Next Article in Journal
GDF-15 Deficiency Reduces Autophagic Activity in Human Macrophages In Vitro and Decreases p62-Accumulation in Atherosclerotic Lesions in Mice
Previous Article in Journal
Measurement of Chromosomal Arms and FISH Reveal Complex Genome Architecture and Standardized Karyotype of Model Fish, Genus Carassius
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

A2A Adenosine Receptor as a Potential Biomarker and a Possible Therapeutic Target in Alzheimer’s Disease

1
Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy
2
Department of Neurology & Neuropathology, Golgi-Cenci Foundation, 20081 Abbiategrasso, Italy
3
Department of Neurology, School of Medicine and Surgery, University of Milano-Bicocca, 20126 Monza, Italy
*
Authors to whom correspondence should be addressed.
Cells 2021, 10(9), 2344; https://doi.org/10.3390/cells10092344
Submission received: 28 July 2021 / Accepted: 31 August 2021 / Published: 7 September 2021
(This article belongs to the Section Cellular Pathology)

Abstract

:
Alzheimer’s disease (AD) is one of the most common neurodegenerative pathologies. Its incidence is in dramatic growth in Western societies and there is a need of both biomarkers to support the clinical diagnosis and drugs for the treatment of AD. The diagnostic criteria of AD are based on clinical data. However, it is necessary to develop biomarkers considering the neuropathology of AD. The A2A receptor, a G-protein coupled member of the P1 family of adenosine receptors, has different functions crucial for neurodegeneration. Its activation in the hippocampal region regulates synaptic plasticity and in particular glutamate release, NMDA receptor activation and calcium influx. Additionally, it exerts effects in neuroinflammation, regulating the secretion of pro-inflammatory cytokines. In AD patients, its expression is increased in the hippocampus/entorhinal cortex more than in the frontal cortex, a phenomenon not observed in age-matched control brains, indicating an association with AD pathology. It is upregulated in peripheral blood cells of patients affected by AD, thus reflecting its increase at central neuronal level. This review offers an overview on the main AD biomarkers and the potential role of A2A adenosine receptor as a new marker and therapeutic target.

Graphical Abstract

1. Alzheimer’s Disease

According to DSM-V, the mental deficit described as neurocognitive disorder (NCD) is caused by pathologies affecting neuronal circuits. Then, the type of underlying pathology defines the etiology of the disorder. While the early stages of NCD (mild-NCD/MCI) are characterized by functional preservation of everyday activities, major-NCD (dementia) has a functional impact on daily life [1].
The main age-related NCD is Alzheimer’s disease (AD); AD pathology progressively involves all cortical areas, and during its evolution, all cognitive domains are weakened with profound changes in behavior and functional abilities. Research over the last few decades has led to the discovery of some risk factors, including non-modifiable genetic factors (AD-related polymorphisms, APO-E4 allele, and pathogenic mutations in PSN-1-2 and APP genes) [2,3] and modifiable factors (favorable behaviors: regular physical and mental activity, healthy diet, high education and social engagement, and harmful conditions: midlife obesity, hypertension, diabetes, smoke, excessive alcohol and hearing loss) [4,5]. Nonetheless, the pathogenetic trajectory of AD is complex and individual, and largely unknown; hence, there is a need to assess the neuropathological picture and the related biomarkers to explore etiopathogenesis.
According to the amyloidogenic theory, the extensive presence of cortical amyloid and, therefore, of toxic β-amyloid oligomers induces a synaptic and neuronal dysfunction, mainly through TAU protein hyperphosphorylation, in turn causing synaptic collapse, fibers degeneration and neuronal loss. Furthermore, amyloid species seem to determine a reduction in the blood flow of brain capillaries and glial inflammatory activation [6,7]. Together, these neurodegenerative processes determine both macroscopic and microscopic brain changes. Typically, progressive brain atrophy is observed, starting in the parahippocampal cortex, hippocampus, medial and basal temporal lobe and parietal lobe, and spreading to the whole cortex in the advanced stages of the disease [8,9]. The underlying microscopic features are characterized by a dual proteinopathy, which consists of the deposition of amyloid and phosphorylated TAU (pTAU). The first proteinopathy is characterized by cortical plaques (β-amyloid or senile plaques) composed by an extracellular accumulation of β-amyloid peptides; the second one is represented by the hyperphosphorylated tau protein that aggregates inside the dying neuron, generating the so-called neurofibrillary tangles (NFT) and neuropil threads (NTs). The combination of the two proteinopathies constitutes the neuritic plaque (NP), which is the hallmark of AD neuropathology. Therefore, the aggregate scores for Amyloid (Thal stages), TAU (Braak stages) and NP (CERAD grading) constitute the ABC criteria for AD pathology, and define the neuropathological diagnosis of AD [10,11,12,13].

2. Biomarkers of AD

As stated previously, AD is defined as a clinical-pathologic entity, which is diagnosed during the course of life as possible or probable disease, and definitively at autopsy [14]. In order to comprehend the mechanism underlying the clinical expression of AD, a biological- as well as a syndrome-based definition is necessary. Furthermore, in order to identify therapies or interventions that prevent or delay the initial onset of symptoms, a biological-based definition including the preclinical phase of the disease is pivotal. Several studies are attempting to find novel biomarkers that reflect the biology of the disease, improving current diagnosis across the AD continuum [15].
Currently, various CSF and imaging biomarkers reflecting neuropathological changes are widely used in AD. An unbiased descriptive classification scheme for the biomarkers named “ATN” system was proposed; it includes seven major AD biomarkers divided in three categories based on the nature of the pathologic process measured by them. Biomarkers of β-amyloid plaques, labeled as “A”, are related to the extent of cortical amyloid deposition, and demonstrated by amyloid-PET or low CSF values of β-amyloid 42 (increased brain amyloid production and/or reduced amyloid “cleaning”). Biomarkers of pTAU, labeled as “T”, are related to cortical pTAU deposition and determined through TAU-PET or elevated CSF pTAU. Biomarkers of neurodegeneration and neuronal injury, labeled as “N”, are expressed by the demonstration of decreased synaptic activity through [(18)F]-fluorodeoxyglucose PET (FDG-PET hypometabolism), volume reduction in specific regions of interest (ROI) of the brain (atrophy on CT/MRI), or total-TAU release from dying neurons (increase in CSF total-TAU, as an effect of neuronal lysis). Each biomarker is evaluated positive or negative and, throughout their combination, the individual status of the biomarkers is defined; in this way, three categories are identifiable: (1) individuals with normal AD biomarkers; (2) those in the Alzheimer’s continuum, characterized by the presence of a significant amyloid load, and divided in amyloid deposition only, amyloid deposition and non-TAU neurodegeneration (suspected non-AD pathology), amyloid deposition and TAU pathology (early AD and AD); (3) non-Alzheimer’s neurodegenerative diseases, including those with no amyloid deposition (normal “A” biomarkers) but showing TAU pathology and/or neurodegeneration (abnormal “T” and/or “N”) [15,16], see Table 1.
The last category has been recently identified thanks to the use of biomarkers; it includes some forms of dementia of very elderly people with amnesic pictures, of varying severity, which mimic AD but have a different biological history. These pathologies mainly involve the temporo-mesial structures and are associated with the deposition of pTAU in the absence of amyloid, as occurs in primary TAUopathies such as primary age-related TAUopathy (PART) or argyrophilic grain disease [17], or with the presence of non-TAU neurodegeneration, as occurs in limbic-predominant age-related TDP-43 encephalopathy (LATE) [18].
The ATN system represents the set of markers most directly linked to the neuropathological picture and, therefore, constitutes the core biomarkers in neurodegeneration. However, the compensatory capacity of the brain and the cognitive reserve should be taken into consideration because protein deposits and atrophic aspects are linked to aging regardless of the presence of a clinically relevant disease, which can be prevented or delayed by the cognitive reserve. Indeed, the ATN system may have a high sensitivity but a low specificity, and, precisely for this reason, it should be interpreted in the light of the clinical and neuropsychological picture. For instance, according to the Cochrane review, amyloid-PET has a sensitivity of 95% and a specificity of 60%, while the FDG-PET has a sensitivity of 75% and a specificity of 85%. The combination of the two exams is more accurate but much more expensive; indeed, both are not recommended for routine diagnostics [19,20]. However, the sensitivity and specificity for AD diagnosis slightly improve using CSF to detect the ATN markers [21]. Therefore, the best diagnostic accuracy is obtained combining different assessments.
The “ATN” system utilizes expensive cerebral imaging or invasive procedures, such as lumbar puncture to obtain CSF. Thus, researchers are looking for AD biomarkers obtainable from more easily accessible biological fluids, such as plasma or serum. The most promising new AD fluid biomarkers include the plasmatic determination of neurofilaments, pTAU species, Aβ-amyloid oligomers (AβOs) and Aβ42/Aβ40 ratio; they might also be used as screening to select cases for further investigation through the ATN system. Overall, the reported sensitivity and specificity of these new fluid markers are similar, both being between 70% and 90% [22,23,24,25,26], but their diagnostic accuracy is still under investigation. The neurofilaments light (NfL) marker consists of light protein chains indicative of axonal damage, whose concentration is increased in the plasma samples of the early stage of AD and increased over time [22,27]. Assays have been developed for the detection of blood pTAU phosphorylated at threonine 181 (pTAU-181), which is increased along the AD continuum and allows the differentiation between AD and non-AD neurodegenerative diseases. Moreover, plasma pTAU-181 is strictly related to its increase in the CSF, and it predicts positive TAU-PET scans [23]. Besides pTAU-181, plasma levels of pTAU phosphorylated at threonine-217 (pTAU-217) is a new candidate tool as biomarker of AD; an increase occurs in the early stage of AD and correlates with worsening of cognition and brain atrophy [28]. Multimeric Detection System (MDS) is a new enzyme-linked immunosorbent assay used to detect AβOs selectively in the plasma of patients with AD. AβOs are the toxic forms of Aβ peptides and their plasma level is higher compared to controls without AD [24]. According to different studies, a significantly lower level of plasmatic Aβ42/Aβ40 ratio (TP42/40) was found in MCI patients compared to healthy controls. Furthermore, TP42/40 inversely correlates with the neocortical amyloid deposition evaluated with amyloid-PET, and was in accordance with the AD biomarkers in CSF [25,26].
Furthermore, Neurogranin (Ng) and inflammatory markers should be mentioned. Ng is a postsynaptic protein, which is increased in the CSF of patients with AD and is supposed to predict the decline in memory and executive function during the early stage of the disease [29]. Regarding inflammation and its important role in AD pathogenesis and its clinical worsening [30,31,32], it should be considered that there are no inflammatory markers in body fluids currently recognized as diagnostic for AD. Reports suggest a possible role of CSF proinflammatory cytokines levels, such as TNF-α, IL-1β or IL-6, as biomarkers of conversion of MCI to AD [33]. However, common inflammatory biomarkers are related to a state of systemic inflammation. Thus, the information obtained from these markers is non-specific and scarcely indicative of what happens in the brain tissue. Therefore, current efforts are moving towards specific markers of microglial/astroglial activation, such as YKL-40 (also known as chitinase-3-like protein), a tracking biomarker of astroglial-related neuroinflammation, which is increased in CSF of AD patients contributing to differentiate AD from non-AD pathologies [34,35], see Table 2.
Body fluid biomarkers play a continuously more important role in clinical trials; they may be used to assess the biological response to therapy and, thus, its efficacy on the course of the disease [36]. The recent approval of Aducanumab by the Food and Drug Administration has brought to the fore the importance of biomarkers to follow AD progression from a biological point of view. Indeed, Aducanumab appears to be more effective on biomarkers than on clinical manifestations, being very effective in removing amyloid and lowering pTAU, but with a modest clinical benefit, which remains uncertain in the long term [37,38,39]. This raises the still-unsolved problem of how much the clinical course of neurodegenerative diseases is predictable through the use of biomarkers. Given the pathogenetic complexity of these diseases, this is not surprising. On the other hand, the study of biomarkers provides indispensable interpretative keys. The study of adenosine receptors fits into this framework; they are very important in the functionality of the hippocampus, which is the nodal center for various neurodegenerative diseases, particularly for AD.

