Skip to main content

OPINION article

Front. Pharmacol., 22 March 2021
Sec. Neuropharmacology
This article is part of the Research Topic Purinergic Signaling 2020: the State-of-The-Art Commented by the Members of the Italian Purine Club View all 41 articles

An Open Question: Is the A2A Adenosine Receptor a Novel Target for Alzheimer’s Disease Treatment?

  • 1Department of Translational Medicine and for Romagna, University of Ferrara, Ferrara, Italy
  • 2Department of Neurology and Neuropathology, Golgi-Cenci Foundation and ASP Golgi-Redaelli, Abbiategrasso, Italy
  • 3University of Ferrara, Ferrara, Italy

Introduction

Neurocognitive Disorder due to Alzheimer’s Disease: A Brief Overview

According to DSM5, the term neurocognitive disorder (NCD) emphasizes that the cause of mental deficit lies in a pathology affecting neuronal circuits. The early clinical stages of NCD (mild-NCD/MCI) are characterized by functional preservation of everyday activities. Instead, if the disorder has a functional impact it is defined as major-NCD (dementia). On the other hand, the definition of the underlying pathology allows for the etiological classification of NCD (American Psychiatric Association, 2013; Sachdev et al., 2015). Based on the pathological deposition of proteins in brain tissue, NCD due to AD is characterized by a dual proteinopathy in which neurodegeneration is associated with the deposition of amyloid and phosphorylated TAU protein (pTAU). AD is the main age-related degenerative NCD progressively involving memory, complex attention, executive functions, language, and visual-perceptual functions. Personality and behavioural changes are also frequent further complicating the clinical course. On the other hand, due to the late involvement of the movement centers, motor function is usually spared until the most advanced stages of the disease. The AD syndromic evolution reflects the progressive spread of pTAU pathology from the allocortex (entorhinal cortex and hippocampus) to the neocortex (Elahi and Miller, 2017; Hanseeuw et al., 2019). Observing the neuropathology of AD is the starting point for deciphering its pathophysiological mechanisms and, therefore, identifying the biomarkers of the disease and the possible therapeutic targets. The macroscopic pathological feature characterizing advanced AD is diffuse brain atrophy due to widespread neurodegeneration causing synaptic and neuronal loss. Actually, the disease begins decades earlier with amyloid accumulation in the neocortex but amyloid deposition, which is very common even in physiological aging, is not sufficient to cause AD. The fundamental question is: what triggers neurodegeneration? Probably, the excess of amyloid-beta (Aβ) induces neurodegeneration through toxic oligomers. Indeed, soluble Aβ oligomers cause a synaptic reduction with a decrease in long-term potentiation and memory. Moreover, oligomers can reduce blood flow in brain capillaries and induce hyperphosphorylation of the AD-relevant epitopes of TAU protein (Selkoe and Hardy 2016; Nortley et al., 2019). Thus, Aβ load triggers neurodegeneration through oligomers which induce unbalanced activation of neuronal kinases resulting in excessive production of pTAU that, in turn, aggregates in pTAU toxic oligomers and spreads from its initial location in allocortex to neocortex. Together, oligomeric Aβ, synaptic pTAU aggregates and glial inflammatory activation are the main neurotoxic factors involved in the manifestation of a clinically relevant neurocognitive disorder (Perez-Nievas et al., 2013; Jack et al., 2018a). Typically, AD pathology shows extracellular accumulation of Aβ peptides (Aβ or senile plaques), as well as the hyperphosphorylated tau protein aggregates inside the dying neurons named neurofibrillary tangles (NFT) and neuropil threads (NT). Their combination constitutes the neuritic plaque (NP), which is the most typical feature of AD neuropathology. Thus, the neuropathological definition of AD requires a combination of scores for Amyloid, TAU (Braak stages), and NP (CERAD), which constitute the ABC criteria for the grading of AD related pathology (Mirra et al., 1991; Braak et al., 2006; Montine et al., 2012). Senile and neuritic plaques, consisting of protein and cellular debris, activate reactive and inflammatory processes by astrocytes and microglia which produce cytokines (IL-1β, IL-6) and NLRP3 inflammosome activation that, in turn, increase neurotoxic phenomena (Serrano-Pozo et al., 2016; Ising et al., 2019). On the basis of the neuropathological picture, several biomarkers have been developed for the in vivo definition of the pathology. Thus, the ATN system (Amyloid-TAU-Neurodegeneration) has been set up including 1) estimate of the amyloid load: Aβ decrease in cerebrospinal fluid (CSF) and/or Aβ cortical accumulation at amyloid-PET; 2) pTAU valuation: pTAU increase in CSF and/or pTAU cortical accumulation at TAU-PET; 3) extent of neurodegeneration: atrophic pattern at brain MRI and/or hypometabolism at FDG-PET and/or increase of total-TAU in CSF (Jack et al., 2018b; Chételat et al., 2020). These markers can allow for early diagnosis or even can identify those most at risk of developing AD in a preclinical phase (before mild-NCD) in order to implement timely therapeutic interventions (Dubois et al., 2016). Nonetheless, there is now no cure for AD and this approach poses ethical problems, as well as being invasive and expensive; therefore, an intensive search for biomarkers obtainable from peripheral blood is still in progress (Lewczuk et al., 2018; Molinuevo et al., 2018).

The early mechanisms leading to Aβ accumulation and initial generation of toxic molecules are elusive and multiple, and belong to the individual trajectory of cerebral aging linked to non-modifiable genetic factors (AD-related polymorphisms, APO-E4 allele, and pathogenic mutations in PSN-1-2 and APP genes, and Williamson et al., 2009; Vermunt et al., 2019), and to modifiable factors related to the individual’s personal history including favorable behaviors (regular physical and mental activity, high education, healthy diet, social engagement) and harmful conditions (midlife obesity, diabetes, hypertension, smoke, excessive alcohol, and hearing loss) (Lourida et al., 2019; Livingston et al., 2020). Early pathogenesis of sporadic AD is quite complex. Just as there are different forms of hepatitis that lead to cirrhosis, there are different pathophysiological paths that lead to AD. However, the sine qua non for the development of AD pathophysiology is the accumulation of amyloid in the cerebral cortex. Indeed, many efforts are being made to reduce the presence of amyloid in the cerebral cortex, especially through the use of costly monoclonal antibodies (e.g., phase3 trials: Aducanumab, Gantenerumab; phase2 trial: Crenezumab). Actually, amyloid reduction is only one aspect of the therapeutic approach and there is increasing attention to non-amyloid targets with 121 agents having clinical trials in course for the treatment of AD (Cummings et al., 2020). Particularly, a new challenge is the development of immunotherapies capable of blocking the toxic pTAU species (Bittar et al., 2020). The multifactorial nature of the AD would require an early, long-lasting, and multi-dimensional therapeutic approach which should be personalized and based on the patient’s clinical and biomarker characteristics (Sperling et al., 2011; Cummings et al., 2018; Hara et al., 2019). The current possible intervention strategies to improve the AD course depend on the stage of pathology and progressively include: prevention measures (healthy and active lifestyle, reduction of detrimental factors), disease modifying treatments (reduction of Aβ load and toxic oligomers, containment of TAU phosphorylation, toxic pTAU species and neuroinflammation, enhancement of neuronal resilience), symptomatic therapies (modulation of synaptic functions and improvement of synaptic efficiency, and Long and Holtzman 2019). Nonetheless, it should be taken into account that many senile cases of AD present mixed pathologies and in the extreme stages of senility it becomes unrealistic to stem neurodegeneration. In this framework, adenosine receptors, especially in the hippocampus, constitute a new and interesting therapeutic target through which it is possible to modulate and improve synaptic activity, obtaining symptomatic and perhaps disease-modifying effects.

