP-1 cells and murine microglia. J Neurochem. 2000;74:1017–1025. doi: 10.1046/j.1471-4159.2000.0741017.x. [DOI] [PubMed] [Google Scholar]
- Yu N, Erb L, Shivaji R, Weisman GA, Seye CI. Binding of the P2Y2 nucleotide receptor to filamin A regulates migration of vascular smooth muscle cells. Circ Res. 2008;102:581–588. doi: 10.1161/CIRCRESAHA.107.162271. [DOI] [PMC free article] [PubMed] [Google Scholar]