3. A2A Adenosine Receptors Biology

A2A receptors belong to the family of G-protein coupled purinergic P1 proteins, including four subtypes, named A1, A2A, A2B and A3, activated by the ubiquitous nucleoside adenosine deriving from ATP [40]. From a structural point of view, the A2A subtype has been cloned and pharmacologically characterized, and shows seven transmembrane domains connected to three extracellular and three intracellular loops [41]. It contains a long intracellular COOH terminus presenting sites for phosphorylation and palmitoylation that may affect the process of receptor desensitization and internalization. It is present on the cell surface not only as a monomer but also in association with other receptors, e.g., A1 adenosine and D2 dopamine subtypes, forming heteromers distinguished from homomers by different functional properties [42]. The A2A receptor localization affects striatum, the olfactory tubercle, and the immune system, presenting the highest expression, followed by the cerebral cortex, hippocampus, heart, lung, and vasculature. Specifically, as for neurons, astrocytes, microglia, and oligodendrocytes, they are present at pre- and postsynaptic level, regulating a series of effects associated to excitotoxicity, e.g., glutamate efflux, glial activation, and blood–brain barrier permeability, thus increasing leukocyte migration from the periphery. Concerning the peripheral immune system, A2A receptors are abundant in neutrophils, monocytes, macrophages, dendritic and T cells as well in platelets, and the blood vessels, where they mediate numerous antiinflammatory, antiaggregatory, and vasodilatory effects, respectively [43]. The A2A receptor signaling involves coupling to Gs and Golf proteins, in the periphery and brain, respectively, associated with adenylate cyclase and PKA stimulation leading to activation of several intracellular proteins [44,45]. In addition, it regulates MAPK signaling [46,47,48]. The ubiquitous nature of adenosine, of which the levels increase from nanomolar concentrations up to micromolar levels, following cellular and tissues damages, the wide distribution of A2A receptor and their upregulation mediated by injuries, renders this subtype an interesting target for several pathologies of both the central and peripheral nervous system, including neurodegenerative and inflammatory diseases as well as cancer.

4. Role of A2A Adenosine Receptors in AD

4.1. Neuronal Injury

Pioneering literature data report that loss of synaptic markers leading to synaptic dysfunction and degeneration is documented as one of the more important events correlated with cognitive impairment, before Aβ plaques and tangle formation [49]. More recently, it has been shown that the loss of synapses in the hippocampus and posterior cingulate gyrus is the first neuropathological alteration affecting brains of MCI and early AD patients and is an early process of memory alterations [50,51]. For this reason, AD has been defined as a synaptic-based disease, and the importance of saving synaptic structure and function has been underlined [52,53]. As for synaptic degeneration, a role for A2A adenosine receptors has been recognized, recently linking it in the pathogenesis of AD [54]. Indeed, hippocampal synapses present A2A adenosine receptors regulating synaptic plasticity (Figure 1) [55,56,57].
While A2A receptor hippocampal expression is generally protective, facilitating BDNF modulation of hippocampal synaptic transmission, in aging its overexpression takes place triggering deleterious synaptic effect causing an LTP-to-LTD shift and a reduction of hippocampal-dependent learning and memory processes. This was due to an increase A2A-mediated glutamate release, mGluR5-dependent NMDA receptor activation, and calcium influx from overexpressed voltage-dependent calcium channels [58,59,60,61]. A similar synaptic plasticity shift, A2A receptor-dependent, was observed in the hippocampus of aged and APP/PS1 animals [61,62]. It has been hypothesized that this phenomenon may be due to a shift in receptor cross-talk involving A2A-A1 heteromers, with the loss of A2A-mediated inhibition of presynaptic inhibitory A1 receptors in aged versus young rats and a direct facilitatory effect of A2A stimulation [63]. Specifically, an increase in A2A expression has been found in hippocampal neurons of aged or AD animal models as well as in astrocytes of AD patients and aged mice [62,64,65,66,67,68,69]. Interestingly, an increased hippocampal density of A2A receptors has also been observed in AD patients [61,70,71] (Table 3).
In addition, adenosine concentration is higher in parietal and temporal in comparison to the frontal cortex of post-mortem AD brains, thus suggesting an increased activation of A2A-upregulated receptors in these areas [78]. The hyperactivation of A2A adenosine receptors provokes memory disabilities, LTP damage, and alterations of synaptic markers [69].
A huge body of literature demonstrates the efficacy of A2A adenosine receptors antagonists, including caffeine, the world’s most popular psychoactive drug, to rescue synaptic damage and cognitive deficit in animal models of AD, proposing for them a role against synaptic toxicity [68,79,80,81,82,83]. Interestingly, chronic consumption of caffeine, or genetic deletion of A2A receptors, decreases TAU hyperphosphorylation in the hippocampus, reduces neuroinflammation, and contrasts related memory deficit. Accordingly, overexpression of A2A adenosine receptors increases TAU hyperphosphorylation and consequent TAU-dependent memory impairments in transgenic animal models of TAUopathy [84,85,86]. All these data are relevant because it is well known that tau pathology plays a role in memory impairment present in ageing and AD.

4.2. Neuroinflammation

Neuroinflammation includes a broad series of cellular effects in response to damage occurring in the nervous system as a consequence of ischemic insult, infection, and neurodegenerative pathologies. The main players of neuroinflammation are activated astrocytes and microglia that produce abnormal pro-inflammatory cytokines, such as TNF-α, IL-1β, and IFN-γ, and increase reactive oxygen and nitrogen species. It is accepted that neuroinflammation plays a crucial role in numerous neurodegenerative forms including AD, and represents an important aspect of aging that is the greatest risk factor for AD [87]. The function of cells involved in immune responses and inflammation is quite difficult to elucidate. On the one hand, microglial cells, under a moderate condition of activation, may positively destroy amyloid accumulation with beneficial effects. On the other hand, in the elderly, “inflammaging”, a chronic low-grade sterile inflammation occurring with age, induces uncontrolled production of inflammatory mediators. Indeed, this condition, characterized by a high level of cytokines, is associated with a decrease of cognition [88]. In this context, another crucial aspect of the A2A adenosine receptor activation in AD concerns its regulation of neuroinflammation. Indeed, it is present in both astrocytes and microglia, regulating the secretion of pro-inflammatory cytokines, as described below (Figure 1) [89].

4.2.1. Astrocytes

These cells include the majority of glial cells and are crucial in the regulation of brain homeostasis, due to their ability to affect synaptic plasticity, neuron metabolism, as well as ions and neurotransmitter homeostasis. Their pathological alteration is found in neurodegenerative conditions, including AD [90].
The first work documenting a role for A2A receptors in astrocytes showed that they were involved in the decrease of Aβ-triggered glutamate uptake, contributing to glutamatergic synaptic dysfunction and excitotoxicity in AD [91]. This reduction was associated with a depletion of the GLT-1 glutamate transporter, Na+/K+-ATPase-dependent, that is modulated by A2A. Other important astrocytic functions regulated by A2A receptors include calcium efflux from the endoplasmic reticulum, glutamate, and ATP release as well as GABA transport [92]. It is well known that memory is a process strictly influenced by astrocytes, where an A2A receptors upregulation occurs in aging human APP mice. In this AD animal model, the conditional genetic ablation of A2A receptor provides memory enhancement [68]. Accordingly, in amyloid plaque-bearing mice, administration of low doses of the A2A antagonist istradefylline increased spatial memory and habituation, providing evidence that A2A receptor blockers might be able to contrast memory deficits in AD patients [76]. The same effect was observed in the presence of an ENT1 inhibitor that increased adenosine and worsened memory dysfunction and neuronal plasticity in an APP/PS1 mouse model of AD [93]. Even though the pathophysiological consequence of this astrocytic A2A receptor increase deserves further investigations, recent data reported that A2A receptor overexpression induced important modifications, at transcriptional level, of genes involved in immune responses, angiogenesis, and cell activation [94].