Role of A2A Adenosine Receptors in AD: State of the Art and Discussion

Adenosine is an ubiquitous autacoid derived by ATP dephosphorylation, that modulates several responses in CNS, by activating four G-protein coupled receptors, A1, A2A, A2B, and A3 present on both neuronal and glial cells (Borea et al., 2018). This nucleoside is generated at both intra– and extracellular level following AMP dephosphorylation by 5′-nucleotidases and its extracellular concentration is regulated both from equilibrative nucleoside transporters as well as through exocytosis by neurons and astrocytes (Borea et al., 2016).

Adenosine regulates several physiological functions including sleep, cognitive performances, and memory and its main role is to regulate neuron excitatory synaptic transmission by inhibitory A1 receptors and synaptic plasticity via facilitatory A2A receptors (Cunha, 2008; Cunha, 2016; Cieślak and Wojtczak, 2018). In particular, the A2A subtype, mainly present in striatal area, has been now recognized in other cerebral regions including cortex and hippocampus, where due to its expression at presynaptic level, it affects the release of excitatory neurotransmitters, like glutamate (Cunha et al., 1994; Lopes et al., 2002; Marchi et al., 2002). As for its synaptic expression in spite of episodic evidence (Canas et al., 2018), it is still debatable if there is A2A receptors expression in synapses, although it is well established that A2A receptors are located in hippocampal synapses, with a density about 20–time lower than in the striatum (Lopes et al., 2004; Rebola et al., 2005). Although in healthy human brains the A2A receptor may exert protective functions, by regulating other proteins as BDNF, its signaling is strongly modified in the hippocampus following aging (Rebola et al., 2003; Tebano et al., 2010; Temido-Ferreira et al., 2019; Temido-Ferreira et al., 2020). In this condition, there is a rise of A2A receptor and G proteins-coupling, leading to an increase in glutamate release, mGluR5-dependent NMDA receptor overstimulation, and enhanced calcium influx responsible for synaptic alterations and memory dysfunction (Temido-Ferreira et al., 2020). These findings suggest a role for this receptor subtype in the pathogenesis of different neurocognitive disorders and specifically AD (Costenla et al., 2011; Rebola et al., 2011; Horgusluoglu-Moloch et al., 2017; Temido-Ferreira et al., 2019). Indeed, synaptic dysfunction and damage are key features in early AD (Selkoe, 2002; Coleman et al., 2004). Interestingly, A2A receptor is overexpressed in both frontal cortex and hippocampus of aged and transgenic-AD animals as well as in AD patients (Lopes et al., 1999; Arendash et al., 2006; Albasanz et al., 2008; Espinosa et al., 2013; Li et al., 2015; Orr et al., 2015; Pagnussat et al., 2015; Gonçalves et al., 2019; Temido-Ferreira et al., 2020). APP/PS1 mouse model of AD amyloidosis show an upregulation and activation of A2A adenosine receptors hampering long-term synaptic potentiation (LTP) in hippocampal CA3 pyramidal cells (Viana da Silva et al., 2016). Several literature data report the use of pharmacological and genetic approaches to demonstrate that A2A adenosine receptors block prevents synaptic damage and cognitive impairments in animal models following Aβ exposure, suggesting that A2A receptor antagonists might reduce synaptotoxicity (Dall'Igna et al., 2007; Canas et al., 2009; Orr et al., 2018). Moreover, antagonism of A2A adenosine receptors in animal models of Tau pathology inhibits Tau hyperphosphorylation, hippocampal neuroinflammation, while protects spatial memory and hippocampal long-term depression (Laurent et al., 2016). Accordingly, overexpression of A2A adenosine receptors, in a tauopathy mouse model, increases tau hyperphosphorylation and consequent tau-dependent memory impairments (Carvalho et al., 2019).

Adenosine, deriving from an increase of ecto-5′-nucleotidase (CD73) activity in animal model of early AD, induced memory deficits, LTP impairment and synaptic markers reduction in a CD73 or A2A adenosine receptor-dependent way (Gonçalves et al., 2019). Indeed, among the early mechanisms involved in memory deterioration, synaptic dysfunction and selective synaptic degeneration stand out as one of the more robust and reproducible events. In fact, the early works on neuropathological changes associated with dementia established the loss of synaptic markers as a key process (Terry et al., 1991). More recent work showed that the loss of synapses is indeed of the earliest neuropathological changes in the brains of MCI and early AD patients, namely in the hippocampus (Scheff et al., 2007; Scheff et al., 2015). Accordingly, animal studies confirmed that synaptic dysfunction is an early event at the onset of memory perturbations (Canas et al., 2009; Viana da Silva et al., 2016; Silva et al., 2018). This justifies the proposal that AD is a synaptic-based disease (Selkoe, 2002) and that synaptic modulators may be paramount to control early AD (Coleman et al., 2004). Another crucial role for the A2A adenosine receptor is its important ability to modulate glial cell functions, affecting pro-inflammatory cytokines release and neuroinflammation (Illes et al., 2020). Specifically, it plays an essential role in activated microglia, located near amyloid plaques typical of AD (Franco et al., 2020), where its upregulation is responsible for a raise of M1 microglial markers (IL-1β, IL-6, TNF-α) and its antagonism prevents hippocampal LTP impairments, as well as IL-1β production, paventing a regulatory function for it in reducing memory dysfunction (Colella et al., 2018; Franco et al., 2019). Several studies support a role of A2A receptor as drug target in both neurons and microglia to revert memory deficit and neurodegeneration in AD (Santiago et al., 2014; Cunha, 2016). Recently, it has been reported that A2A subtypes interact with NMDA receptors producing A2A-NMDA heteromers, mainly in microglia, characterized by bidirectional cross-antagonism, where A2A receptor inhibition decreases hyperactivation of glutamatergic signalling by blocking NMDA receptor-mediated currents (Rebola et al., 2008; Mouro et al., 2018; Franco et al., 2020; Temido-Ferreira et al., 2020). In addition, it forms important complexes with CB2 cannabinoid receptor subtypes, presenting cross-interaction, thus modifying the pathway of each other. In this heteromer, A2A receptor antagonism provides an increase in CB2 receptor activity suggesting, for the first time, that A2A receptor block rises the neuroprotective action of endocannabinoids important for AD therapy (Franco et al., 2019). Interestingly, these receptorial complexes were enhanced in a transgenic AD mouse model (Franco et al., 2020). Although these heteromers might have a role, recently, A2A receptor-mediated effects in AD-related features have been attributed to monomeric forms (Temido-Ferreira et al., 2020).