4.2.2. Microglia

As the brain’s resident macrophages, these cells play essential functions in the modulation of cerebral activities, by eliminating dying neurons, deleting non-functional synapses, and producing molecules important for neuronal vitality. Microglia respond to neuronal activation and prevent excessive neurostimulation, exerting a fundamental protection of the brain from excessive activation like that occurring in AD [95].
Evaluation of post-mortem brains revealed an overexpression of A2A receptors only in microglia in proximity of pathological signs of AD but not in nondemented age-matched control brains [74]. Interestingly, it is known that neuroinflammation alone blocks neurogenesis and that contrasting inflammation reactivates this process, suggesting that the role of A2A receptor in the control of neuroinflammation might be a crucial mechanism in neurodegenerative diseases [96]. As the NMDA receptor, present in both neurons and microglia, is one of the principal targets to fight AD, it is relevant that it interacts with A2A receptors mainly in microglia. This interaction provides a novel functional complex, where A2A blockade is useful to hamper NMDA overactivation, useful in the protection of microglial cells. Interestingly, these interacting entities were upregulated in the hippocampal cells from the APPSw, Ind mice [97]. In general, activated microglia can present two opposite phenotypes, M1 promoting inflammation and cytotoxic effects and M2 fighting inflammation and providing neuroprotection [98]. It was found that cells from APP mice presented a relevant increase of a typical M1 marker, such as inducible nitric oxide synthase (iNOS), and in M2 marker arginase-1 (Arg-1). Interestingly, the A2A receptor blockade was able to decrease iNOS and increase Arg-1, providing evidence for a shift of microglia versus the beneficial M2 phenotype [97]. Accordingly, other works in animal models of neuroinflammation reported that overexpression of A2A receptors caused an increase in IL-1β, IL-6, TNF-α, and typical M1 microglial markers and that its block hampered LTP deficit in hippocampus [99]. Furthermore, A2A receptor constitutes heteromers with CB2 cannabinoid receptor subtypes, through which A2A receptor antagonists may increase both endo- and exo-cannabinoid effects, providing neuroprotection [77].

5. A2A Adenosine Receptor as a Novel Peripheral Biomarker in AD

Due to several pieces of evidence for the role of A2A receptors to trigger synaptic and cognitive problems, it has been suggested that it might be a good candidate biomarker of some chronic neurodegenerative diseases, including AD [100,101]. Its principal localization is the striatum important for locomotor activity, followed by the frontal cortex and hippocampus, crucial for memory and cognition. Specifically, in a study evaluating A2A expression in brain tissues from AD patients, an overexpression of them has been found in the hippocampus/entorhinal cortex in comparison with both frontal gray and white matter, which was not found in age-matched control brains, suggesting a relation with the presence of AD pathology [71]. Furthermore, the lowest A2A presence was detected in the frontal white matter, a region less affected by AD. In other words, the pattern of expression of A2A receptors seems to reflect the same distribution of AD pathology that affects the hippocampus/entorhinal cortex areas [12]. In any case, the finding of an A2A overexpression in the frontal white matter, rich in glial cells, supports a role for them in glial modifications that are found in AD [71,97]. In general, it is recognized that alterations occurring in CNS pathologies may be reflected at peripheral level in the blood, representing a useful and accessible substrate to evaluate proteins playing a crucial role in the pathology [102,103]. This happens, for example, in the field of adenosine receptors, in different diseases, such as heart and respiratory failure, Parkinson, and colon cancer [104,105,106,107]. As for AD, platelets showing biochemical activities in common with neurons, such as augmented β-secretase activity as well as amyloidogenic processing of amyloid protein precursor, may be used as a peripheral model for the study of cortical pathology [108]. Interestingly, very recently, it has been demonstrated that platelets from AD patients express a higher density of A2A receptors in comparison to platelets from control subjects not affected by dementia, thus providing evidence that this adenosine receptor subtype could mirror brain AD pathology in the periphery, revealing to be a promising indicator of disease [71]. These data are interesting in the diagnostic field of AD because they offer a peripheral marker that is cheap and easily accessible, through a blood withdrawal, reflecting changes in the CNS. According to the need for early diagnostic sentinels and therapeutic targets of disease, the A2A adenosine receptor may satisfy both these requirements, offering a novel opportunity to find and cure AD pathology. Similarly, previous works suggested the involvement of them at the beginning of AD, with their level being higher in blood cells from patients with mild cognitive impairment (MCI) with respect to healthy subjects [109,110]. This finding should be better investigated in future studies, comparing MCI and AD patients in the same setting at different stages of the pathology to gain information about the sensitivity of the A2A receptor as a biomarker of AD. In addition, other studies reported a lower amount of A2A mRNA in vascular dementia versus AD patients, but no difference between these ones and control subjects [111]. All these data suggest that A2A adenosine receptors may be differentially regulated in different forms of dementia, including vascular dementia and AD, probably depending on the origin/pathogenesis of the disease.
An important issue that needs to be addressed concerns the specificity of this adenosine receptor subtype as a potential biomarker of AD. Regarding specificity in particular, an upregulation of them was already found in lymphocytes from ischemic stroke patients [112,113,114], amyotrophic lateral sclerosis and multiple sclerosis [115,116], Huntington’s and Parkinson’s disease [106,117], chronic heart failure and cardiac transplantation [105], and in PMBCs from atrial fibrillation patients [118]. In spite of this wide overexpression of A2A receptors in lymphocytes and neutrophils of patients with pathologies of different nature and affecting different organs, the evidence related to their upregulation in AD has been relieved in platelets, and whether the same result is present in lymphocytes has yet to be determined.

6. A2A Adenosine Receptor as a Possible Therapeutic Target in AD

On the basis of the World Alzheimer Report, AD pathology is expected to rise up to more than 150 million in the next 30 years, starting from 50 million in 2018. This common type of dementia accounts for two-thirds of all dementia cases [119]. The most important risk factor for it is age, but others include both genetic and environmental factors, lifestyle, and cardiovascular pathologies. Despite many efforts employed in several years of research in disease-modifying drug development, there is no effective therapy to delay the beginning and the progression of AD. Today, the symptomatic therapies used to treat AD include nootropic molecules, reversible inhibitors of cholinesterase (donepezil or rivastigmine) improving memory and stabilizing patient’s behavior, and memantine (antagonist of NDMA receptors) producing positive effects in the cognition, behavior, and memory. Nutraceutics, containing a mix of omega-3, phospholipids, vitamins B12, B6, folate, Thiamine, vit. C and E, oligoelements, antioxidant agents (Curcumin), and coenzyme Q, are also prescribed. Unfortunately, there have been no new symptomatic drugs for 20 years, the last being memantine in 2002. A reason for this unsuccessful result may be found in the complex etiology and pathophysiology of AD.
Numerous efforts of several scientists around the world, working in the purinergic field, point to a role of A2A receptor antagonists in the therapy of different neurological ailments [88,120,121,122]. Specifically, clinical studies led to the development of the new first-in-class drug istradefylline, Nourias and Nourianz, in Japan and the US, respectively, for the treatment of Parkinson’s disease [98]. Interestingly, the therapeutic value of this molecule and of the whole class of A2A antagonists is not limited to improving locomotor disabilities and reducing the drawback of classical antiparkinson drugs, but also to ameliorate cognitive dysfunctions. Furthermore, their clinical development as new drugs for AD therapy may be fastened by the acquired knowledge that virtually all A2A blockers, tested in animal models and clinical studies, seem very safe [83].
Indeed, possible drawbacks related to the wide distribution of A2A receptors and their biological effects, including regulation of the immune system, inflammation, sleep, platelet aggregation and vasodilation, have not been revealed in clinical trials [123]. This is in agreement with the general safety of caffeine, by human assumption of 2–4 cup of coffee/daily, which was found able to reduce death for all causes by a recent meta-analysis [124]. Surprisingly, an exception to this safe profile of A2A antagonists is represented by tozadenant, having a different chemical structure with respect to istradefylline, which induced unfortunately fatal agranulocytosis in five patients in a clinical phase III trial with 409 participants [125]. However, additional experiments are necessary to clarify if these dramatic events were due to A2A blocking or other issue linked to its chemical structure.

7. Conclusions

Biomarkers are a primary tool in the development of AD research, but the interpretation of them should always be evaluated in light of the clinical picture. Since there are no disease-modifying therapies available on a large scale, and the diagnostic and prognostic value of biomarkers is not yet completely clear, inaccurate early diagnoses may cause deleterious psychic effects on the subjects under examination. However, beyond the early (pre-clinical) diagnosis of AD that poses such ethical problems, we believe it is possible to identify biological markers that allow us to follow the course of the disease even during its clinical phase, addressing symptomatic therapy in a personalized way. The possible use of the A2A receptor as a biomarker as well as a drug target fits into this framework. Indeed, all evidence remarqued in this review underline the importance of A2A receptor as a novel symptomatic drug target for AD treatment. Future clinical studies on AD patients, treated with istradefylline or with other old and new A2A antagonists, will be required, and the role of A2A receptor as biomarker may help to optimize patients’ enrollment and assumption protocol and dosages. In other words, molecular imaging of A2A receptors in the brain or its peripheral evaluation, as a specific and sensitive platelet biomarker, may help to select patients for which the drug would be more indicated, providing the desirable approach of a personalized medicine, with a reduction of costs and wait times. Notably, this research strategy for the discovery of a new therapy for AD is of particular importance, as it may positively impact the sustainability of the system, from which both public health and the economy could take advantage.