Clinical data, based on the effect of caffeine consumption in elderly, encourage the use of A2A adenosine antagonists to prevent memory deficits. Indeed, caffeine is the most widely consumed psychostimulant substance, present in coffee, tea, cola, chocolate, and other foods, exerting benefical effects in dementia and AD (Eskelinen et al., 2009; Eskelinen and Kivipelto, 2010; Santos et al., 2010; Gelber et al., 2011; Liu et al., 2016; Sugiyama et al., 2016; Reyes and Cornelis, 2018; Domenici et al., 2019; Dong et al., 2020; Iranpour et al., 2020). Interestingly it has been reported that non-toxic doses/concentrations of caffeine mostly act on A2A receptors in the brain (Yu et al., 2009; Lopes et al., 2019). Accordingly, antagonism of A2A receptors is one of the main effects of caffeine (Jacobson et al., 2020).

Specifically, coffee consumption correlated with reduction of cognitive function, with significant effects obtained with three cups of coffee per day (Ritchie et al., 2007; van Gelder et al., 2007). Indeed, a retrospective analysis during 20 years before AD development, revealed a negative correlation between coffee intake and disease diagnosis, with lower quantity of caffeine at day assumed by patients with AD in contrast to higher amounts of caffeine in control subjects (Maia and de Mendonca, 2002). Furthermore, a prospective work evaluating the effect of coffee intake assumed every day on AD development, showed a reduction of AD risk by 31%, following 5 years examination (Lindsay et al., 2002). In addition, lower caffeine levels were observed in plasma of mild cognitive impairments patients developing later dementia, in comparison to those who did not develop the disease. Therefore, high levels of caffeine were related to the lack of dementia development in a window of 2/4 years (Cao et al., 2012). Accordingly, a high daily consumption of 3–5 cups of coffee reduced the risk of dementia and AD of 65–70 and 62–64%, respectively, in comparison to a lower assumption (Eskelinen et al., 2009). More generally, caffeine intake was associated to the absence of dementia and cerebral injuries typical of AD and to an increase of long-term memories consolidation in humans (Gelber et al., 2011; Borota et al., 2014; Favila and Kuhl, 2014).

In animal models of AD, administration of caffeine, has been associated to a reduced risk for memory decline and dysfunction, beta-amyloid production and tau hyperphosphorylation (Costa et al., 2008; Arendash et al., 2009; Canas et al., 2009; Cao et al., 2009; Eskelinen and Kivipelto, 2010; Santos et al., 2010; Laurent et al., 2014; Kaster et al., 2015; Kolahdouzan and Hamadeh, 2017). Finally, recent works support the utility of caffeine intake as antioxidant and antiinflammatory agent (Janitschke et al., 2019; Sinyor et al., 2020). However, it has to be remarqued that although caffeine is an abundant bioactive molecule in coffee beverages, these have over 2,000 other chemicals that may have biological effects. In this respect, it is interesting that it is the amounts of a caffeine metabolite, the obrominel, rather than caffeine levels in the CSF that correlate with amyloid/tau markers in demented patients (Travassos et al., 2015). Interestingly, the intake of chocolate, rich in the obromine, is inversely correlated with memory deterioration (Moreira et al., 2016).

It is important to remarque that the neuroprotective effects of caffeine, associated to A2A adenosine receptor inhibition, have been observed also in Parkinson’s disease, where much work has been carried out to demonstrate safety of the first A2A adenosine receptor antagonist, istradefylline, recently launched as a new drug for this pathology in Japan (Nouriast) and in the United States (Nourianz) (Borea et al., 2016, Borea et al., 2017; Chen and Cunha, 2020). Istradefylline has been also shown to exert protective effects by reducing memory dysfunction in animal models of AD and for the future it would be crucial to determine whether it could also induce memory improvement in patients with AD (Orr et al., 2018). However, it should be underlined that istradefylline has a narrow therapeutic window in aging and experimental models of AD and PD, leading to the hypothesis that age and other factors may affect safety of A2A receptor antagonists.

Finally, this opinion article has presented the main findings supporting the role of A2A adenosine receptor antagonists on AD. Even though further work is necessary to better elucidate the mechanisms involved in the shift of A2A receptor from beneficial target in normal synapses to detrimental one in aging and disease, its capability to modulate synaptotoxicity, glutamate-dependent NMDA signaling and calcium dysfunction, together with its effect on neuroinflammation, suggest a crucial role for its antagonism to prevent AD pathology.

Author Contributions

SM, SG, PAB, and TEP conceived the work and wrote the manuscript. LP, SP, FV, and KV contributed to writing and editing. All authors contributed to the article and approved the submitted version.

Conflict of Interest

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

References

Albasanz, J. L., Perez, S., Barrachina, M., Ferrer, I., and Martín, M. (2008). Up-regulation of adenosine receptors in the frontal cortex in Alzheimer’s disease. Brain Pathol. 18, 211–219. doi:10.1111/j.1750-3639.2007.00112.x

PubMed Abstract | CrossRef Full Text | Google Scholar

American Psychiatric Association (2013). Diagnostic and statistical manual of mental disorders (DSM-5). 5th Edn. American Psychiatric Publishing, Inc. Available at: https://books.google.it/books?id=-JivBAAAQBAJ (Accessed May 18, 2013).

Arendash, G. W., Mori, T., Cao, C., Mamcarz, M., Runfeldt, M., Dickson, A., et al. (2009). Caffeine reverses cognitive impairment and decreases brain amyloid-beta levels in aged Alzheimer's disease mice. J. Alzheimers Dis. 17, 661–680. doi:10.3233/JAD-2009-1087

PubMed Abstract | CrossRef Full Text | Google Scholar

Arendash, G. W., Schleif, W., Rezai-Zadeh, K., Jackson, E. K., Zacharia, L. C., Cracchiolo, J. R., et al. (2006). Caffeine protects Alzheimer’s mice against cognitive impairment and reduces brain beta-amyloid production. Neuroscience 142, 941–952. doi:10.1016/j.neuroscience.2006.07.021

PubMed Abstract | CrossRef Full Text | Google Scholar

Bittar, A., Bhatt, N., and Kayed, R. (2020). Advances and considerations in AD tau-targeted immunotherapy. Neurobiol. Dis. 134, 104707. doi:10.1016/j.nbd.2019.104707

PubMed Abstract | CrossRef Full Text | Google Scholar

Borea, P. A., Gessi, S., Merighi, S., and Varani, K. (2016). Adenosine as a multi-signalling guardian angel in human diseases: when, where and how does it exert its protective effects?. Trends Pharmacol. Sci. 37, 419–434. doi:10.1016/j.tips.2016.02.006

PubMed Abstract | CrossRef Full Text | Google Scholar

Borea, P. A., Gessi, S., Merighi, S., Vincenzi, F., and Varani, K. (2017). Pathological overproduction: the bad side of adenosine. Br. J. Pharmacol. 174, 1945–1960. doi:10.1111/bph.13763

PubMed Abstract | CrossRef Full Text | Google Scholar

Borea, P. A., Gessi, S., Merighi, S., Vincenzi, F., and Varani, K. (2018). Pharmacology of adenosine receptors: the state of the art. Physiol. Rev. 98, 1591–1625. doi:10.1152/physrev.00049.2017

PubMed Abstract | CrossRef Full Text | Google Scholar

Borota, D., Murray, E., Keceli, G., Chang, A., Watabe, J. M., Ly, M., et al. (2014). Post-study caffeine administration enhances memory consolidation in humans. Nat. Neurosci. 17, 201–203. doi:10.1038/nn.3623

PubMed Abstract | CrossRef Full Text | Google Scholar

Braak, H., Alafuzoff, I., Arzberger, T., Kretzschmar, H., and Del Tredici, K. (2006). Staging of Alzheimer disease-associated neurofibrillary pathology using paraffin sections and immunocytochemistry. Acta Neuropathol. 112, 389–404. doi:10.1007/s00401-006-0127-z

PubMed Abstract | CrossRef Full Text | Google Scholar

Canas, P. M., Porciúncula, L. O., Cunha, G. M., Silva, C. G., Machado, N. J., Oliveira, J. M., et al. (2009). Adenosine A2A receptor blockade prevents synaptotoxicity and memory dysfunction caused by beta-amyloid peptides via p38 mitogen-activated protein kinase pathway. J. Neurosci. 29, 14741–14751. doi:10.1523/JNEUROSCI.3728-09.2009

PubMed Abstract | CrossRef Full Text | Google Scholar

Canas, P. M., Porciúncula, L. O., Simões, A. P., Augusto, E., Silva, H. B., Machado, N. J., et al. (2018). Neuronal adenosine A2A receptors are critical mediators of neurodegeneration triggered by convulsions. eNeuro 5 (6), 0385–418. doi:10.1523/ENEURO.0385-18.2018

CrossRef Full Text | Google Scholar

Cao, C., Cirrito, J. R., Lin, X., Wang, L., Wang, L., Verges, D. K., et al. (2009). Caffeine suppresses amyloid-beta levels in plasma and brain of Alzheimer’s disease transgenic mice. J. Alzheimer’s Dis. 17, 681–697. doi:10.3233/JAD-2009-1071

CrossRef Full Text | Google Scholar

Cao, C., Loewenstein, D. A., Lin, X., Zhang, C., Wang, L., Duara, R., et al. (2012). High Blood caffeine levels in MCI linked to lack of progression to dementia. J. Alzheimer’s Dis. 30, 559–572. doi:10.3233/JAD-2012-111781

CrossRef Full Text | Google Scholar

Carvalho, K., Faivre, E., Pietrowski, M. J., Marques, X., Gomez-Murcia, V., Deleau, A., et al. (2019). Exacerbation of C1q dysregulation, synaptic loss and memory deficits in tau pathology linked to neuronal adenosine A2A receptor. Brain 142, 3636–3654. doi:10.1093/brain/awz288

PubMed Abstract | CrossRef Full Text | Google Scholar

Chen, J. F., and Cunha, R. A. (2020). The belated United States FDA approval of the adenosine A2A receptor antagonist istradefylline for treatment of Parkinson’s disease. Purinergic Signal. 16, 167–174. doi:10.1007/s11302-020-09694-2

PubMed Abstract | CrossRef Full Text | Google Scholar

Chételat, G., Arbizu, J., Barthel, H., Garibotto, V., Law, I., Morbelli, S., et al. (2020). Amyloid-PET and 18F-FDG-PET in the diagnostic investigation of Alzheimer’s disease and other dementias. Lancet Neurol. 19, 951–962. doi:10.1016/S1474-4422(20)30314-8

PubMed Abstract | CrossRef Full Text | Google Scholar

Cieślak, M., and Wojtczak, A. (2018). Role of purinergic receptors in the Alzheimer’s disease. Purinergic Signal. 14, 331–344. doi:10.1007/s11302-018-9629-0

PubMed Abstract | CrossRef Full Text | Google Scholar

Colella, M., Zinni, M., Pansiot, J., Cassanello, M., Mairesse, J., Ramenghi, L., et al. (2018). Modulation of microglial activation by adenosine A2A receptor in animal models of perinatal brain injury. Front. Neurol. 9, 605. doi:10.3389/fneur.2018.00605

PubMed Abstract | CrossRef Full Text | Google Scholar

Coleman, P., Federoff, H., and Kurlan, R. (2004). A focus on the synapse for neuroprotection in Alzheimer disease and other dementias. Neurology 63, 1155–1162. doi:10.1212/01.wnl.0000140626.48118.0a

PubMed Abstract | CrossRef Full Text | Google Scholar

Costa, M. S., Botton, P. H., Mioranzza, S., Souza, D. O., and Porciúncula, L. O. (2008). Caffeine prevents age-associated recognition memory decline and changes brain-derived neurotrophic factor and tirosine kinase receptor (TrkB) content in mice. Neuroscience 153, 1071–1078. doi:10.1016/j.neuroscience.2008.03.038

PubMed Abstract | CrossRef Full Text | Google Scholar

Costenla, A. R., Diógenes, M. J., Canas, P. M., Rodrigues, R. J., Nogueira, C., Maroco, J., et al. (2011). Enhanced role of adenosine A2A receptors in the modulation of LTP in the rat hippocampus upon ageing. Eur. J. Neurosci. 34, 12–21. doi:10.1111/j.1460-9568.2011.07719.x

PubMed Abstract | CrossRef Full Text | Google Scholar

Cummings, J. L., Cohen, S., van Dyck, C. H., Brody, M., Curtis, C., Cho, W., et al. (2018). ABBY: a phase 2 randomized trial of crenezumab in mild to moderate Alzheimer disease. Neurology 90, e1889–e1897. doi:10.1212/WNL.0000000000005550

PubMed Abstract | CrossRef Full Text | Google Scholar

Cummings, J., Morstorf, T., Zhong, K., Sabbagh, M., and Zhong, K. (2020). Alzheimer’s disease drug-development pipeline: few candidates, frequent failures. Alzheimers Res. Ther. 6, e12050. doi:10.1002/trc2.1205010.1186/alzrt269

CrossRef Full Text | Google Scholar

Cunha, R. A., Johansson, B., van der Ploeg, I., Sebastião, A. M., Ribeiro, J. A., and Fredholm, B. B. (1994). Evidence for functionally important adenosine A2A receptors in the rat hippocampus. Brain Res. 649, 208–216. doi:10.1016/0006-8993(94)91066-9

PubMed Abstract | CrossRef Full Text | Google Scholar

Cunha, R. A. (2008). Different cellular sources and different roles of adenosine: A1 receptor-mediated inhibition through astrocytic-driven transmission and synapse-restricted A2A receptor-mediated facilitation of plasticity. Neurochem. Int. 52, 65–72. doi:10.1016/j.neuint.2007.06.026

PubMed Abstract | CrossRef Full Text | Google Scholar

Cunha, R. A. (2016). How does adenosine control neuronal dysfunction and neurodegeneration?. J. Neurochem. 139, 1019–1055. doi:10.1111/jnc.13724

PubMed Abstract | CrossRef Full Text | Google Scholar

Dall'Igna, O. P., Fett, P., Gomes, M. W., Souza, D. O., Cunha, R. A., and Lara, D. R. (2007). Caffeine and adenosine A2A receptor antagonists prevent beta-amyloid (25–35)-induced cognitive deficits in mice. Exp. Neurol. 203, 241–245. doi:10.1016/j.expneurol.2006.08.008