Author Contributions

S.G., T.E.P., G.N., K.V., S.P., F.V., P.A.B., and S.M. wrote the paper. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Conflicts 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

  1. Arlington, V.A.; American Psychiatric Association. Diagnostic and Statistical Manual of Mental Disorders, 5th ed.; American Psychiatric Publishing: Washington, DC, USA, 2013. [Google Scholar]
  2. Vermunt, L.; Sikkes, S.A.M.; van den Hout, A.; Handels, R.; Bos, I.; van der Flier, W.M.; Kern, S.; Ousset, P.J.; Maruff, P.; Skoog, I.; et al. Alzheimer Disease Neuroimaging Initiative; AIBL Research Group; ICTUS/DSA study groups. Duration of preclinical, prodromal, and dementia stages of Alzheimer’s disease in relation to age, sex, and APOE genotype. Alzheimers Dement. 2019, 15, 888–898. [Google Scholar] [CrossRef]
  3. Williamson, J.; Goldman, J.; Marder, K.S. Genetic aspects of Alzheimer disease. Neurologist 2009, 15, 80–86. [Google Scholar] [CrossRef] [Green Version]
  4. Livingston, J.M.; McDonald, M.W.; Gagnon, T.; Jeffers, M.S.; Gomez-Smith, M.; Antonescu, S.; Cron, G.O.; Boisvert, C.; Lacoste, B.; Corbett, D. Influence of metabolic syndrome on cerebral perfusion and cognition. Neurobiol. Dis. 2020, 137, 104756. [Google Scholar] [CrossRef] [PubMed]
  5. Lourida, I.; Hannon, E.; Littlejohns, T.J.; Langa, K.M.; Hyppönen, E.; Kuzma, E.; Llewellyn, D.J. Association of Lifestyle and Genetic Risk With Incidence of Dementia. JAMA 2019, 322, 430–437. [Google Scholar] [CrossRef]
  6. Selkoe, D.J.; Hardy, J. The amyloid hypothesis of Alzheimer’s disease at 25 years. EMBO Mol. Med. 2016, 8, 595–608. [Google Scholar] [CrossRef] [PubMed]
  7. Nortley, R.; Korte, N.; Izquierdo, P.; Hirunpattarasilp, C.; Mishra, A.; Jaunmuktane, Z.; Kyrargyri, V.; Pfeiffer, T.; Khennouf, L.; Madry, C.; et al. Amyloid b oligomers constrict human capillaries in Alzheimer’s disease via signaling to pericytes. Science 2019, 365, eaav9518. [Google Scholar] [CrossRef] [PubMed]
  8. Perez-Nievas, B.G.; Stein, T.D.; Tai, H.C.; Dols-Icardo, O.; Scotton, T.C.; Barroeta-Espar, I.; Fernandez-Carballo, L.; de Munain, E.L.; Perez, J.; Marquie, M.; et al. Dissecting phenotypic traits linked to human resilience to Alzheimer’s pathology. Brain 2013, 136, 2510–2526. [Google Scholar] [CrossRef]
  9. Jack, C.R., Jr.; Wiste, H.J.; Schwarz, C.G.; Lowe, V.J.; Senjem, M.L.; Vemuri, P.; Weigand, S.D.; Therneau, T.M.; Knopman, D.S.; Gunter, J.L.; et al. Longitudinal tau PET in ageing and Alzheimer’s disease. Brain 2018, 141, 1517–1528. [Google Scholar] [CrossRef] [Green Version]
  10. Mirra, S.S.; Heyman, A.; McKeel, D.; Sumi, S.M.; Crain, B.J.; Brownlee, L.M.; Vogel, F.S.; Hughes, J.P.; Belle, G.V.; Berg, L. The Consortium to Establish a Registry for Alzheimer’s Disease (CERAD): Part II. Standardization of the Neuropathologic Assessment of Alzheimer’s Disease. Neurology 1991, 41, 479. [Google Scholar] [CrossRef]
  11. Thal, D.R.; Rüb, U.; Orantes, M.; Braak, H. Phases of Aβ-deposition in the human brain and its relevance for the development of AD. Neurology 2002, 58, 1791–1800. [Google Scholar] [CrossRef]
  12. Braak, H.; Alafuzoff, I.; Arzberger, T.; Kretzschmar, H.; Del Tredici, K. Staging of Alzheimer disease-associated neurofibrillary pathology using paraffin sections and immunocytochemistry. Acta Neuropathol. 2006, 112, 389–404. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Montine, T.J.; Phelps, C.H.; Beach, T.G.; Bigio, E.H.; Cairns, N.J.; Dickson, D.W.; Duyckaerts, C.; Frosch, M.P.; Masliah, E.; Mirra, S.S.; et al. National Institute on Aging-Alzheimer’s Association guidelines for the neuropathologic assessment of Alzheimer’s disease: A practical approach. Acta Neuropathol. 2012, 123, 1–11. [Google Scholar] [CrossRef] [Green Version]
  14. McKhann, G.; Drachman, D.; Folstein, M.; Katzman, R.; Price, D.; Stadlan, E.M. Clinical Diagnosis of Alzheimer’s Disease: Report of the NINCDS-ADRDA Work Group under the Auspices of Department of Health and Human Services Task Force on Alzheimer’s Disease. Neurology 1984, 34, 939–944. [Google Scholar] [CrossRef] [Green Version]
  15. Jack, C.R., Jr.; Bennett, D.A.; Blennow, K.; Carrillo, M.C.; Dunn, B.; Haeberlein, S.B.; Holtzman, D.M.; Jagust, W.; Jessen, F.; Karlawish, J.; et al. NIA-AA Research Framework: Toward a biological definition of Alzheimer’s disease. Alzheimers Dement. 2018, 14, 535–562. [Google Scholar] [CrossRef] [PubMed]
  16. Jack, C.R., Jr.; Bennett, D.A.; Blennow, K.; Carrillo, M.C.; Feldman, H.H.; Frisoni, G.B.; Hampel, H.; Jagust, W.J.; Johnson, K.A.; Knopman, D.S.; et al. A/T/N: An unbiased descriptive classification scheme for Alzheimer disease biomarkers. Neurology 2016, 87, 539–547. [Google Scholar] [CrossRef] [PubMed]
  17. Crary, J.F.; Trojanowski, J.Q.; Schneider, J.A.; Abisambra, J.F.; Abner, E.L.; Alafuzoff, I.; Arnold, S.E.; Attems, J.; Beach, T.G.; Bigio, E.H.; et al. Primary age-related tauopathy (PART): A common pathology associated with human aging. Acta Neuropathol. 2014, 128, 755–766. [Google Scholar] [CrossRef] [Green Version]
  18. Nelson, P.T.; Dickson, D.W.; Trojanowski, J.Q.; Jack, C.R.; Boyle, P.A.; Arfanakis, K.; Rademakers, R.; Alafuzoff, I.; Attems, J.; Brayne, C.; et al. Limbic-predominant age-related TDP-43 encephalopathy (LATE): Consensus working group report. Brain 2019, 142, 1503–1527. [Google Scholar] [CrossRef] [Green Version]
  19. Smailagic, N.; Vacante, M.; Hyde, C.; Martin, S.; Ukoumunne, O.; Sachpekidis, C. ¹⁸F-FDG PET for the early diagnosis of Alzheimer’s disease dementia and other dementias in people with mild cognitive impairment (MCI). Cochrane Database Syst. Rev. 2015, 1, CD010632. [Google Scholar] [CrossRef]
  20. Zhang, S.; Smailagic, N.; Hyde, C.; Noel-Storr, A.H.; Takwoingi, Y.; McShane, R.; Feng, J. (11)C-PIB-PET for the early diagnosis of Alzheimer’s disease dementia and other dementias in people with mild cognitive impairment (MCI). Cochrane Database Syst. Rev. 2014, 7, CD010386. [Google Scholar] [CrossRef] [PubMed]
  21. Counts, S.E.; Ikonomovic, M.D.; Mercado, N.; Vega, I.E.; Mufson, E.J. Biomarkers for the Early Detection and Progression of Alzheimer’s Disease. Neurotherapeutics 2017, 14, 35–53. [Google Scholar] [CrossRef] [Green Version]
  22. Lewczuk, P.; Ermann, N.; Andreasson, U.; Schultheis, C.; Podhorna, J.; Spitzer, P.; Maler, J.M.; Kornhuber, J.; Blennow, K.; Zetterberg, H. Plasma neurofilament light as a potential biomarker of neurodegeneration in Alzheimer’s disease. Alzheimers Res. Ther. 2018, 10, 71. [Google Scholar] [CrossRef]
  23. Janelidze, S.; Mattsson, N.; Palmqvist, S.; Smith, R.; Beach, T.G.; Serrano, G.E.; Chai, X.; Proctor, N.K.; Eichenlaub, U.; Zetterberg, H.; et al. Plasma P-tau181 in Alzheimer’s disease: Relationship to other biomarkers, differential diagnosis, neuropathology and longitudinal progression to Alzheimer’s dementia. Nat. Med. 2020, 26, 379–386. [Google Scholar] [CrossRef]
  24. Wang, M.J.; Yi, S.; Han, J.Y.; Park, S.Y.; Jang, J.W.; Chun, I.K.; Kim, S.E.; Lee, B.S.; Kim, G.J.; Yu, J.S.; et al. Oligomeric forms of amyloid-β protein in plasma as a potential blood-based biomarker for Alzheimer’s disease. Alzheimer’s Res. Ther. 2017, 9, 98. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Fandos, N.; Pérez-Grijalba, V.; Pesini, P.; Olmos, S.; Bossa, M.; Villemagne, V.L.; Doecke, J.; Fowler, C.; Masters, C.L.; Sarasa, M. Plasma amyloid β 42/40 ratios as biomarkers for amyloid β cerebral deposition in cognitively normal individuals. Alzheimers Dement. 2017, 8, 179–187. [Google Scholar] [CrossRef] [PubMed]