PubMed Abstract | CrossRef Full Text | Google Scholar

Domenici, M. R., Ferrante, A., Martire, A., Chiodi, V., Pepponi, R., Tebano, M. T., et al. (2019). Adenosine A2A receptor as potential therapeutic target in neuropsychiatric disorders. Pharmacol. Res. 147, 104338. doi:10.1016/j.phrs.2019.104338

PubMed Abstract | CrossRef Full Text | Google Scholar

Dong, X., Li, S., Sun, J., Li, Y., and Zhang, D. (2020). Association of coffee, decaffeinated coffee and caffeine intake from coffee with cognitive performance in older adults: national health and nutrition examination survey (NHANES) 2011–2014. Nutrients 12, 840. doi:10.3390/nu12030840

CrossRef Full Text | Google Scholar

Dubois, B., Hampel, H., Feldman, H. H., Scheltens, P., Aisen, P., Andrieu, S., et al. (2016). Preclinical Alzheimer’s disease: definition, natural history, and diagnostic criteria. Alzheimers Dement. 12, 292–323. doi:10.1016/j.jalz.2016.02.002

PubMed Abstract | CrossRef Full Text | Google Scholar

Elahi, F. M., and Miller, B. L. (2017). A clinicopathological approach to the diagnosis of dementia. Nat. Rev. Neurol. 13, 457–476. doi:10.1038/nrneurol.2017.96

PubMed Abstract | CrossRef Full Text | Google Scholar

Eskelinen, M. H., and Kivipelto, M. (2010). Caffeine as a protective factor in dementia and Alzheimer’s disease. J. Alzheimers Dis. 20 (Suppl. 1), S167–S174. doi:10.3233/JAD-2010-1404

PubMed Abstract | CrossRef Full Text | Google Scholar

Eskelinen, M. H., Ngandu, T., Tuomilehto, J., Soininen, H., and Kivipelto, M. (2009). Midlife coffee and tea drinking and the risk of late-life dementia: a population-based CAIDE study. J. Alzheimers Dis. 16, 85–91. doi:10.3233/JAD-2009-0920

PubMed Abstract | CrossRef Full Text | Google Scholar

Espinosa, J., Rocha, A., Nunes, F., Costa, M. S., Schein, V., Kazlauckas, V., et al. (2013). Caffeine consumption prevents memory impairment, neuronal damage, and adenosine A2A receptors upregulation in the hippocampus of a rat model of sporadic dementia. J. Alzheimers Dis. 34, 509–518. doi:10.3233/JAD-111982

PubMed Abstract | CrossRef Full Text | Google Scholar

Favila, S. E., and Kuhl, B. A. (2014). Stimulating memory consolidation. Nat. Neurosci. 17, 151–152. doi:10.1038/nn.3638

PubMed Abstract | CrossRef Full Text | Google Scholar

Franco, R., Reyes-Resina, I., Aguinaga, D., Lillo, A., Jiménez, J., Raïch, I., et al. (2019). Potentiation of cannabinoid signaling in microglia by adenosine A2A receptor antagonists. Glia 67, 2410–2423. doi:10.1002/glia.23694

PubMed Abstract | CrossRef Full Text | Google Scholar

Franco, R., Rivas-Santisteban, R., Casanovas, M., Lillo, A., Saura, C. A., and Navarro, G. (2020). Adenosine A2A receptor antagonists affects NMDA glutamate receptor function. Potential to address neurodegeneration in alzheimer’s disease. Cells 9, 1075. doi:10.3390/cells9051075

CrossRef Full Text | Google Scholar

Gelber, R. P., Petrovitch, H., Masaki, K. H., Ross, G. W., and White, L. R. (2011). Coffee intake in midlife and risk of dementia and its neuropathologic correlates. J. Alzheimers Dis. 23, 607–615. doi:10.3233/JAD-2010-101428

PubMed Abstract | CrossRef Full Text | Google Scholar

Gonçalves, F. Q., Lopes, J. P., Silva, H. B., Lemos, C., Silva, A. C., Gonçalves, N., et al. (2019). Synaptic and memory dysfunction in a β-amyloid model of early Alzheimer’s disease depends on increased formation of ATP-derived extracellular adenosine. Neurobiol. Dis. 132, 104570. doi:10.1016/j.nbd.2019.104570

PubMed Abstract | CrossRef Full Text | Google Scholar

Hanseeuw, B. J., Betensky, R. A., Jacobs, H. I. L., Schultz, A. P., Sepulcre, J., Becker, J. A., et al. (2019). Association of amyloid and tau with cognition in preclinical Alzheimer disease. JAMA Neurol. 76, 915–924. doi:10.1001/jamaneurol.2019.1424

CrossRef Full Text | Google Scholar

Hara, Y., McKeehan, N., and Fillit, H. M. (2019). Translating the biology of aging into novel therapeutics for Alzheimer disease. Neurology 92, 84–93. doi:10.1212/WNL.0000000000006745

PubMed Abstract | CrossRef Full Text | Google Scholar

Horgusluoglu-Moloch, E., Nho, K., Risacher, S. L., Kim, S., Foroud, T., Shaw, L. M., et al. (2017). Targeted neurogenesis pathway-based gene analysis identifies ADORA2A associated with hippocampal in mild cognitive impairment and Alzheimer’s disease. Neurobiol. Aging 60, 92–103. doi:10.1016/j.neurobiolaging.2017.08.010

PubMed Abstract | CrossRef Full Text | Google Scholar

Illes, P., Rubini, P., Ulrich, H., Zhao, Y., and Tang, Y. (2020). Regulation of microglial functions by purinergic mechanisms in the healthy and diseased CNS. Cells 9, 1108. doi:10.3390/cells9051108

CrossRef Full Text | Google Scholar

Iranpour, S., Saadati, H. M., Koohi, F., and Sabour, S. (2020). Association between caffeine intake and cognitive function in adults; effect modification by sex: data from national health and nutrition examination survey (NHANES) 2013–2014. Clin. Nutr. 39, 2158–2168. doi:10.1016/j.clnu.2019.09.003

PubMed Abstract | CrossRef Full Text | Google Scholar

Ising, C., Venegas, C., Zhang, S., Scheiblich, H., Schmidt, S. V., Vieira-Saecker, A., et al. (2019). NLRP3 inflammasome activation drives tau pathology. Nature 575, 669–673. doi:10.1038/s41586-019-1769-z

PubMed Abstract | CrossRef Full Text | Google Scholar

Jack, C. R., Wiste, H. J., Schwarz, C. G., Lowe, V. J., Senjem, M. L., Vemuri, P., et al. (2018a). Longitudinal tau PET in ageing and Alzheimer’s disease. Brain 141, 1517–1528. doi:10.1093/brain/awy059

PubMed Abstract | CrossRef Full Text | Google Scholar

Jack, C. R., Bennett, D. A., Blennow, K., Carrillo, M. C., Dunn, B., Haeberlein, S. B., et al. (2018b). NIA-AA research framework: toward a biological definition of Alzheimer’s disease. Alzheimers Dement 14, 535–562. doi:10.1016/j.jalz.2018.02.018