  26. Pérez-Grijalba, V.; Arbizu, J.; Romero, J.; Prieto, E.; Pesini, P.; Sarasa, L.; Guillen, F.; Monleòn, I.; San-Josè, I.; Martìnez-Lage, P.; et al. Plasma Aβ42/40 ratio alone or combined with FDG-PET can accurately predict amyloid-PET positivity: A cross-sectional analysis from the AB255 Study. Alzheimer’s Res. Ther. 2019, 11, 96. [Google Scholar] [CrossRef] [PubMed]
  27. Mattsson, N.; Cullen, N.C.; Andreasson, U.; Zetterberg, H.; Blennow, K. Association Between Longitudinal Plasma Neurofilament Light and Neurodegeneration in Patients With Alzheimer Disease. JAMA Neurol. 2019, 76, 791–799. [Google Scholar] [CrossRef]
  28. Mattsson-Carlgren, N.; Janelidze, S.; Palmqvist, S.; Cullen, N.; Svenningsson, A.L.; Strandberg, O.; Mengel, D.; Walsh, D.M.; Stomrud, E.; Dage, J.L.; et al. Longitudinal plasma p-tau217 is increased in early stages of Alzheimer’s disease. Brain 2020, 143, 3234–3241. [Google Scholar] [CrossRef]
  29. Headley, A.; De Leon-Benedetti, A.; Dong, C.; Levin, B.; Loewenstein, D.; Camargo, C.; Rundek, T.; Zetterberg, H.; Blennow, K.; Wright, C.B.; et al. Alzheimer’s Disease Neuroimaging Initiative. Neurogranin as a predictor of memory and executive function decline in MCI patients. Neurology 2018, 90, e887–e895. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  30. Serrano-Pozo, A.; Betensky, R.A.; Frosch, M.P.; Hyman, B.T. Plaque-Associated Local Toxicity Increases over the Clinical Course of Alzheimer Disease. Am. J. Pathol. 2016, 186, 375–384. [Google Scholar] [CrossRef] [Green Version]
  31. Ising, C.; Venegas, C.; Zhang, S.; Scheiblich, H.; Schmidt, S.V.; Vieira-Saecker, A.; Schwartz, S.; Albasset, S.; McManus, R.M.; Tejera, D.; et al. NLRP3 inflammasome activation drives tau pathology. Nature 2019, 575, 669–673. [Google Scholar] [CrossRef]
  32. Poloni, T.E.; Medici, V.; Moretti, M.; Visonà, S.D.; Cirrincione, A.; Carlos, A.F.; Davin, A.; Gagliard, S.; Pansarasa, O.; Cereda, C.; et al. COVID-19-related neuropathology and microglial activation in elderly with and without dementia. Brain Pathol. 2021, 31, e12997. [Google Scholar] [CrossRef] [PubMed]
  33. Tarkowski, E.; Andreasen, N.; Tarkowski, A.; Blennow, K. Intrathecal inflammation precedes development of Alzheimer’s disease. J. Neurol. Neurosurg. Psychiatry 2003, 74, 1200–1205. [Google Scholar] [CrossRef] [PubMed]
  34. Muszyński, P.; Groblewska, M.; Kulczyńska-Przybik, A.; Kułakowska, A.; Mroczko, B. YKL-40 as a Potential Biomarker and a Possible Target in Therapeutic Strategies of Alzheimer’s Disease. Curr. Neuropharmacol. 2017, 15, 906–917. [Google Scholar] [CrossRef] [Green Version]
  35. Olsson, B.; Hertze, J.; Lautner, R.; Zetterberg, H.; Nägga, K.; Hoglund, K.; Basun, H.; Annas, P.; Lannfelt, L.; Andreasen, N.; et al. Microglial Markers are Elevated in the Prodromal Phase of Alzheimer’s Disease and Vascular Dementia. J. Alzheimer’s Dis. 2013, 33, 45–53. [Google Scholar] [CrossRef]
  36. Zetterberg, H.; Bendlin, B.B. Biomarkers for Alzheimer’s disease-preparing for a new era of disease-modifying therapies. Mol. Psychiatry 2021, 26, 296–308. [Google Scholar] [CrossRef]
  37. Mullard, A. FDA approval for Biogen’s aducanumab sparks Alzheimer disease firestorm. Nat. Rev. Drug Discov. 2021, 20, 496. [Google Scholar]
  38. Sabbagh, M.N.; Cummings, J. Open Peer Commentary to “Failure to demonstrate efficacy of aducanumab: An analysis of the EMERGE and ENGAGE Trials as reported by Biogen December 2019”. Alzheimers Dement. 2021, 17, 702–703. [Google Scholar] [CrossRef] [PubMed]
  39. Knopman, D.S.; Jones, D.T.; Greicius, M.D. Failure to demonstrate efficacy of aducanumab: An analysis of the EMERGE and ENGAGE trials as reported by Biogen, December 2019. Alzheimers Dement. 2021, 17, 696–701. [Google Scholar] [CrossRef]
  40. Borea, P.A.; Gessi, S.; Merighi, S.; Vincenzi, F.; Varani, K. Pharmacology of Adenosine Receptors: The State of the Art. Physiol. Rev. 2018, 98, 1591–1625. [Google Scholar] [CrossRef] [PubMed]
  41. Fredholm, B.B.; Arslan, G.; Halldner, L.; Kull, B.; Schulte, G.; Wasserman, W. Structure and function of adenosine receptors and their genes. Naunyn Schmiedebergs Arch. Pharmacol. 2000, 362, 364–374. [Google Scholar] [CrossRef]
  42. Ferré, S.; Navarro, G.; Casadó, V.; Cortés, A.; Mallol, J.; Canela, E.I.; Lluís, C.; Franco, R. G protein-coupled receptor heteromers as new targets for drug development. Prog. Mol. Biol. Transl. Sci. 2010, 91, 41–52. [Google Scholar]
  43. Antonioli, L.; Fornai, M.; Blandizzi, C.; Pacher, P.; Haskó, G. Adenosine signaling and the immune system: When a lot could be too much. Immunol. Lett. 2019, 205, 9–15. [Google Scholar] [CrossRef] [PubMed]
  44. Kull, B.; Svenningsson, P.; Fredholm, B.B. Adenosine A(2A) receptors are colocalized with and activate g(olf) in rat striatum. Mol. Pharmacol. 2000, 58, 771–777. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Preti, D.; Baraldi, P.G.; Moorman, A.R.; Borea, P.A.; Varani, K. History and perspectives of A2A adenosine receptor antagonists as potential therapeutic agents. Med. Res. Rev. 2015, 35, 790–848. [Google Scholar] [CrossRef]
  46. Baraldi, P.G.; Tabrizi, M.A.; Gessi, S.; Borea, P.A. Adenosine receptor antagonists: Translating medicinal chemistry and pharmacology into clinical utility. Chem. Rev. 2008, 108, 238–263. [Google Scholar] [CrossRef]
  47. Chen, J.F.; Eltzschig, H.K.; Fredholm, B.B. Adenosine receptors as drug targets–what are the challenges? Nat. Rev. Drug Discov. 2013, 12, 265–286. [Google Scholar] [CrossRef] [Green Version]
  48. Schulte, G.; Fredholm, B.B. Signalling from adenosine receptors to mitogen-activated protein kinases. Cell Signal 2003, 15, 813–827. [Google Scholar] [CrossRef]
  49. Terry, R.D.; Masliah, E.; Salmon, D.P.; Butters, N.; DeTeresa, R.; Hill, R.; Hansen, L.A.; Katzman, R. Physical basis of cognitive alterations in Alzheimer’s disease: Synapse loss is the major correlate of cognitive impairment. Ann. Neurol. 1991, 30, 572–580. [Google Scholar] [CrossRef]
  50. Scheff, S.W.; Price, D.A.; Schmitt, F.A.; DeKosky, S.T.; Mufson, E.J. Synaptic alterations in CA1 in mild Alzheimer disease and mild cognitive impairment. Neurology 2007, 68, 1501–1508. [Google Scholar] [CrossRef] [PubMed]
  51. Scheff, S.W.; Price, D.A.; Ansari, M.A.; Roberts, K.N.; Schmitt, F.A.; Ikonomovic, M.D.; Mufson, E.J. Synaptic change in the posterior cingulate gyrus in the progression of Alzheimer’s disease. J. Alzheimers Dis. 2015, 43, 1073–1090. [Google Scholar] [CrossRef] [Green Version]
  52. Selkoe, D.J. Alzheimer’s disease is a synaptic failure. Science 2002, 298, 789–791. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Coleman, P.; Federoff, H.; Kurlan, R. A focus on the synapse for neuroprotection in Alzheimer disease and other dementias. Neurology 2004, 63, 1155–1162. [Google Scholar] [CrossRef]
  54. Horgusluoglu-Moloch, E.; Nho, K.; Risacher, S.L.; Kim, S.; Foroud, T.; Shaw, L.M.; Trojanowski, J.Q.; Aisen, P.S.; Petersen, R.C.; Jack, C.R.; et al. Targeted neurogenesis pathway-based gene analysis identifies ADORA2A associated with hippocampal volume in mild cognitive impairment and Alzheimer’s disease. Neurobiol. Aging 2017, 60, 92–103. [Google Scholar] [CrossRef] [Green Version]
  55. Rebola, N.; Canas, P.M.; Oliveira, C.R.; Cunha, R.A. Different synaptic and subsynaptic localization of adenosine A2A receptors in the hippocampus and striatum of the rat. Neuroscience 2005, 132, 893–903. [Google Scholar] [CrossRef]
  56. Rebola, N.; Lujan, R.; Cunha, R.A.; Mulle, C. Adenosine A2A receptors are essential for long-term potentiation of NMDA-EPSCs at hippocampal mossy fiber synapses. Neuron 2008, 57, 121–134. [Google Scholar] [CrossRef] [Green Version]
  57. Costenla, A.R.; Diógenes, M.J.; Canas, P.M.; Rodrigues, R.J.; Nogueira, C.; Maroco, J.; Agostinho, P.M.; Ribeiro, J.A.; Cunha, R.A.; De Mendonça, A. Enhanced role of adenosine A(2A) receptors in the modulation of LTP in the rat hippocampus upon ageing. Eur. J. Neurosci. 2011, 34, 12–21. [Google Scholar] [CrossRef]
  58. Tebano, M.T.; Martire, A.; Rebola, N.; Pepponi, R.; Domenici, M.R.; Gro, M.C.; Schwarzschild, M.A.; Chen, J.F.; Cunha, R.A.; PopoliAdenosine, P. A2A receptors and metabotropic glutamate 5 receptors are co-localized and functionally interact in the hippocampus: A possible key mechanism in the modulation of N-methyl-D-aspartate effects. J. Neurochem. 2005, 95, 1188–1200. [Google Scholar] [CrossRef] [Green Version]
  59. Tebano, M.T.; Martire, A.; Chiodi, V.; Ferrante, A.; Popoli, P. Role of adenosine A(2A) receptors in modulating synaptic functions and brain levels of BDNF: A possible key mechanism in the pathophysiology of Huntington’s disease. Sci. World J. 2010, 10, 1768–1782. [Google Scholar] [CrossRef] [Green Version]
  60. Temido-Ferreira, M.; Coelho, J.E.; Pousinha, P.A.; Lopes, L.V. Novel players in the aging synapse: Impact on cognition. J. Caffeine Adenosine Res. 2019, 9, 104–127. [Google Scholar] [CrossRef] [PubMed]
  61. Temido-Ferreira, M.; Ferreira, D.G.; Batalha, V.L.; Marques-Morgado, I.; Coelho, J.E.; Pereira, P.; Gomes, R.; Pinto, A.; Carvalho, S.; Canas, P.M.; et al. Age-related shift in LTD is dependent on neuronal adenosine A 2A receptors interplay with mGluR5 and NMDA receptors. Mol. Psychiatry 2020, 25, 1876–1900. [Google Scholar] [CrossRef] [PubMed]
  62. Viana da Silva, S.; Haberl, M.G.; Zhang, P.; Bethge, P.; Lemos, C.; Gonçalves, N.; Gorlewicz, A.; Malezieux, M.; Gonçalves, F.Q.; Grosjean, N.; et al. Early synaptic deficits in the APP/PS1 mouse model of Alzheimer’s disease involve neuronal adenosine A2A receptors. Nat. Commun. 2016, 7, 11915. [Google Scholar] [CrossRef]
  63. Lopes, L.V.; Cunha, R.A.; Ribeiro, J.A. Cross talk between A(1) and A(2A) adenosine receptors in the hippocampus and cortex of young adult and old rats. J. Neurophysiol. 1999, 82, 3196–3203. [Google Scholar] [CrossRef]
  64. Lopes, L.V.; Cunha, R.A.; Ribeiro, J.A. Increase in the Number, G Protein Coupling, and Efficiency of Facilitatory Adenosine A2A Receptors in the Limbic Cortex, but not Striatum, of Aged Rats. J. Neurochem. 1999, 73, 1733–1738. [Google Scholar] [CrossRef]
  65. Arendash, G.W.; Schleif, W.; Rezai-Zadeh, K.; Jackson, E.K.; Zacharia, L.C.; Cracchiolo, J.R.; Shippy, D.; Tan, J. Caffeine protects Alzheimer’s mce against cognitive impairment and reduces brain beta-amyloid production. Neuroscience 2006, 142, 941–952. [Google Scholar] [CrossRef]
  66. Canas, P.M.; Duarte, J.M.; Rodrigues, R.J.; Köfalvi, A.; Cunha, R.A. Modification upon aging of the density of presynaptic modulation systems in the hippocampus. Neurobiol. Aging 2009, 30, 1877–1884. [Google Scholar] [CrossRef] [Green Version]
  67. Espinosa, J.; Rocha, A.; Nunes, F.; Costa, M.S.; Schein, V.; Kazlauckas, V.; Kalinine, E.; Souza, D.O.; Rodrigo, A.; Cunha, R.A.; et al. 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. 2013, 34, 509–518. [Google Scholar] [CrossRef]
  68. Orr, A.G.; Hsiao, E.C.; Wang, M.M.; Ho, K.; Kim, D.H.; Wang, X.; Guo, W.; Kang, J.; Yu, G.Q.; Adame, A.; et al. Astrocytic adenosine receptor A2A and Gs-coupled signaling regulate memory. Nat. Neurosci. 2015, 18, 423–434. [Google Scholar] [CrossRef] [Green Version]
  69. Gonçalves, F.Q.; Lopes, J.P.; Silva, H.B.; Lemos, C.; Silva, A.C.; Gonçalves, N.; Tomé, A.R.; Ferreira, S.G.; Canas, P.M.; Rial, D.; et al. 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. 2019, 132, 104570. [Google Scholar] [CrossRef]
  70. Albasanz, J.L.; Perez, S.; Barrachina, M.; Ferrer, I.; Martín, M. Up-regulation of adenosine receptors in the frontal cortex in Alzheimer’s disease. Brain Pathol. 2008, 18, 211–219. [Google Scholar] [CrossRef]
  71. Merighi, S.; Battistello, E.; Casetta, I.; Gragnaniello, D.; Poloni, T.E.; Medici, V.; Cirrincione, A.; Varani, K.; Vincenzi, F.; Borea, P.A.; et al. Upregulation of Cortical A2A Adenosine Receptors Is Reflected in Platelets of Patients with Alzheimer’s Disease. J. Alzheimers Dis. 2021, 80, 1105–1117. [Google Scholar] [CrossRef]
  72. Cunha, R.A.; Constantino, M.C.; Sebastião, A.M.; Ribeiro, J.A. Modification of A1 and A2a adenosine receptor binding in aged striatum, hippocampus and cortex of the rat. Neuroreport 1995, 6, 1583–1588. [Google Scholar] [CrossRef]
  73. Rebola, N.; Sebastião, A.M.; de Mendonca, A.; Oliveira, C.R.; Ribeiro, J.A.; Cunha, R.A. Enhanced adenosine A2A receptor facilitation of synaptic transmission in the hippocampus of aged rats. J. Neurophysiol. 2003, 90, 1295–1303. [Google Scholar] [CrossRef]
  74. Angulo, E.; Casadó, V.; Mallol, J.; Canela, E.I.; Viñals, F.; Ferrer, I.; 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. [Google Scholar] [CrossRef] [PubMed]
  75. Diógenes, M.J.; Assaife-Lopes, N.; Pinto-Duarte, A.; Ribeiro, J.A.; Sebastião, A.M. Influence of age on BDNF modulation of hippocampal synaptic transmission: Interplay with adenosine A2A receptors. Hippocampus 2007, 17, 577–585. [Google Scholar] [CrossRef]
  76. Orr, A.G.; Lo, I.; Schumacher, H.; Ho, K.; Gill, M.; Guo, W.; Kim, D.H.; Knox, A.; Saito, T.; Saido, T.C.; et al. Istradefylline reduces memory deficits in aging mice with amyloid pathology. Neurobiol. Dis. 2018, 110, 29–36. [Google Scholar] [CrossRef] [PubMed]
  77. Franco, R.; Reyes-Resina, I.; Aguinaga, D.; Lillo, A.; Jiménez, J.; Raïch, I.; Borroto-Escuela, D.O.; Ferreiro-Vera, C.; Canela, E.I.; Sánchez de Medina, V.; et al. Potentiation of cannabinoid signaling in microglia by adenosine A2A receptor antagonists. Glia 2019, 67, 2410–2423. [Google Scholar] [CrossRef]
  78. Alonso-Andrés, P.; Albasanz, J.L.; Ferrer, I.; Martín, M. Purine-related metabolites and their converting enzymes are altered in frontal, parietal and temporal cortex at early stages of Alzheimer’s disease pathology. Brain Pathol. 2018, 28, 933–946. [Google Scholar] [CrossRef] [Green Version]
  79. Arendash, G.W.; Cao, C. Caffeine and coffee as therapeutics against Alzheimer’s disease. J. Alzheimers Dis. 2010, 20 (Suppl. S1), S117–S126. [Google Scholar] [CrossRef] [Green Version]
  80. Dall’Igna, O.P.; Fett, P.; Gomes, M.W.; Souza, D.O.; Cunha, R.A.; Lara, D.R. Caffeine and adenosine A(2a) receptor antagonists prevent beta-amyloid (25-35)-induced cognitive deficits in mice. Exp. Neurol. 2007, 203, 241–245. [Google Scholar] [CrossRef] [PubMed]
  81. Canas, P.M.; Porciúncula, L.O.; Cunha, G.M.; Silva, C.G.; Machado, N.J.; Oliveira, J.M.; Oliveira, C.R.; Cunha, R.A. Adenosine A2A receptor blockade prevents synaptotoxicity and memory dysfunction caused by beta-amyloid peptides via p38 mitogen-activated protein kinase pathway. J. Neurosci. 2009, 29, 14741–14751. [Google Scholar] [CrossRef]
  82. Faivre, E.; Coelho, J.E.; Zornbach, K.; Malik, E.; Baqi, Y.; Schneider, M.; Cellai, L.; Carvalho, K.; Sebda, S.; Figeac, M.; et al. Beneficial Effect of a Selective Adenosine A2A Receptor Antagonist in the APPswe/PS1dE9 Mouse Model of Alzheimer’s Disease. Front. Mol. Neurosci. 2018, 11, 235. [Google Scholar] [CrossRef]