PubMed Abstract | CrossRef Full Text | Google Scholar

Jacobson, K. A., Gao, Z. G., Matricon, P., Eddy, M. T., and Carlsson, J. (2020). Adenosine A2A receptor antagonists: from caffeine to selective non‐xanthines. Br. J. Pharmacol. doi:10.1111/bph.15103

CrossRef Full Text | Google Scholar

Janitschke, D., Nelke, C., Lauer, A., Regner, L., Winkler, J., Thiel, A., et al. (2019). Effect of caffeine and other methylxanthines on Aβ-homeostasis in SH-SY5Y cells. Biomolecules 9, 689. doi:10.3390/biom9110689

CrossRef Full Text | Google Scholar

Kaster, M. P., Machado, N. J., Silva, H. B., Nunes, A., Ardais, A. P., Santana, M., et al. (2015). Caffeine acts through neuronal adenosine A2A receptors to prevent mood and memory dysfunction triggered by chronic stress. Proc. Natl. Acad. Sci. U.S.A. 112, 7833–7838. doi:10.1073/pnas.1423088112

PubMed Abstract | CrossRef Full Text | Google Scholar

Kolahdouzan, M., and Hamadeh, M. J. (2017). The neuroprotective effects of caffeine in neurodegenerative diseases. CNS Neurosci. Ther. 23, 272–290. doi:10.1111/cns.12684

PubMed Abstract | CrossRef Full Text | Google Scholar

Laurent, C., Eddarkaoui, S., Derisbourg, M., Leboucher, A., Demeyer, D., Carrier, S., et al. (2014). Beneficial effects of caffeine in a transgenic model of Alzheimer’s disease-like tau pathology. Neurobiol. Aging 35, 2079–2090. doi:10.1016/j.neurobiolaging.2014.03.027

PubMed Abstract | CrossRef Full Text | Google Scholar

Laurent, C., Burnouf, S., Ferry, B., Batalha, V. L., Coelho, J. E., Baqi, Y., et al. (2016). A2A adenosine receptor deletion is protective in a mouse model of Tauopathy. Mol. Psychiatry 21, 97–107. doi:10.1038/mp.2014.151

PubMed Abstract | CrossRef Full Text | Google Scholar

Lewczuk, P., Ermann, N., Andreasson, U., Schultheis, C., Podhorna, J., Spitzer, P., et al. (2018). Plasma neurofilament light as a potential biomarker of neurodegeneration in Alzheimer’s disease. Alzheimers Res. Ther. 10, 71. doi:10.1186/s13195-018-0404-9

PubMed Abstract | CrossRef Full Text | Google Scholar

Li, P., Rial, D., Canas, P. M., Yoo, J. H., Li, W., Zhou, X., et al. (2015). Optogenetic activation of intracellular adenosine A2A receptor signaling in the hippocampus is sufficient to trigger CREB phosphorylation and impair memory. Mol. Psychiatry 20, 1481. doi:10.1038/mp.2015.43

PubMed Abstract | CrossRef Full Text | Google Scholar

Lindsay, J., Laurin, D., Verreault, R., Hébert, R., Helliwell, B., Hill, G. B., et al. (2002). Risk factors for Alzheimer’s disease: a prospective analysis from the Canadian study of health and aging. Am. J. Epidemiol. 156, 445–453. doi:10.1093/aje/kwf074

PubMed Abstract | CrossRef Full Text | Google Scholar

Liu, Q. P., Wu, Y. F., Cheng, H. Y., Xia, T., Ding, H., Wang, H., et al. (2016). Habitual coffee consumption and risk of cognitive decline/dementia: a systematic review and meta-analysis of prospective cohort studies. Nutrition 32, 628–636. doi:10.1016/j.nut.2015.11.015

PubMed Abstract | CrossRef Full Text | Google Scholar

Livingston, J. M., McDonald, M. W., Gagnon, T., Jeffers, M. S., Gomez-Smith, M., Antonescu, S., et al. (2020). Influence of metabolic syndrome on cerebral perfusion and cognition. Neurobiol. Dis. 137, 104756. doi:10.1016/j.nbd.2020.104756

PubMed Abstract | CrossRef Full Text | Google Scholar

Long, J. M., and Holtzman, D. M. (2019). Alzheimer disease: an update on pathobiology and treatment strategies. Cell 179, 312–339. doi:10.1016/j.cell.2019.09.001

PubMed Abstract | CrossRef Full Text | Google Scholar

Lopes, J. P., Pliássova, A., and Cunha, R. A. (2019). The physiological effects of caffeine on synaptic transmission and plasticity in the mouse hippocampus selectively depend on adenosine A1 and A2A receptors. Biochem. Pharmacol. 166, 313–321. doi:10.1016/j.bcp.2019.06.008

PubMed Abstract | CrossRef Full Text | Google Scholar

Lopes, L. V., Cunha, R. A., Kull, B., Fredholm, B. B., and Ribeiro, J. A. (2002). Adenosine A2A receptor facilitation of hippocampal synaptic transmission is dependent on tonic A1 receptor inhibition. Neuroscience 112, 319–329. doi:10.1016/s0306-4522(02)00080-5

PubMed Abstract | CrossRef Full Text | Google Scholar

Lopes, L. V., Cunha, R. A., and Ribeiro, J. A. (1999). Increase in the number, G protein coupling, and efficiency of facilitatory adenosine A2A receptors in the limbic cortex, but not striatum, of aged rats. J. Neurochem. 73, 1733–1738. doi:10.1046/j.1471-4159.1999.731733.x

PubMed Abstract | CrossRef Full Text | Google Scholar

Lopes, L. V., Halldner, L., Rebola, N., Johansson, B., Ledent, C., Chen, J. F., et al. (2004). Binding of the prototypical adenosine A2A receptor agonist CGS 21680 to the cerebral cortex of adenosine A1 and A2A receptor knockout mice. Br. J. Pharmacol. 141, 1006–1014. doi:10.1038/sj.bjp.0705692

PubMed Abstract | CrossRef Full Text | Google Scholar

Lourida, I., Hannon, E., Littlejohns, T. J., Langa, K. M., Hyppönen, E., Kuzma, E., et al. (2019). Association of lifestyle and genetic risk with incidence of dementia. JAMA 322, 430–437. doi:10.1001/jama.2019.9879

CrossRef Full Text | Google Scholar

Maia, L., and de Mendonça, A. (2002). Does caffeine intake protect from Alzheimer’s disease?. Eur. J. Neurol. 9, 377–382. doi:10.1046/j.1468-1331.2002.00421.x

PubMed Abstract | CrossRef Full Text | Google Scholar

Marchi, M., Raiteri, L., Risso, F., Vallarino, A., Bonfanti, A., Monopoli, A., et al. (2002). Effects of adenosine A1 and A2A receptor activation on the evoked release of glutamate from rat cerebrocortical synaptosomes. Br. J. Pharmacol. 136, 434–440. doi:10.1038/sj.bjp.0704712

PubMed Abstract | CrossRef Full Text | Google Scholar

Mirra, S. S., Heyman, A., McKeel, D., Sumi, S. M., Crain, B. J., Brownlee, L. M., et al. (1991). The consortium to establish a registry for Alzheimer’s disease (CERAD). Part II. Standardization of the neuropathologic assessment of Alzheimer’s disease. Neurology 41, 479–486. doi:10.1212/wnl.41.4.479

PubMed Abstract | CrossRef Full Text | Google Scholar

Molinuevo, J. L., Ayton, S., Batrla, R., Bednar, M. M., Bittner, T., Cummings, J., et al. (2018). Current state of Alzheimer's fluid biomarkers. Acta Neuropathol. 136, 821–853. doi:10.1007/s00401-018-1932-x

PubMed Abstract | CrossRef Full Text | Google Scholar

Montine, T. J., Phelps, C. H., Beach, T. G., Bigio, E. H., Cairns, N. J., Dickson, D. W., et al. (2012). National institute on aging-Alzheimer’s association guidelines for the neuropathologic assessment of Alzheimer’s disease: a practical approach. Acta Neuropathol. 123, 1–11. doi:10.1007/s00401-011-0910-3

PubMed Abstract | CrossRef Full Text | Google Scholar

Moreira, A., Diógenes, M. J., de Mendonça, A., Lunet, N., and Barros, H. (2016). Chocolate consumption is associated with a lower risk of cognitive decline. J. Alzheimers Dis. 53, 85–93. doi:10.3233/JAD-160142

PubMed Abstract | CrossRef Full Text | Google Scholar

Mouro, F. M., Rombo, D. M., Dias, R. B., Ribeiro, J. A., and Sebastião, A. M. (2018). Adenosine A2A receptors facilitate synaptic NMDA currents in CA1 pyramidal neurons. Br. J. Pharmacol. 175, 4386–4397. doi:10.1111/bph.14497

PubMed Abstract | CrossRef Full Text | Google Scholar

Nortley, R., Korte, N., Izquierdo, P., Hirunpattarasilp, C., Mishra, A., Jaunmuktane, Z., et al. (2019). Amyloid β oligomers constrict human capillaries in Alzheimer’s disease via signaling to pericytes. Science 365, eaav9518. doi:10.1126/science.aav9518

PubMed Abstract | CrossRef Full Text | Google Scholar

Orr, A. G., Hsiao, E. C., Wang, M. M., Ho, K., Kim, D. H., Wang, X., et al. (2015). Astrocytic adenosine receptor A2A and Gs-coupled signaling regulate memory. Nat. Neurosci. 18, 423–434. doi:10.1038/nn.3930

PubMed Abstract | CrossRef Full Text | Google Scholar

Orr, A. G., Lo, I., Schumacher, H., Ho, K., Gill, M., Guo, W., et al. (2018). Istradefylline reduces memory deficits in aging mice with amyloid pathology. Neurobiol. Dis. 110, 29–36. doi:10.1016/j.nbd.2017.10.014

PubMed Abstract | CrossRef Full Text | Google Scholar

Pagnussat, N., Almeida, A. S., Marques, D. M., Nunes, F., Chenet, G. C., Botton, P. H., et al. (2015). Adenosine A2A receptors are necessary and sufficient to trigger memory impairment in adult mice. Br. J. Pharmacol. 172 (15), 3831–3845. doi:10.1111/bph.13180

PubMed Abstract | CrossRef Full Text | Google Scholar

Perez-Nievas, B. G., Stein, T. D., Tai, H. C., Dols-Icardo, O., Scotton, T. C., Barroeta-Espar, I., et al. (2013). Dissecting phenotypic traits linked to human resilience to Alzheimer’s pathology. Brain 136, 2510–2526. doi:10.1093/brain/awt171

PubMed Abstract | CrossRef Full Text | Google Scholar

Rebola, N., Sebastião, A. M., de Mendonca, A., Oliveira, C. R., Ribeiro, J. A., and Cunha, R. A. (2003). Enhanced adenosine A2A receptor facilitation of synaptic transmission in the hippocampus of aged rats. J. Neurophysiol. 90, 1295–1303. doi:10.1152/jn.00896.2002

PubMed Abstract | CrossRef Full Text | Google Scholar

Rebola, N., Canas, P. M., Oliveira, C. R., and Cunha, R. A. (2005). Different synaptic and subsynaptic localization of adenosine A2A receptors in the hippocampus and striatum of the rat. Neuroscience 132, 893–903. doi:10.1016/j.neuroscience.2005.01.014

PubMed Abstract | CrossRef Full Text | Google Scholar

Rebola, N., Lujan, R., Cunha, R. A., and Mulle, C. (2008). Adenosine A2A receptors are essential for long-term potentiation of NMDA-EPSCs at hippocampal mossy fiber synapses. Neuron 57, 121–134. doi:10.1016/j.neuron.2007.11.023

PubMed Abstract | CrossRef Full Text | Google Scholar

Rebola, N., Simões, A. P., Canas, P. M., Tomé, A. R., Andrade, G. M., Barry, C. E., et al. (2011). Adenosine A2A receptors control neuroinflammation and consequent hippocampal neuronal dysfunction. J. Neurochem. 117, 100–111. doi:10.1111/j.1471-4159.2011.07178.x

PubMed Abstract | CrossRef Full Text | Google Scholar

Reyes, C., and Cornelis, M. (2018). Caffeine in the diet: country-level consumption and guidelines. Nutrients 10, 1772. doi:10.3390/nu10111772

CrossRef Full Text | Google Scholar

Ritchie, K., Carrière, I., de Mendonca, A., Portet, F., Dartigues, J. F., Rouaud, O., et al. (2007). The neuroprotective effects of caffeine: a prospective population study (the three city study). Neurology 69, 536–545. doi:10.1212/01.wnl.0000266670.35219.0c

PubMed Abstract | CrossRef Full Text | Google Scholar

Sachdev, P. S., Mohan, A., Taylor, L., and Jeste, D. V. (2015). DSM-5 and mental disorders in older individuals: an overview. Harv. Rev. Psychiatry 23, 320–328. doi:10.1097/HRP.0000000000000090

PubMed Abstract | CrossRef Full Text | Google Scholar

Santiago, A. R., Baptista, F. I., Santos, P. F., Cristóvão, G., Ambrósio, A. F., Cunha, R. A., et al. (2014). Role of microglia adenosine A2A receptors in retinal and brain neurodegenerative diseases. Mediators Inflamm. 2014, 465694. doi:10.1155/2014/465694

PubMed Abstract | CrossRef Full Text | Google Scholar

Santos, C., Costa, J., Santos, J., Vaz-Carneiro, A., and Lunet, N. (2010). Caffeine intake and dementia: systematic review and meta-analysis. J. Alzheimers. Dis. 20 (Suppl. 1), S187–S204. doi:10.3233/JAD-2010-091387

PubMed Abstract | CrossRef Full Text | Google Scholar

Scheff, S. W., Price, D. A., Ansari, M. A., Roberts, K. N., Schmitt, F. A., Ikonomovic, M. D., et al. (2015). Synaptic change in the posterior cingulate gyrus in the progression of Alzheimer’s disease. J. Alzheimers Dis. 43, 1073–1090. doi:10.3233/JAD-141518

PubMed Abstract | CrossRef Full Text | Google Scholar

Scheff, S. W., Price, D. A., Schmitt, F. A., DeKosky, S. T., and Mufson, E. J. (2007). Synaptic alterations in CA1 in mild Alzheimer disease and mild cognitive impairment. Neurology 68, 1501–1508. doi:10.1212/01.wnl.0000260698.46517.8f