  83. Jacobson, K.A.; Gao, Z.G.; Matricon, P.; Eddy, M.T.; Carlsson, J. Adenosine A2A receptor antagonists: From caffeine to selective non-xanthines. Br. J. Pharmacol. 2020, 1–16. [Google Scholar] [CrossRef]
  84. Laurent, C.; Eddarkaoui, S.; Derisbourg, M.; Leboucher, A.; Demeyer, D.; Carrier, S.; Schneider, M.; Hamdane, M.; Müller, C.E.; Buée, L.; et al. Beneficial effects of caffeine in a transgenic model of Alzheimer’s disease-like tau pathology. Neurobiol. Aging 2014, 35, 2079–2090. [Google Scholar] [CrossRef]
  85. Laurent, C.; Burnouf, S.; Ferry, B.; Batalha, V.L.; Coelho, J.E.; Baqi, Y.; Malik, E.; Mariciniak, E.; Parrot, S.; Van der Jeugd, A.; et al. A2A adenosine receptor deletion is protective in a mouse model of Tauopathy. Mol. Psychiatry 2016, 21, 97–107. [Google Scholar] [CrossRef] [Green Version]
  86. Carvalho, K.; Faivre, E.; Pietrowski, M.J.; Marques, X.; Gomez-Murcia, V.; Deleau, A.; Huin, V.; Hansen, J.N.; Kozlov, S.; Danis, C.; et al. Exacerbation of C1q dysregulation, synaptic loss and memory deficits in tau pathology linked to neuronal adenosine A2A receptor. Brain 2019, 142, 3636–3654. [Google Scholar] [CrossRef]
  87. Martí Navia, A.; Dal Ben, D.; Lambertucci, C.; Spinaci, A.; Volpini, R.; Marques-Morgado, I.; Coelho, J.E.; Lopes, L.V.; Marucci, G.; Buccioni, M. Adenosine Receptors as Neuroinflammation Modulators: Role of A(1) Agonists and A(2A) Antagonists. Cells 2020, 9, 1739. [Google Scholar] [CrossRef]
  88. Chambers, E.S.; Akbar, A.N. Can blocking inflammation enhance immunity during aging? J. Allergy Clin. Immunol. 2020, 145, 1323–1331. [Google Scholar] [CrossRef]
  89. Illes, P.; Rubini, P.; Ulrich, H.; Zhao, Y.; Tang, Y. Regulation of Microglial Functions by Purinergic Mechanisms in the Healthy and Diseased CNS. Cells 2020, 9, 1108. [Google Scholar] [CrossRef]
  90. Verkhratsky, A.; Nedergaard, M. Physiology of Astroglia. Physiol. Rev. 2018, 98, 239–389. [Google Scholar] [CrossRef] [PubMed]
  91. Matos, M.; Augusto, E.; Machado, N.J.; Dos Santos-Rodrigues, A.; Cunha, R.A.; Agostinho, P. Astrocytic adenosine A2A receptors control the amyloid-beta peptide-induced decrease of glutamate uptake. J. Alzheimers Dis. 2012, 31, 555–567. [Google Scholar] [CrossRef]
  92. Lopes, C.R.; Cunha, R.A.; Agostinho, P. Astrocytes and Adenosine A(2A) Receptors: Active Players in Alzheimer’s Disease. Front. Neurosci. 2021, 15, 666710. [Google Scholar] [CrossRef]
  93. Lee, C.C.; Chang, C.P.; Lin, C.J.; Lai, H.L.; Kao, Y.H.; Cheng, S.J.; Chen, H.M.; Liao, Y.P.; Faivre, E.; Buée, L.; et al. Adenosine Augmentation Evoked by an ENT1 Inhibitor Improves Memory Impairment and Neuronal Plasticity in the APP/PS1 Mouse Model of Alzheimer’s Disease. Mol. Neurobiol. 2018, 55, 8936–8952. [Google Scholar] [CrossRef]
  94. Paiva, I.; Carvalho, K.; Santos, P.; Cellai, L.; Pavlou, M.A.S.; Jain, G.; Gnad, T.; Pfeifer, A.; Vieau, D.; Fischer, A.; et al. A2AR-induced transcriptional deregulation in astrocytes: An in vitro study. Glia 2019, 67, 2329–2342. [Google Scholar] [CrossRef]
  95. Badimon, A.; Strasburger, H.J.; Ayata, P.; Chen, X.; Nair, A.; Ikegami, A.; Hwang, P.; Chan, A.T.; Graves, S.M.; Uweru, J.O.; et al. Negative feedback control of neuronal activity by microglia. Nature 2020, 586, 417–423. [Google Scholar] [CrossRef]
  96. Monje, M.L.; Toda, H.; Palmer, T.D. Inflammatory blockade restores adult hippocampal neurogenesis. Science 2003, 302, 1760–1765. [Google Scholar] [CrossRef]
  97. Franco, R.; Rivas-Santisteban, R.; Casanovas, M.; Lillo, A.; Saura, C.A.; Navarro, G. Adenosine A(2A) receptor antagonists affects NMDA glutamate receptor function. Potential to address neurodegeneration in Alzheimer’s disease. Cells 2020, 9, 1075. [Google Scholar] [CrossRef]
  98. Franco, R.; Lillo, A.; Rivas-Santisteban, R.; Reyes-Resina, I.; Navarro, G. Microglial Adenosine Receptors: From Preconditioning to Modulating the M1/M2 Balance in Activated Cells. Cells 2021, 10, 1124. [Google Scholar] [CrossRef] [PubMed]
  99. Colella, M.; Zinni, M.; Pansiot, J.; Cassanello, M.; Mairesse, J.; Ramenghi, L.; Baud, O. Modulation of Microglial Activation by Adenosine A2a Receptor in Animal Models of Perinatal Brain Injury. Front. Neurol. 2018, 9, 605. [Google Scholar] [CrossRef] [PubMed]
  100. Borea, P.A.; Gessi, S.; Merighi, S.; Varani, K. Adenosine as a multisignalling guardian angel in human diseases: When, where and how does it exert its protective effects? Trends Pharmacol. Sci. 2016, 37, 419–434. [Google Scholar] [CrossRef]
  101. Borea, P.A.; Gessi, S.; Merighi, S.; Vincenzi, F.; Varani, K. Pathological overproduction: The bad side of adenosine. Br. J. Pharmacol. 2017, 174, 1945–1960. [Google Scholar] [CrossRef] [Green Version]
  102. Brodde, O.E.; Beckering, J.J.; Michel, M.C. Human heart β-adrenoceptors: A fair comparison with lymphocyte β-adrenoceptors? Trends Pharmacol. Sci. 1987, 8, 403–407. [Google Scholar] [CrossRef]
  103. Nijhuis, E.W.; Oostervink, F.; Hinloopen, B.; Rozing, J.; Nagelkerken, L. Differences in dexamethasone-sensitivity between lymphocytes from patients with Alzheimer’s disease and patients with multi-infarct dementia. Brain Behav. Immun. 1996, 10, 115–125. [Google Scholar] [CrossRef]
  104. Varani, K.; Caramori, G.; Vincenzi, F.; Adcock, I.; Casolari, P.; Leung, E.; Maclennan, S.; Gessi, S.; Morello, S.; Barnes, P.J.; et al. Alteration of adenosine receptors in patients with chronic obstructive pulmonary disease. Am. J. Respir. Crit. Care Med. 2006, 173, 398–406. [Google Scholar] [CrossRef]
  105. Varani, K.; Laghi-Pasini, F.; Camurri, A.; Capecchi, P.L.; Maccherini, M.; Diciolla, F.; Ceccatelli, L.; Lazzerini, P.E.; Ulouglu, C.; Cattabeni, F.; et al. Changes of peripheral A2A adenosine receptors in chronic heart failure and cardiac transplantation. FASEB J. 2003, 17, 280–282. [Google Scholar] [CrossRef] [Green Version]
  106. Varani, K.; Vincenzi, F.; Tosi, A.; Gessi, S.; Casetta, I.; Granieri, G.; Fazio, P.; Leung, E.; MacLennan, S.; Granieri, E.; et al. A2A adenosine receptor overexpression and functionality, as well as TNF-alpha levels, correlate with motor symptoms in Parkinson’s disease. FASEB J. 2010, 24, 587–598. [Google Scholar] [CrossRef]
  107. Gessi, S.; Cattabriga, E.; Avitabile, A.; Gafa’, R.; Lanza, G.; Cavazzini, L.; Bianchi, N.; Gambari, R.; Feo, C.; Liboni, A.; et al. Elevated expression of A3 adenosine receptors in human colorectal cancer is reflected in peripheral blood cells. Clin. Cancer Res. 2004, 10, 5895–5901. [Google Scholar] [CrossRef] [Green Version]
  108. Pluta, R.; Ułamek-Kozioł, M.; Januszewski, S.; Czuczwar, S.J. Platelets, Lymphocytes and erythrocytes from Alzheimer’s disease patients: The quest for blood cell-based biomarkers. Folia Neuropathol. 2018, 56, 14–20. [Google Scholar] [CrossRef] [PubMed]
  109. Arosio, B.; Mastronardi, L.; As, C.; Nicolini, P.; Casè, A.; Ziglioli, E.; Bergamaschini, L. Adenosine A(2A) Receptor and IL-10 in Peripheral Blood Mononuclear Cells of Patients with Mild Cognitive Impairment. Int. J. Alzheimers Dis. 2011, 2011, 484021. [Google Scholar]
  110. Arosio, B.; Viazzoli, C.; Mastronardi, L.; Bilotta, C.; Vergani, C.; Bergamaschini, L. Adenosine A2A receptor expression in peripheral blood mononuclear cells of patients with mild cognitive impairment. J. Alzheimers.Dis. 2010, 20, 991–996. [Google Scholar] [CrossRef] [PubMed]