PubMed Abstract | CrossRef Full Text | Google Scholar

Selkoe, D. J. (2002). Alzheimer’s disease is a synaptic failure. Science 298, 789–791. doi:10.1126/science.1074069

PubMed Abstract | CrossRef Full Text | Google Scholar

Selkoe, D. J., and Hardy, J. (2016). The amyloid hypothesis of Alzheimer’s disease at 25 years. EMBO Mol. Med. 8, 595–608. doi:10.15252/emmm.201606210

PubMed Abstract | CrossRef Full Text | Google Scholar

Serrano-Pozo, A., Betensky, R. A., Frosch, M. P., and Hyman, B. T. (2016). Plaque-associated local toxicity increases over the clinical course of Alzheimer disease. Am. J. Pathol. 186, 375–384. doi:10.1016/j.ajpath.2015.10.010

PubMed Abstract | CrossRef Full Text | Google Scholar

Silva, A. C., Lemos, C., Gonçalves, F. Q., Pliássova, A. V., Machado, N. J., Silva, H. B., et al. (2018). Blockade of adenosine A2A receptors recovers early deficits of memory and plasticity in the triple transgenic mouse model of Alzheimer’s disease. Neurobiol. Dis. 117, 72–81. doi:10.1016/j.nbd.2018.05.024

PubMed Abstract | CrossRef Full Text | Google Scholar

Sinyor, B., Mineo, J., and Ochner, C. (2020). Alzheimer’s disease, inflammation, and the role of antioxidants. J. Alzheimers. Dis. Rep. 4, 175–183. doi:10.3233/ADR-200171

PubMed Abstract | CrossRef Full Text | Google Scholar

Sperling, R. A., Jack, C. R., and Aisen, P. S. (2011). Testing the right target and right drug at the right stage. Sci. Transl. Med. 3, 111cm33. doi:10.1126/scitranslmed.3002609

PubMed Abstract | CrossRef Full Text | Google Scholar

Sugiyama, K., Tomata, Y., Kaiho, Y., Honkura, K., Sugawara, Y., and Tsuji, I. (2016). Association between coffee consumption and incident risk of disabling dementia in elderly Japanese: the ohsaki cohort 2006 study. J. Alzheimers. Dis. 50, 491–500. doi:10.3233/JAD-150693

PubMed Abstract | CrossRef Full Text | Google Scholar

Tebano, M. T., Martire, A., Chiodi, V., Ferrante, A., and Popoli, P. (2010). Role of adenosine A2A receptors in modulating synaptic functions and brain levels of BDNF: a possible key mechanism in the pathophysiology of Huntington’s disease. Sci. World J. 10, 1768–1782. doi:10.1100/tsw.2010.164

CrossRef Full Text | Google Scholar

Temido-Ferreira, M., Coelho, J. E., Pousinha, P. A., and Lopes, L. V. (2019). Novel players in the aging synapse: impact on cognition. J. Caffeine Adenosine Res. 9, 104–127. doi:10.1089/caff.2019.0013

PubMed Abstract | CrossRef Full Text | Google Scholar

Temido-Ferreira, M., Ferreira, D. G., Batalha, V. L., Marques-Morgado, I., Coelho, J. E., Pereira, P., et al. (2020). Age-related shift in LTD is dependent on neuronal adenosine A2A receptors interplay with mGluR5 and NMDA receptors. Mol. Psychiatry 25, 1876–1900. doi:10.1038/s41380-018-0110-9

PubMed Abstract | CrossRef Full Text | Google Scholar

Terry, R. D., Masliah, E., Salmon, D. P., Butters, N., DeTeresa, R., Hill, R., et al. (1991). Physical basis of cognitive alterations in Alzheimer’s disease: synapse loss is the major correlate of cognitive impairment. Ann. Neurol. 30, 572–580. doi:10.1002/ana.410300410

PubMed Abstract | CrossRef Full Text | Google Scholar

Travassos, M., Santana, I., Baldeiras, I., Tsolaki, M., Gkatzima, O., Sermin, G., et al. (2015). Does caffeine consumption modify cerebrospinal fluid amyloid-β levels in patients with alzheimer's disease?. J. Alzheimers Dis. 47, 1069–1078. doi:10.3233/JAD-150374

PubMed Abstract | CrossRef Full Text | Google Scholar

van Gelder, B. M., Buijsse, B., Tijhuis, M., Kalmijn, S., Giampaoli, S., Nissinen, A., et al. (2007). Coffee consumption is inversely associated with cognitive decline in elderly European men: the FINE Study. Eur. J. Clin. Nutr. 61, 226–232. doi:10.1038/sj.ejcn.1602495

PubMed Abstract | CrossRef Full Text | Google Scholar

Vermunt, L., Sikkes, S. A. M., van den Hout, A., Handels, R., Bos, I., van der Flier, W. M., et al. (2019). Duration of preclinical, prodromal, and dementia stages of Alzheimer’s disease in relation to age, sex, and APOE genotype. Alzheimers Dement. 15, 888–898. doi:10.1016/j.jalz.2019.04.001

PubMed Abstract | CrossRef Full Text | Google Scholar

Viana da Silva, S., Haberl, M. G., Zhang, P., Bethge, P., Lemos, C., Gonçalves, N., et al. (2016). Early synaptic deficits in the APP/PS1 mouse model of Alzheimer’s disease involve neuronal adenosine A2A receptors. Nat. Commun. 7, 11915. doi:10.1038/ncomms11915

PubMed Abstract | CrossRef Full Text | Google Scholar

Williamson, J., Goldman, J., and Marder, K. S. (2009). Genetic aspects of Alzheimer disease. Neurologist 15, 80–86. doi:10.1097/NRL.0b013e318187e76b

PubMed Abstract | CrossRef Full Text | Google Scholar

Yu, L., Coelho, J. E., Zhang, X., Fu, Y., Tillman, A., Karaoz, U., et al. (2009). Uncovering multiple molecular targets for caffeine using a drug target validation strategy combining A2A receptor knockout mice with microarray profiling. Physiol. Genomics. 37, 199–210. doi:10.1152/physiolgenomics.90353.2008

PubMed Abstract | CrossRef Full Text | Google Scholar

Keywords: A2A adenosine receptor, Alzheimer’s disease, biomarker, neuroinflammation, therapeutic target

Citation: Merighi S, Poloni TE, Pelloni L, Pasquini S, Varani K, Vincenzi F, Borea PA and Gessi S (2021) An Open Question: Is the A2A Adenosine Receptor a Novel Target for Alzheimer’s Disease Treatment?. Front. Pharmacol. 12:652455. doi: 10.3389/fphar.2021.652455

Received: 14 January 2021; Accepted: 17 February 2021;
Published: 22 March 2021.

Edited by:

Francisco Ciruela, University of Barcelona, Spain

Reviewed by:

Yijuang Chern, National Research Program for Biopharmaceuticals, Taiwan
Rodrigo A. Cunha, University of Coimbra, Portugal

Copyright © 2021 Merighi, Poloni, Pelloni, Pasquini, Varani, Vincenzi, Borea and Gessi. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Stefania Merighi, mhs@unife.it; Stefania Gessi, gss@unife.it

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