  111. Gussago, C.; Arosio, B.; Casati, M.; Ferri, E.; Gualandris, F.; Tedone, E.; Nicolini, P.; Rossi, P.D.; Abbate, C.; Mari, D. Different adenosine A2A receptor expression in peripheral cells from elderly patients with vascular dementia and Alzheimer’s disease. J. Alzheimer’s Dis. 2014, 40, 45–49. [Google Scholar] [CrossRef]
  112. Pasquini, S.; Vincenzi, F.; Casetta, I.; Laudisi, M.; Merighi, S.; Gessi, S.; Borea, P.A.; Varani, K. Adenosinergic System Involvement in Ischemic Stroke Patients’ Lymphocytes. Cells 2020, 9, 1072. [Google Scholar] [CrossRef]
  113. Ravani, A.; Vincenzi, F.; Bortoluzzi, A.; Padovan, M.; Pasquini, S.; Gessi, S.; Merighi, S.; Borea, P.A.; Govoni, M.; Varani, K. Role and Function of A(2A) and A3 Adenosine Receptors in Patients with Ankylosing Spondylitis, Psoriatic Arthritis and Rheumatoid Arthritis. Int. J. Mol. Sci. 2017, 18, 697. [Google Scholar] [CrossRef] [PubMed]
  114. Bortoluzzi, A.; Vincenzi, F.; Govoni, M.; Padovan, M.; Ravani, A.; Borea, P.A.; Varani, K. A2A adenosine receptor upregulation correlates with disease activity in patients with systemic lupus erythematosus. Arthritis Res. Ther. 2016, 18, 192. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  115. Vincenzi, F.; Corciulo, C.; Targa, M.; Casetta, I.; Gentile, M.; Granieri, E.; Borea, P.A.; Popoli, P.; Varani, K. A2A adenosine receptors are up-regulated in lymphocytes from amyotrophic lateral sclerosis patients. Amyotroph. Lateral Scler. Front. Degener. 2013, 14, 406–413. [Google Scholar] [CrossRef]
  116. Vincenzi, F.; Corciulo, C.; Targa, M.; Merighi, S.; Gessi, S.; Casetta, I.; Gentile, M.; Granieri, E.; Borea, P.A.; Varani, K. Multiple sclerosis lymphocytes upregulate A2A adenosine receptors that are antiinflammatory when stimulated. Eur. J. Immunol. 2013, 43, 2206–2216. [Google Scholar] [CrossRef]
  117. Varani, K.; Abbracchio, M.P.; Cannella, M.; Cislaghi, G.; Giallonardo, P.; Mariotti, C.; Cattabriga, E.; Cattabeni, F.; Borea, P.A.; Squitieri, F.; et al. Aberrant A2A receptor function in peripheral blood cells in Huntington’s disease. FASEB J. 2003, 17, 2148–2150. [Google Scholar] [CrossRef]
  118. Godoy-Marín, H.; Duroux, R.; Jacobson, K.A.; Soler, C.; Colino-Lage, H.; Jiménez-Sábado, V.; Montiel, J.; Hove-Madsen, L.; Ciruela, F. Adenosine A2A Receptors Are Upregulated in Peripheral Blood Mononuclear Cells from Atrial Fibrillation Patients. Int. J. Mol. Sci. 2021, 22, 3467. [Google Scholar] [CrossRef]
  119. Yang, K.; Yang, Z.; Chen, X.; Li, W. The Significance of Sialylation on the Pathogenesis of Alzheimer’s Disease. Brain Res. Bull. 2021, 173, 116–123. [Google Scholar] [CrossRef] [PubMed]
  120. Merighi, S.; Poloni, T.E.; Pelloni, L.; Varani, K.; Vincenzi, F.; Borea, P.A.; Gessi, S. An open question: Is the A2A adenosine receptor a novel target for Alzheimer’s disease treatment? Front. Pharmacol. 2021, 12, 652455. [Google Scholar] [CrossRef] [PubMed]
  121. Merighi, S.; Poloni, T.E.; Terrazzan, A.; Moretti, E.; Gessi, S.; Ferrari, D. Alzheimer and Purinergic Signaling: Just a matter of inflammation? Cells 2021, 10, 1267. [Google Scholar] [CrossRef]
  122. Atif, M.; Alsrhani, A.; Naz, F.; Imran, M.; Imran, M.; Ullah, M.I.; Alameen, A.A.M.; Gondal, T.A.; Raza, Q. Targeting Adenosine Receptors in Neurological Diseases. Cell Reprogram 2021, 23, 57–72. [Google Scholar] [CrossRef] [PubMed]
  123. Chen, J.F.; Cunha, R. The belated US FDA approval of the adenosine A2A receptor antagonist istradefylline for treatment of Parkinson’s disease. Purinergic Signal. 2020, 16, 167–174. [Google Scholar] [CrossRef] [PubMed]
  124. Kim, Y.; Je, Y.; Giovannucci, E. Coffee consumption and all-cause and cause-specific mortality: A meta-analysis by potential modifiers. Eur. J. Epidemiol. 2019, 34, 731–752. [Google Scholar] [CrossRef] [PubMed]
  125. Charvin, D.; Medori, R.; Hauser, R.A.; Rascol, O. Therapeutic strategies for Parkinson disease: Beyond dopaminergic drugs. Nat. Rev. Drug Discov. 2018, 17, 804–822. [Google Scholar] [CrossRef]
Figure 1. Schematic diagram illustrating the role of A2A adenosine receptors in AD.
Figure 1. Schematic diagram illustrating the role of A2A adenosine receptors in AD.
Cells 10 02344 g001
Table 1. Individual score obtained from the combination of ATN [15,16].
Table 1. Individual score obtained from the combination of ATN [15,16].
ATN System
A-T-N-No biomarkers of degenerative brain pathology
A + T-N-Amyloid deposition (Alzheimer’s continuum)
A + T-N+Amyloid deposition and non-tau degeneration (Alzheimer’s continuum; suspected non-AD pathology)
A + T + N-Early Alzheimer’s Disease (Alzheimer’s continuum)
A + T + N+Alzheimer’s Disease (Alzheimer’s continuum)
A-T + N-
A-T-N+
A-T + N+
Non-Alzheimer neurodegenerative Diseases
(e.g., primary TAUpahies, Fronto-Temporal Dementia due to TDP-43, LATE, other rare forms)
Table 2. Biomarkers of Alzheimer’s Disease.
Table 2. Biomarkers of Alzheimer’s Disease.
Biomarkers of Alzheimer’s Disease
ATN System [15,16]
Biomarkers of β-amyloid plaques (A)Cortical amyloid PET
Low CSF β-amyloid 42
Biomarkers of tau (T)Cortical tau PET
Elevated CSF phospho-tau
Biomarkers of neurodegeneration and neuronal injury (N)[(18)F]-fluorodeoxyglucose PET hypometabolism
Atrophy on MRI
Elevated CSF total-tau
Fluid biomarkers
Increased levels of plasma Neurofilament light (NfL)
[22,27]
Increased levels of plasma tau phosphorylated at threonine 181 (P-tau181)
[23]
Increased levels of plasma tau phosphorylated at threonine-217 (P-tau217)
[28]
Increased levels of plasma Aβ-amyloid oligomers [24]
Lower levels of plasma Aβ42/Aβ40 ratio (TP42/40)
[25,26]
Increased levels of CSF Neurogranin (Ng) [29]
Increased levels of CSF YKL-40 [34,35]
Table 3. Upregulation of A2A adenosine receptors in aged or animal models of AD and in AD patients.
Table 3. Upregulation of A2A adenosine receptors in aged or animal models of AD and in AD patients.
Aged/Animal Models of AD/AD PatientsTissue/CellReferences
aged ratscortex and hippocampus[72]
aged vs. young ratscortical membranes[64]
aged vs. young ratshippocampal neurons[57,73]
AD patientsmicroglia[74]
APPsw tg micehyppocampus[65]
adult and aged ratshyppocampus[75]
AD patientsfrontal cortex[70]
Rats with different agesnerve terminals purified from the hippocampus[66]
AD rat modelhippocampus[67]
AD patients/aged miceastrocytes[68,76]
APP/PS1 miceCA3 synaptic membranes[62]
Early AD mice modelhippocampal synaptosomes [69]
APPSw/Ind AD transgenic mice modelmicroglia[77]
Aged subjects and AD patientshippocampal neurons[60]
AD patientscortex, hippocampus, platelets[71]
Abbreviations: swedish mutation (Sw) transgenic (Tg), Indiana (Ind) mutations.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Gessi, S.; Poloni, T.E.; Negro, G.; Varani, K.; Pasquini, S.; Vincenzi, F.; Borea, P.A.; Merighi, S. A2A Adenosine Receptor as a Potential Biomarker and a Possible Therapeutic Target in Alzheimer’s Disease. Cells 2021, 10, 2344. https://doi.org/10.3390/cells10092344

AMA Style

Gessi S, Poloni TE, Negro G, Varani K, Pasquini S, Vincenzi F, Borea PA, Merighi S. A2A Adenosine Receptor as a Potential Biomarker and a Possible Therapeutic Target in Alzheimer’s Disease. Cells. 2021; 10(9):2344. https://doi.org/10.3390/cells10092344

Chicago/Turabian Style

Gessi, Stefania, Tino Emanuele Poloni, Giulia Negro, Katia Varani, Silvia Pasquini, Fabrizio Vincenzi, Pier Andrea Borea, and Stefania Merighi. 2021. "A2A Adenosine Receptor as a Potential Biomarker and a Possible Therapeutic Target in Alzheimer’s Disease" Cells 10, no. 9: 2344. https://doi.org/10.3390/cells10092344

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop