Skip to main content
Springer logoLink to Springer
. 2019 May 4;44(10):2413–2422. doi: 10.1007/s11064-019-02798-1

Purinergic Signalling in Parkinson’s Disease: A Multi-target System to Combat Neurodegeneration

Adrián Tóth 1,2,3, Zsófia Antal 2, Dániel Bereczki 1, Beáta Sperlágh 2,
PMCID: PMC6776560  PMID: 31054067

Abstract

Parkinson’s disease (PD) is the second most common neurodegenerative disorder, characterized by progressive loss of dopaminergic neurons that results in characteristic motor and non-motor symptoms. l-3,4 dihydroxyphenylalanine (l-DOPA) is the gold standard therapy for the treatment of PD. However, long-term use of l-DOPA leads to side effects such as dyskinesias and motor fluctuation. Since purines have neurotransmitter and co-transmitter properties, the function of the purinergic system has been thoroughly studied in the nervous system. Adenosine and adenosine 5′-triphosphate (ATP) are modulators of dopaminergic neurotransmission, neuroinflammatory processes, oxidative stress, excitotoxicity and cell death via purinergic receptor subtypes. Aberrant purinergic receptor signalling can be either the cause or the result of numerous pathological conditions, including neurodegenerative disorders. Many data confirm the involvement of purinergic signalling pathways in PD. Modulation of purinergic receptor subtypes, the activity of ectonucleotidases and ATP transporters could be beneficial in the treatment of PD. We give a brief summary of the background of purinergic signalling focusing on its roles in PD. Possible targets for pharmacological treatment are highlighted.

Keywords: Adenosine, Adenosine receptors, ATP, Parkinson’s disease, Purinergic receptors

Introduction

Parkinsons’s Disease: Pathophysiological Background

Parkinson’s disease (PD) is the second most common neurodegenerative disorder, characterized by progressive loss of dopaminergic neurons in the substantia nigra pars compacta that results in dopamine (DA) deficiency in the striatum. The ongoing degeneration of this peculiar pathway causes the characteristic motor symptoms such as resting tremor, rigidity, bradykinesia and postural instability [1, 2]. Besides dopaminergic neural degeneration, the presence of Lewy bodies (protein aggregates) due to misfolding of α-synuclein occurs in various regions of the affected brain [3]. In spite of many studies on the pathogenesis of PD, the precise mechanism underlying these events has not been unraveled yet. However, a genetic predisposition associated with disturbed proteostasis due to impaired ubiquitin–proteasome system, mitochondrial dysfunction, oxidative stress and neuroinflammation seems to play cardinal roles for the α-synuclein aggregation and the progression of pathology in PD [47]. Among these factors, the pathological, self-amplifying interaction between mitochondrial dysfunction and oxidative stress has been early recognized, which might be a key factor responsible for the selective vulnerability of dopaminergic neurons in PD, and one potential reason behind the clinical failures of neuroprotective therapies so far [8]. Dysfunction of the mitochondrial complex I results in an enhanced production of reactive oxygen species (ROS), which, in turn will inhibit complex I and other vital metabolic enzymes such as alpha-ketoglutarate dehydrogenase, whilst the latter also serves as a source of ROS generation in mitochondria [9, 10]. Simultaneous or preceding mitochondrial dysfunction exacerbates the effect of oxidative stress on pathological monoamine release from nerve terminals [11, 12]. This process leads to the formation of toxic, oxidative DA metabolites, such as dopamine quinone, which might further amplify the ongoing degeneration process [13]. Therefore, disease-modifying potential could be primarily expected from those novel multi-target therapies, which simultaneously target the above mentioned pivotal pathological pathways and prevent their pathological interaction [14, 15].

The Current Treatment of PD

As for the symptomatic treatment of PD, the clinical break-through came with the first clinical trials of DA replacement therapy using the high dosage of the DA precursor l-3,4 dihydroxyphenylalanine (l-DOPA) [1619]. l-DOPA is able to cross the blood–brain barrier and converts into DA that engages specific DA receptor subtypes (D1 to D5) [20]. However, long-term use of l-DOPA leads to a dysbalance of striatal circuits of the motor system and leads to side effects such as l-DOPA induced dyskinesias and motor fluctuation in 50% of patients after 5 years of continuous treatment [21, 22]. The therapeutic management of these complications is difficult and there is a need for developing effective and new pharmacological therapies against motor fluctuation and dyskinesias [23].

Purinergic Signalling: Concept and Purinergic Receptors

The concept of purinergic signalling, being adenosine 5′-triphosphate (ATP) as an extracellular signalling molecule with neurotransmitter properties was proposed in the early 1970s [24, 25]. A couple of years later, purines were also described as co-transmitters and neuromodulators in the peripheral and central nervous system (CNS), as they are able to modulate other signalling pathways and neurotransmitter systems [2628]. ATP is co-released with acetylcholine, catecholamines, γ-amino butyric acid (GABA), glutamate and DA in the CNS [2934]. Extracellular ATP is released from cells under physiological conditions. The levels of extracellular ATP are controlled by ectonucleotidases that catalyze its degradation [35, 36].

There are two families of purinergic receptors, which are distinguished by their main agonists [37]. P1 receptors are G protein-coupled metabotropic receptors activated by adenosine and can be subdivided into four subtypes (A1, A2A, A2B, A3). P2 receptors are subdivided into two classes: P2X(1-7) ionotropic receptors, activated by ATP and G protein-coupled metabotropic P2Y(1-2,4,6,11-14) receptors, activated by ATP, adenosine diphosphate (ADP), uridine di- and triphosphate (UDP and UTP), or UDP-glucose depending on the receptor subtype [3840]. ATP is able to bind to the extracellular ligand-binding site of P2X receptors and leading to conformational change that opens a permeable channel to Na+, K+ and Ca2+. The activation of these ionotropic receptors is important for Ca2+-induced intracellular signalling pathways [4143]. Depending on the activated adenosine and P2 receptor subtype, the induced signalling pathway may vary. These activated receptors are able to make alterations in Ca2+ levels, which modulate the activity of several secondary messengers involved in physiological processes [4446]. The final effects of purinergic receptor-mediated signalling depend on the cell type and other physiological (neurogenesis, proliferation, cell death, stem cell differentiation) or pathological cellular conditions (inflammatory, neurological, psychiatric, oncological, cognitive, neuromuscular and neuromotor diseases) [4766]. Purinergic receptor activation may have para- or autocrine nature, which is characteristic for astrocytes in the regulation of neuronal activity [67]. Not only purinergic receptors but membrane nucleotide/nucleoside transporters, channels and ectonucleotidases also play important role in purinergic signalling [36, 6870].

Adenosine is the predominant, presynaptic modulator of neurotransmitter release in the CNS, although ATP has presynaptic modulator effect as well [7173]. Adenosine is produced by enzymatic breakdown of released ATP, but some CNS cells are able to release adenosine directly [74]. A1 and A2A receptors have higher affinity (activated by physiological extracellular levels of adenosine) and A2B and A3 receptors have lower affinity (activated by higher extracellular levels of adenosine) for the ribonucleoside [7577]. The adenosine A1 and A2A receptors are highly expressed in the brain and CNS, where they have profound influence on neuronal activity. Adenosine A1 receptor is the dominant adenosine receptor subtype in the CNS. Adenosine A1 receptors can be found in various cortical and subcortical regions of the brain, while A2A receptors are mainly expressed in the striatum [7881] (Table 1). In contrast, adenosine A2B and A3 receptors are mainly found in peripheral tissues, even though low levels of these receptors are also expressed in some regions of the brain [8284].

Table 1.

Localization of adenosine receptor subtypes in CNS [80, 81]

CNS
A1 High levels in striatum, thalamus and moderate levels in cortex, pons
A2A High levels in striatum, thalamus, hippocampus
A2B Low levels in microglia cells, astrocytes
A3 Low levels in cortex, hippocampus, striatum, cerebellum

There is a heterogeneous distribution of P2 purinergic receptors in the CNS as well. For instance P2X1 receptors are predominantly expressed in the cerebellum, while P2X3 receptors are expressed in the brainstem [85, 86], and they can be found in the basal ganglia with variable expression level [87] (Table 2). Various P1 and P2 receptor subtypes are also expressed by microglia, astrocytes and oligodendrocytes [8893]. Extracellular nucleotides act as messengers between neuronal and non-neuronal cells, thereby integrating functional activity between neurons, glial and vascular cells in the CNS [9498]. Adenosine and ATP—as key players in neuron–glia interaction and microglial activation—are modulators of neuroinflammatory processes, oxidative stress, excitotoxicity and cell death [99102]. Aberrant purinergic receptor signalling can be the cause or result of numerous pathological conditions, including neurodegenerative disorders [103]. Here, we explore the importance of purinergic signalling in PD to suggest potential targets for novel therapies.

Table 2.

Expression of P2 receptor subtypes in the basal ganglia (striatum and substantia nigra) [87]

Striatum Substantia nigra
P2X1 ↑↑↑ ↑↑↑
P2X2 ↑↑↑ ↑↑↑
P2X3 ↑↑ ↑↑
P2X4 ↑↑↑ ↑↑↑
P2X5 ↑↑↑
P2X6 ↑↑
P2X7 ↑↑ ↑↑
P2Y1 ↑↑
P2Y2 ↑↑↑ ↑↑↑
P2Y4 ↑↑↑ ↑↑↑
P2Y6 ↑↑↑
P2Y11
P2Y12 ↑↑↑ ↑↑↑
P2Y13
P2Y14 ↑↑↑

Expression level of P2 receptor subtypes: – = no expression, ↑ = low expression, ↑↑ = medium expression, ↑↑↑ = high expression

Purinergic Signalling Involvement in PD

Purinergic Gene Polymorphisms in PD

Two ADORA2A (A2A receptor) polymorphisms (rs71651683, a 5′ variant or rs5996696, a promoter region variant) were inversely associated with genetic PD risk, moreover, there was evidence of interaction with coffee consumption [104]. CYP1A2a is an enzyme, which is responsible for caffeine metabolism, two polymorphisms (rs762551 or rs5996696) of the enzyme in homozygous coffee drinkers reduced PD risk [104]. Humans with R1628P variant (LRRK2 risk variant) who did not take caffeine had a 15 times increased risk of PD [105]. GRIN2A encodes an N-methyl-d-aspartate-2A (NMDA) glutamate receptor subunit involved in central excitatory neurotransmission, which is associated with A2A receptor activation. Carriers of GRIN2A rs4998386-T allele had a lower risk of PD, than carriers of rs4998386-CC genotype among heavy coffee drinkers [106]. There is evidence that creatine is able to hasten PD progression in GRIN2A coffee drinkers, which demonstrates an example of a genetic factor interacting with environmental factors exemplifying the complexity of environment–gene interactions in the progression of PD [107]. In addition, P2X7 receptor 1513A>C polymorphism is a risk factor for sporadic PD, late-onset PD and male PD in Han Chinese population [108].

Adenosine Receptor-Mediated Signalling in PD

A2A receptors are enriched in dopaminergic brain areas (the highest expression of these receptors are in the striatum), thus pointing to a significant role of purines in motor control [109]. A2A and DA D2 receptors are mainly expressed in the neurons of the indirect pathway of striatal circuits projecting to the globus pallidus, in contrast to A1 and DA D1 receptors, which are mainly found on the neurons of the direct pathway of motor control projecting to the internal globus pallidus and substantia nigra pars reticulata. The main adenosine signalling mechanism is via the cyclic adenosine monophosphate (cAMP)-dependent pathway. Activated A2A receptors stimulate the enzymatic function of adenyl cyclase that increases cAMP levels and depresses the signalling mediated by D2 receptors. Activation of protein Gi-coupled DA D2 receptors leads to reduction in the cAMP level. There is a reciprocal situation in the direct pathway of motor control with protein Gs-coupled D1 and protein Gi/o-coupled A1 receptors. Generally, adenosine acts as a negative modulator of D1- and D2-mediated actions in the direct and indirect pathways [110112].

The antagonistic functional interaction between adenosine A2A and DA D2 receptors may depend on the formation of receptor heterodimers (A2A-D2 heteroreceptor complexes) in the striatum thereby balancing the inhibitory and excitatory impulses in the striatal circuits [112]. Not only dopaminergic mechanisms, but non-dopaminergic modes of action of A2A receptors may involve interactions with various non-dopaminergic receptors, possibly by forming heterodimeric and/or multimeric receptor complexes [23]. Thus, adenosine A2A receptors may adjust the actions of striatal adenosine A1 receptors (A1-A2A heteroreceptor complexes), metabotropic glutamate receptors (mGlu) 5 (A2A-mGlu5 heteroreceptor complexes), cannabinoid receptor type 1 (CB1) receptors (A2A-CB1 heteroreceptor complexes) and serotonin 1A (5-HT1A) receptors [113115]. Moreover, studies also suggested the presence of multimeric A2A-D2-mGlu5 and A2A-CB1-D2 receptor complexes in the striatum [116, 117]. These functional interactions between receptors may modulate the activity of striatal efferent neurons and influence motor behavior [23]. In general, adenosine tone appears as a key for the fine tune control of DA dependent actions in the basal ganglia and affects non-dopaminergic mechanisms also [20].

Adenosine receptor antagonists (especially non-selective A2A receptor antagonists, such as methylxanthines, caffeine, or selective A2A antagonists) have been shown to enhance therapeutic effect of l-DOPA in a wide range of animal models of PD [118121]. A2A homoreceptor complexes are in balance with DA D2 homoreceptor complexes in intact striatum [122126]. Dysbalance of striatal circuits leads to motor inhibition and disruption of this balance in PD leads to increased signalling via A2A receptors and decreased signalling via DA D2 receptors. These changes explain the beneficial effect of A2A receptor antagonists on increasing motor functions without worsening l-DOPA-induced dyskinesias [20, 127].

A2A receptor antagonists have been used in clinical trials in patients with PD (Table 3). Istradefylline is a xanthine-based compound with increased selectivity for A2A receptors against A1 receptors, which is used concomitantly with l-DOPA [128]. The drug was not approved in the USA because there was no significant reduction in off time compared to l-DOPA treatment [129]. In contrast, istradefylline was approved in Japan in 2013 with the trade name Nouriast® to enhance the antiparkinsonian effect of l-DOPA with less long-term side effects [130, 131]. Preladenant is a second-generation A2A receptor antagonist, which failed in phase III clinical trials in the treatment of PD because the compound was not superior to placebo in reducing off state [132, 133]. Vipadenant is a triazolopyromidine-based drug, which has increased selectivity for A2A receptors versus A1 and A3 receptors [134]. Its development as an antiparkinsonian medication was stopped; however, A2A receptor antagonists have considerable potential in novel immune-oncology and cardiology therapies [113, 135137]. Another adenosine A2A receptor antagonist, tozadenant was safe, well tolerated and effective in reducing off time in PD patients in phase II trial but phase III clinical trial was discontinued because of serious adverse events (agranulocytosis) [23, 133, 138]. There have been many drug trials for selective A2A receptor antagonists. Most of them were shown to be safe, well tolerated and beneficial; however, the majority did not reach the regulatory threshold for efficacy to be approved as PD drugs [139, 140]. Development of bivalent drugs (able to bind to two receptors simultaneously) to target A2A-D2 heteroreceptor complexes acting on A2A and DA D2 receptors may be a good therapeutic approach in the future. Heterobivalent drugs offers the opportunity to target the orthosteric sites of the receptors in the heterodimer with a higher affinity and a higher specificity versus corresponding homomers and reduce the dose required for therapy and, accordingly, the side effects [20].

Table 3.

Pre-clinical and clinical studies with purinergic receptor antagonists in PD

Compounds Mechanism of effect Models Published Results
KW-6002 (istradefylline) A2A receptor antagonism PD patients 2003 Improved PD motor scores when added to low-dose l-DOPA
KW-6002 (istradefylline) A2A receptor antagonism LPS treated rats 2013 Enhanced therapeutic effect of l-DOPA
Caffeine A2A receptor antagonism LPS treated rats 2013 Reduced motor impairment
Preladenant A2A receptor antagonism MPTP treated mice 2014 Enhanced therapeutic effect of low doses of l-DOPA
8-Ethoxy-9-ethyladenine A2A receptor antagonism 6-OHDA lesioned rats 2015 Enhanced effect of low doses of l-DOPA without increased dyskinesia
SCH 58261 A2A receptor antagonism A2A receptor knockout mice, SH-SY5Y cells 2015 Decreased α-synuclein aggregation, prevented neuronal death
ZM 241385 A2A receptor antagonism A2A receptor knockout mice, SH-SY5Y cells 2015 Decreased α-synuclein aggregation, prevented neuronal death
Preladenant A2A receptor antagonism PD patients 2017 Failed (was not superior to placebo) in phase III clinical trial
Vipadenant A2A receptor antagonism PD patients 2009 Failed (was not superior to placebo)
Tozadenant A2A receptor antagonism PD patients 2017 Failed in phase III clinical trial (induced agranulocytosis)
NF449 P2X1 receptor antagonism H4 cells 2015 Prevented α-synuclein aggregation
A-438079 P2X7 receptor antagonism 6-OHDA lesioned rats 2010 Prevented depletion of DA in striatum
BBG P2X7 receptor antagonism 6-OHDA lesioned rats 2014 Reverted dopaminergic neurons loss in substantia nigra
BBG P2X7 receptor antagonism BV2 microglia cells 2015 Decreased ROS production induced by α-synuclein
PPADS P2X7 receptor antagonism SH-SY5Y cells 2017 Prevented abnormal calcium influx induced by α-synuclein
AZ 11645373 P2X7 receptor antagonism SH-SY5Y cells 2017 Prevented abnormal calcium influx induced by α-synuclein
AP4A P2Y2/P2Y4 antagonism 6-OHDA lesioned rats 2003 Reduced dopaminergic neurons loss
MRS2578 P2Y6 receptor antagonism SH-SY5Y cells 2017 Delayed neuronal loss

The list is not comprehensive and is restricted to studies mentioned in the article. For further references, see [111, 113]

Adenosine A2A receptor antagonists may also involve direct or indirect actions at microglia and inflammatory processes. Pre-treatment of slices from 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-injected mice with preladenant facilitates the ability of activated microglia to respond to tissue damage [141]. The nonselective A1/A2A adenosine receptor antagonist caffeine and the selective A2A receptor antagonist (KW-6002) had anti-inflammatory potential in a rat model of lipopolysaccharide (LPS)-induced neuroinflammation [142].

The Role of A2A Receptors in Synucleopathy

Increased striatal A2A receptor expression was observed as an early pathological event in PD and increased A2A receptor expression was detected after hippocampal injection of α-synuclein in mice [143, 144]. A2A receptor-knock out mice showed resistance against α-synuclein induced insults [145]. A2A receptor antagonism restrained hyperactivation of NMDA-glutamate receptors and decreased the aggregation of α-synucleins [146]. Based upon these results, A2A receptors seem to have role in the pathological process of synucleinopathy [111].

P2 Receptor-Mediated Signalling in PD

P2 ionotropic and metabotropic receptors are widely expressed in basal ganglia and in various cell types, such as neurons and astrocytes [87, 147, 148]. 6-Hydroxidopamine (6-OHDA) induced lesions of nigral dopaminergic neurons generate a significant decrease in the expression of P2X and P2Y receptor proteins from striatal spiny neurons and GABAergic interneurons, thus confirming the involvement of P2 receptors and extracellular ATP in the striatal circuits [87]. P2Y1 and P2X1-4, 6 receptor protein subtypes are expressed in dopaminergic neurons with co-expression of P2X1 with DA D1 receptors, therefore stimulation of P2 receptors by ATP induces an increased release of DA in the striatum [149152]. In a neuronal cell model, extracellular ATP induced a significant increase in intracellular α-synuclein levels, which was the result of lysosome dysfunction caused by P2X1 receptor activation [153].

Many data have implicated the role of P2X7 receptor in PD. P2X7 receptor antagonism with A-438059 or Brilliant Blue G (BBG) prevented DA deficit in the striatum and 6-OHDA-induced hemiparkinsonian behavior [154, 155]. However, P2X7 receptor deficiency or inhibition did not promote the survival of dopaminergic neurons in rotenone and MPTP induced animal models of PD [156]. It is presumed that there is a massive release of ATP during cell death in the lesioned striatum and substantia nigra, which activates cell death pathways via purinergic receptors and is able to activate further purinergic subtypes [20]. Permanent purinergic receptor activation and ATP release seem to play a key role in the neuronal death, which exacerbates α-synuclein aggregation in PD [87]. The accumulation of α-synuclein might overwhelm the capacity of intracellular protein-degradation mechanisms and induce neuroinflammation, which creates a positive feedback loop promoting the degeneration of dopaminergic cells [7]. α-Synuclein-induced intracellular free calcium mobilization in neuronal cells depends on the activation of purinergic P2X7 receptors. In the same study, activation of P2X7 receptors lead to ATP release with the recruitment of the pore forming protein pannexin1, whilst α-synuclein decreased the activity of extracellular ecto-ATPase which is responsible for ATP degradation [157]. Stimulation of the microglial P2X7 receptor by extracellular α-synuclein increased oxidative stress, which was prevented with the use of P2X7 receptor antagonist [158].

DA neurotransmission has been linked to calcium signalling. There is data that P2Y1 receptor is involved in the regulation of calcium signalling [159]. Neurodegeneration induced by 6-OHDA in nigrostriatal dopaminergic neurons was reduced by pretreatment with diadenosine tetraphosphate (AP4A, an endogenous diadenosine polyphosphate) possibly through an anti-apoptotic mechanism and the activation of P2Y1 and P2Y4 receptors [160]. Recently, expression levels of P2Y6 receptor in PD patients younger than 80 years were higher than healthy controls and multiple system atrophy (MSA) patients and P2Y6 receptor could thereby be a potential clinical biomarker of PD. P2Y6 receptor was also upregulated in LPS-treated microglial cells and involved in proinflammatory cytokine release through UDP secretion [161]. Another study showed that expression of P2Y6 receptor on neuronal SH-SY5Y cell is associated with the progression of oxidative stress and cell death induced by 1-methyl-4-phenylpyridinium (MPP+) [162]. In vivo, LPS induced microglial activation and delayed neuronal loss was prevented by selective inhibition of P2Y6 receptor with MRS2578 [163]. Based on these studies P2Y6 receptor subtype seems to be involved in the process of neuroinflammation in PD and blocking UDP/P2Y6 receptor signalling could reverse these pathological processes [161].

Conclusion

In general, many data confirm the involvement of purinergic signalling pathways in PD. Modulation of purinergic receptor subtypes, the activity of ectonucleotidases and ATP transporters could be beneficial in the treatment of PD. Antagonism of A2A, P2X1, P2X7 and P2Y6 receptor subtypes is a promising weapon against PD via various ways: reducing l-DOPA induced dyskinesia, influencing neuroinflammation, preventing α-synuclein aggregation, reducing microglia activation. Development of new bivalent compounds to target A2A-D2 heteroreceptor complexes, which are orally bioavailable and can cross the blood–brain barrier could be a potential therapeutic tool. In addition, multi-target compounds targeting self-amplifying circuits controlled by purinergic and non-purinergic receptors could be a viable strategy to obtain the desired disease-modifying effect [164]. Additional studies and better quality PD animal models are required for the deeper understanding of underlying unknown pathological processes in PD and the role of purinergic signalling in it.

Acknowledgements

Open access funding provided by MTA Institute of Experimental Medicine (MTA KOKI). This study was supported by Research Grants from Hungarian Research and Development Fund (Grant K116654 to BS), Hungarian Brain Research Program (2017-1.2.1.-NKP-2017-00002 to BS) and the European Union’s Horizon 2020 Research and Innovation Programme under the Marie Sklodowska-Curie Grant Agreement No. 766124.

Abbreviations

ADORA2A

Adenosine A2A receptor

ADP

Adenosine 5′-diphosphate

AP4A

Diadenosine tetraphosphate

ATP

Adenosine 5′-triphosphate

cAMP

Cyclic adenosine monophosphate

CB1

Cannabinoid receptor type 1

DA

Dopamine

GABA

γ-Amino butyric acid

GRIN2A

Glutamate ionotropic receptor NMDA type subunit 2A

5-HT1A

5-Hydroxytryptamine/serotonin receptor 1A

l-DOPA

l-3,4 dihydroxyphenylalanine

LPS

Lipopolysaccharide

LRRK2

Leucine-rich repeat kinase 2

6-OHDA

6-hydroxydopamine

mGlu

Metabotropic glutamate receptor

MPP+

1-Methyl-4-phenylpyridinium

MPTP

1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine

MSA

Multiple system atrophy

NMDA

N-methyl-d-aspartate

PD

Parkinson’s disease

ROS

Reactive oxygen species

UDP

Uridine 5′-diphosphate

UTP

Uridine 5′-triphosphate

Footnotes

The authors are pleased to be part of the SI dedicated to Professor Vera Adam-Vizi and grateful for the great inspiration and collaboration.

Special Issue of Neurochemical Research: In honour of Professor Vera Adam-Vizi.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

References

  • 1.Dehay B, Bourdenx M, Gorry P, et al. Targeting α-synuclein for treatment of Parkinson’s disease: mechanistic and therapeutic considerations. Lancet Neurol. 2015;14:855–866. doi: 10.1016/S1474-4422(15)00006-X. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Olanow CW, Kieburtz K, Odin P, et al. Continuous intrajejunal infusion of levodopa–carbidopa intestinal gel for patients with advanced Parkinson’s disease: a randomised, controlled, double-blind, double-dummy study. Lancet Neurol. 2014;13:141–149. doi: 10.1016/S1474-4422(13)70293-X. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Braak H, Del Tredici K, Rüb U, et al. Staging of brain pathology related to sporadic Parkinson’s disease. Neurobiol Aging. 2013;24:197–211. doi: 10.1016/s0197-4580(02)00065-9. [DOI] [PubMed] [Google Scholar]
  • 4.Ghavami S, Shojaei S, Yeganeh B, et al. Autophagy and apoptosis dysfunction in neurodegenerative disorders. Prog Neurobiol. 2014;112:24–49. doi: 10.1016/j.pneurobio.2013.10.004. [DOI] [PubMed] [Google Scholar]
  • 5.Osellame LD, Duchen MR. Defective quality control mechanisms and accumulation of damaged mitochondria link Gaucher and Parkinson diseases. Autophagy. 2013;9:1633–1635. doi: 10.4161/auto.25878. [DOI] [PubMed] [Google Scholar]
  • 6.Tansey MG, Goldberg MS. Neuroinflammation in Parkinson’s disease: its role in neuronal death and implications for therapeutic intervention. Neurobiol Dis. 2010;37:510–518. doi: 10.1016/j.nbd.2009.11.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Zhang G, Xia Y, Wan F, et al. New perspectives on roles of alpha-synuclein in Parkinson’s disease. Front Aging Neurosci. 2018;10:370. doi: 10.3389/fnagi.2018.00370. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Tretter L, Sipos I, Adam-Vizi V. Initiation of neuronal damage by complex I deficiency and oxidative stress in Parkinson’s disease. Neurochem Res. 2004;29:569–577. doi: 10.1023/b:nere.0000014827.94562.4b. [DOI] [PubMed] [Google Scholar]
  • 9.Tretter L, Adam-Vizi V. Generation of reactive oxygen species in the reaction catalyzed by alpha-ketoglutarate dehydrogenase. J Neurosci. 2004;24:7771–7778. doi: 10.1523/JNEUROSCI.1842-04.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Adam-Vizi V, Tretter L. The role of mitochondrial dehydrogenases in the generation of oxidative stress. Neurochem Int. 2013;62:757–763. doi: 10.1016/j.neuint.2013.01.012. [DOI] [PubMed] [Google Scholar]
  • 11.Milusheva E, Sperlagh B, Shikova L, et al. Non-synaptic release of [3H]noradrenaline in response to oxidative stress combined with mitochondrial dysfunction in rat hippocampal slices. Neuroscience. 2003;120:771–781. doi: 10.1016/s0306-4522(03)00340-3. [DOI] [PubMed] [Google Scholar]
  • 12.Milusheva E, Baranyi M, Kittel Á, et al. Increased sensitivity of striatal dopamine release to H2O2 upon chronic rotenone treatment. Free Radic Biol Med. 2005;39:133–142. doi: 10.1016/j.freeradbiomed.2005.02.034. [DOI] [PubMed] [Google Scholar]
  • 13.Baranyi M, Milusheva E, Vizi ES, et al. Chromatographic analysis of dopamine metabolism in a Parkinsonian model. J Chromatogr. 2006;1120:13–20. doi: 10.1016/j.chroma.2006.03.018. [DOI] [PubMed] [Google Scholar]
  • 14.Milusheva E, Baranyi M, Kormos E, et al. The effect of antiparkinsonian drugs on oxidative stress induced pathological [3H]dopamine efflux after in vitro rotenone exposure in rat striatal slices. Neuropharmacology. 2010;58:816–825. doi: 10.1016/j.neuropharm.2009.11.017. [DOI] [PubMed] [Google Scholar]
  • 15.Baranyi M, Porceddu PF, Gölöncsér F, et al. Novel (hetero)arylalkenyl propargylamine compounds are protective in toxin-induced models of Parkinson’s disease. Mol Neurodegener. 2016;11:6. doi: 10.1186/s13024-015-0067-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Birkmayer W, Hornykiewicz O. The l-dihydroxyphenylalanine (l-DOPA) effect in Parkinson’s syndrome in man: on the pathogenesis and treatment of Parkinson akinesis. Arch Psychiatr Nervenkrankh Z Gesamte Neurol Psychiatr. 1962;203:560–574. doi: 10.1007/BF00343235. [DOI] [PubMed] [Google Scholar]
  • 17.Birkmayer W, Hornykiewicz O. Additional experimental studies on l-DOPA in Parkinson’ syndrome and Reserpine Parkinsonism. Arch Psychiatr Nervenkrankh. 1964;206:367–381. doi: 10.1007/BF00341704. [DOI] [PubMed] [Google Scholar]
  • 18.Cotzias GC, Van Woert MH, Schiffer LM. Aromatic amino acids and modification of Parkinsonism. N Engl J Med. 1967;276:374–379. doi: 10.1056/NEJM196702162760703. [DOI] [PubMed] [Google Scholar]
  • 19.Cotzias GC, Papavasiliou PS, Gellene R. Modification of Parkinonism-chronic treatment with l-DOPA. N Engl J Med. 1969;280:337–345. doi: 10.1056/NEJM196902132800701. [DOI] [PubMed] [Google Scholar]
  • 20.Navarro G, Borroto-Escuela DO, Fuxe K, et al. Purinergic signaling in Parkinson’s disease. Relevance for treatment. Neuropharmacology. 2016;104:161–168. doi: 10.1016/j.neuropharm.2015.07.024. [DOI] [PubMed] [Google Scholar]
  • 21.Lang AE. When and how should treatment be started in Parkinson disease? Neurology. 2009;72:S39–S43. doi: 10.1212/WNL.0b013e318198e177. [DOI] [PubMed] [Google Scholar]
  • 22.Olanow CW, Stern MB, Sethi K. The scientific and clinical basis for the treatment of Parkinson disease. Neurology. 2009;72:S1–S136. doi: 10.1212/WNL.0b013e3181a1d44c. [DOI] [PubMed] [Google Scholar]
  • 23.Pinna A, Serra M, Morelli M, et al. Role of adenosine A2A receptors in motor control: relevance to Parkinson’s disease and dyskinesia. J Neural Transm (Vienna) 2018;125:1273–1286. doi: 10.1007/s00702-018-1848-6. [DOI] [PubMed] [Google Scholar]
  • 24.Burnstock G, Campbell G, Satchell D, et al. Evidence that adenosine triphosphate or a related nucleotide is the transmitter substance released by non-adrenergic inhibitory nerves in the gut. Br J Pharmacol. 1970;40:668–688. doi: 10.1111/j.1476-5381.1970.tb10646.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Burnstock G. Purinergic nerves. Pharmacol Rev. 1972;24:509–581. [PubMed] [Google Scholar]
  • 26.Burnstock G. Do some nerve cells release more than one transmitter? Neuroscience. 1976;1:239–248. doi: 10.1016/0306-4522(76)90054-3. [DOI] [PubMed] [Google Scholar]
  • 27.Burnstock G. The past, present and future of purine nucleotides as signaling molecules. Neuropharmacology. 1997;36:1127–1139. doi: 10.1016/s0028-3908(97)00125-1. [DOI] [PubMed] [Google Scholar]
  • 28.Burnstock G. Purinergic cotransmission. Exp Physiol. 2009;94:20–24. doi: 10.1113/expphysiol.2008.043620. [DOI] [PubMed] [Google Scholar]
  • 29.Potter P, White TD. Release of adenosine 5′-triphosphate from synaptosomes from different regions of rat brain. Neuroscience. 1980;5:1351–1356. doi: 10.1016/0306-4522(80)90207-9. [DOI] [PubMed] [Google Scholar]
  • 30.Poelchen W, Sieler D, Wirkner K, et al. Co-transmitter function of ATP in central catecholaminergic neurons of the rat. Neuroscience. 2001;102:593–602. doi: 10.1016/s0306-4522(00)00529-7. [DOI] [PubMed] [Google Scholar]
  • 31.Sperlágh B, Sershen H, Lajtha A, et al. Co-release of endogenous ATP and [3H]noradrenaline from rat hypothalamic slices: origin and modulation by α2-adrenoreceptors. Neuroscience. 1998;82:511–520. doi: 10.1016/s0306-4522(97)00306-0. [DOI] [PubMed] [Google Scholar]
  • 32.Jo YH, Role LW. Coordinate release of ATP and GABA at in vitro synapses of lateral hypothalamic neurons. J Neurosci. 2002;22:4794–4804. doi: 10.1523/JNEUROSCI.22-12-04794.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Mori M, Heuss C, Gahwiler BH, et al. Fast synaptic transmission mediated by P2X receptors in CA3 pyramidal cells of rat hippocampal slice cultures. J Physiol. 2001;535:115–123. doi: 10.1111/j.1469-7793.2001.t01-1-00115.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Krügel U, Kittner H, Franke H, et al. Purinergic modulation of neuronal activity in the mesolimbic dopaminergic system in vivo. Synapse. 2003;47:134–142. doi: 10.1002/syn.10162. [DOI] [PubMed] [Google Scholar]
  • 35.Zimmermann H. Ectonucleotidases: some developments and a note on nomenclature. Drug Dev Res. 2001;52:44–56. [Google Scholar]
  • 36.Zimmermann H. Ectonucleotidases in nervous system. Purinergic Signal Neuron–Glia Interact. 2006;276:113–130. [Google Scholar]
  • 37.Burnstock G. A basis for distinguishing two types of purinergic receptor. In: Straub RW, Bolis L, editors. Cell membrane receptors for drugs and hormones: a multidisciplinary approach. New York: Raven Press; 1978. pp. 107–118. [Google Scholar]
  • 38.Burnstock G. Purine and pyrimidine receptors. Cell Mol Life Sci. 2007;64:1471–1483. doi: 10.1007/s00018-007-6497-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Ciruela F, Albergaria C, Soriano C, et al. Adenosine receptors interacting proteins (ARIPs): behind the biology of adenosine signaling. Biochim Biophys Acta. 2010;1798:9–20. doi: 10.1016/j.bbamem.2009.10.016. [DOI] [PubMed] [Google Scholar]
  • 40.Burnstock G. Purinergic signalling: from discovery to current developments. Exp Physiol. 2014;99:16–34. doi: 10.1113/expphysiol.2013.071951. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Khakh BS, Burnstock G, Kennedy C, et al. International Union Of Pharmacology. XXIV. Current status of the nomenclature and properties of P2X receptors and their subunits. Pharmacol Rev. 2001;53:107–118. [PubMed] [Google Scholar]
  • 42.Surprenant A, North RA. Signaling at purinergic P2X receptors. Annu Rev Physiol. 2009;71:333–359. doi: 10.1146/annurev.physiol.70.113006.100630. [DOI] [PubMed] [Google Scholar]
  • 43.Puchalowicz K, Baranowska-Bosiacka I, Dziedziejko V, et al. Purinergic signaling and the functioning of the nervous system cells. Cell Mol Biol Lett. 2015;20:867–918. doi: 10.1515/cmble-2015-0050. [DOI] [PubMed] [Google Scholar]
  • 44.Dubyak GR, el-Moatassim C. Signal transduction via P2-purinergic receptors for extracellular ATP and other nucleotides. Am J Physiol. 1993;265:C577–C606. doi: 10.1152/ajpcell.1993.265.3.C577. [DOI] [PubMed] [Google Scholar]
  • 45.Abbracchio MP, Burnstock G, Boeynaems JM, et al. International Union of Pharmacology LVIII: update on the P2Y G protein coupled nucleotide receptors: from molecular mechanisms and pathophysiology to therapy. Pharmacol Rev. 2006;58:281–341. doi: 10.1124/pr.58.3.3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Oliveira A, Illes P, Ulrich H. Purinergic receptors in embryonic and adult neurogenesis. Neuropharmacology. 2016;104:272–281. doi: 10.1016/j.neuropharm.2015.10.008. [DOI] [PubMed] [Google Scholar]
  • 47.Beamer E, Gölöncsér F, Horváth G, et al. Purinergic mechanisms in neuroinflammation: an update from molecules to behavior. Neuropharmacology. 2016;104:94–104. doi: 10.1016/j.neuropharm.2015.09.019. [DOI] [PubMed] [Google Scholar]
  • 48.Madeira MH, Boia R, Ambrósio AF, et al. Having a coffee break: the impact of caffeine consumption on microglia-mediated inflammation in neurodegenerative diseases. Mediat Inflamm. 2017;2017:1–12. doi: 10.1155/2017/4761081. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Przybyla T, Sakowicz-Burkiewicz M, Pawelczyk T. Purinergic signaling in B cells. Acta Biochim Pol. 2018;65:1–7. doi: 10.18388/abp.2017_1588. [DOI] [PubMed] [Google Scholar]
  • 50.Allard B, Beavis PA, Darcy PK, et al. Immunosuppressive activities of adenosine in cancer. Curr Opin Pharmacol. 2016;29:7–16. doi: 10.1016/j.coph.2016.04.001. [DOI] [PubMed] [Google Scholar]
  • 51.Vijayan D, Young A, Teng M, et al. Targeting immunosuppressive adenosine in cancer. Nat Rev Cancer. 2017;17:709–724. doi: 10.1038/nrc.2017.86. [DOI] [PubMed] [Google Scholar]
  • 52.Whiteside TL. Targeting adenosine in cancer immunotherapy: a review of recent progress. Expert Rev Anticancer Ther. 2017;17:527–535. doi: 10.1080/14737140.2017.1316197. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Kazemi MH, Raoofi Mohseni S, Hojjat-Farsangi M, et al. Adenosine and adenosine receptors in the immunopathogenesis and treatment of cancer. J Cell Physiol. 2018;233:2032–2057. doi: 10.1002/jcp.25873. [DOI] [PubMed] [Google Scholar]
  • 54.Burnstock G, Fredholm BB, Verkhratsky A. Adenosine and ATP receptors in the brain. Curr Top Med Chem. 2011;11:973–1011. doi: 10.2174/156802611795347627. [DOI] [PubMed] [Google Scholar]
  • 55.Stockwell J, Jakova E, Cayabyab FS. Adenosine A1 and A2A receptors in the brain: current research and their role in neurodegeneration. Molecules. 2017;22:676. doi: 10.3390/molecules22040676. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Lindberg D, Shan D, Ayers-Ringler J, et al. Purinergic signaling and energy homeostasis in psychiatric disorders. Curr Mol Med. 2015;15:275–295. doi: 10.2174/1566524015666150330163724. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Csóka B, Töro G, Vindeirinho J, et al. A2A adenosine receptors control pancreatic dysfunction in high-fat-diet-induced obesity. FASEB J. 2017;31:4985–4997. doi: 10.1096/fj.201700398R. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Parpura V, Fisher ES, Lechleiter JD, et al. Glutamate and ATP at the interface between signaling and metabolism in astroglia: examples from pathology. Neurochem Res. 2017;42:19–34. doi: 10.1007/s11064-016-1848-6. [DOI] [PubMed] [Google Scholar]
  • 59.Tozzi M, Novak I. Purinergic receptors in adipose tissue as potential targets in metabolic disorders. Front Pharmacol. 2017;8:878. doi: 10.3389/fphar.2017.00878. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Labazi H, Teng B, Mustafa SJ. Functional changes in vascular reactivity to adenosine receptor activation in type I diabetic mice. Eur J Pharmacol. 2018;820:191–197. doi: 10.1016/j.ejphar.2017.12.034. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Ortiz R, Ulrich H, Zarate CA, et al. Purinergic system dysfunction in mood disorders: a key target for developing improved therapeutics. Prog Neuropsychopharmacol Biol Psychiatry. 2015;57:117–131. doi: 10.1016/j.pnpbp.2014.10.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Krügel U. Purinergic receptors is psychiatric disorders. Neuropharmacology. 2016;104:212–225. doi: 10.1016/j.neuropharm.2015.10.032. [DOI] [PubMed] [Google Scholar]
  • 63.Cheffer A, Castillo AR, Corrêa-Velloso JC, et al. Purinergic system in psychiatric diseases. Mol Psychiatry. 2017;23:94–106. doi: 10.1038/mp.2017.188. [DOI] [PubMed] [Google Scholar]
  • 64.Illes P, Verkhratsky A. Purinergic neurone–glia signalling in cognitive-related pathologies. Neuropharmacology. 2016;104:62–75. doi: 10.1016/j.neuropharm.2015.08.005. [DOI] [PubMed] [Google Scholar]
  • 65.Burnstock G, Arnett TR, Orriss IR. Purinergic signaling in the musculoskeletal system. Purinergic Signal. 2013;9:541–572. doi: 10.1007/s11302-013-9381-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Safarzadeh E, Jadidi-Niaragh F, Motallebnezhad M, et al. The role of adenosine and adenosine receptors in the immunopathogenesis of multiple sclerosis. Inflamm Res. 2016;65:511–520. doi: 10.1007/s00011-016-0936-z. [DOI] [PubMed] [Google Scholar]
  • 67.Pascual O, Casper KB, Kubera C, et al. Astrocytic purinergic signaling coordinates synaptic networks. Science. 2005;310:113–116. doi: 10.1126/science.1116916. [DOI] [PubMed] [Google Scholar]
  • 68.Scemes E, Suadicani SO, Dahl G, et al. Connexin and pannexin mediated cell-cell communication. Neuron–Glia Biol. 2007;3:199–208. doi: 10.1017/S1740925X08000069. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Abbrachio MP, Burnstock G, Verkhratsky A, et al. Purinergic signaling in the nervous system: an overview. Trends Neurosci. 2009;32:19–29. doi: 10.1016/j.tins.2008.10.001. [DOI] [PubMed] [Google Scholar]
  • 70.Lapato AS, Tiwari-Woodruff SK. Connexins and pannexins: at the junction of neuro-glial homeostasis and disease. J Neurosci Res. 2017;96:31–44. doi: 10.1002/jnr.24088. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Vizi ES, Knoll J. The inhibitory effect of adenosine and related nucleotides on the release of acetylcholine. Neuroscience. 1976;1:391–398. doi: 10.1016/0306-4522(76)90132-9. [DOI] [PubMed] [Google Scholar]
  • 72.Dunwiddie TV. The physiological role of adenosine in the central nervous system. Int Rev Neurobiol. 1985;27:63–139. doi: 10.1016/s0074-7742(08)60556-5. [DOI] [PubMed] [Google Scholar]
  • 73.Cunha RA, Ribeiro JA. ATP as a presynaptic modulator. Life Sci. 2000;68:119–137. doi: 10.1016/s0024-3205(00)00923-1. [DOI] [PubMed] [Google Scholar]
  • 74.Wall MJ, Dale N. Auto-inhibition of rat parallel fibre-Purkinje cell synapses by activity-dependent adenosine release. J Physiol. 2007;581:553–565. doi: 10.1113/jphysiol.2006.126417. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Fredholm BB, IJzerman AP, Jacobson KA, et al. International Union of Basic and Clinical Pharmacology. LXXXI. Nomenclature and classification of adenosine receptors—an update. Pharmacol Rev. 2011;63:1–34. doi: 10.1124/pr.110.003285. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Chen JF, Pedata F. Modulation of ischemic brain injury and neuroinflammation by adenosine A2A receptors. Curr Pharm Des. 2008;14:1490–1499. doi: 10.2174/138161208784480126. [DOI] [PubMed] [Google Scholar]
  • 77.Pedata F, Dettori I, Coppi E, et al. Purinergic signalling in brain ischemia. Neuropharmacology. 2016;104:105–130. doi: 10.1016/j.neuropharm.2015.11.007. [DOI] [PubMed] [Google Scholar]
  • 78.Latini S, Pedata F. Adenosine in the central nervous system: release mechanisms and extracellular concentrations. J Neurochem. 2001;79:463–484. doi: 10.1046/j.1471-4159.2001.00607.x. [DOI] [PubMed] [Google Scholar]
  • 79.Augood SJ, Emson PC. Adenosine A2A receptor mRNA is expressed by enkephalin cells but not somatostatin cells in rat striatum: a co-expression study. Mol Brain Res. 1994;22:204–210. doi: 10.1016/0169-328x(94)90048-5. [DOI] [PubMed] [Google Scholar]
  • 80.Dixon AK, Gubitz AK, Sirinathsinghji DJ, et al. Tissue distribution of adenosine receptor mRNAs in the rat. Br J Pharmacol. 1996;118:1461–1468. doi: 10.1111/j.1476-5381.1996.tb15561.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Sebastião AM, Ribeiro JA. Adenosine receptors and the central nervous system. Handb Exp Pharmacol. 2009;193:471–534. doi: 10.1007/978-3-540-89615-9_16. [DOI] [PubMed] [Google Scholar]
  • 82.Feoktistov I, Biaggioni I. Adenosine A2B receptors. Pharmacol Rev. 1997;49:381–402. [PubMed] [Google Scholar]
  • 83.Hammarberg C, Schulte G, Fredholm BB. Evidence for functional adenosine A3 receptors in microglia cells. J Neurochem. 2003;86:1051–1054. doi: 10.1046/j.1471-4159.2003.01919.x. [DOI] [PubMed] [Google Scholar]
  • 84.Rivkees SA, Thevananther S, Hao H. Are A3 adenosine receptors expressed in the brain? NeuroReport. 2000;11:1025–1030. doi: 10.1097/00001756-200004070-00026. [DOI] [PubMed] [Google Scholar]
  • 85.Burnstock G, Knight GE. Cellular distribution and functions of P2 receptor subtypes in different systems. Int Rev Cytol. 2004;240:31–304. doi: 10.1016/S0074-7696(04)40002-3. [DOI] [PubMed] [Google Scholar]
  • 86.Guo W, Xu X, Gao X, et al. Expression of P2X5 receptors in the mouse central nervous system. Neuroscience. 2008;128:697–712. doi: 10.1016/j.neuroscience.2008.07.062. [DOI] [PubMed] [Google Scholar]
  • 87.Amadio S, Montilli C, Picconi B, et al. Mapping P2X and P2Y receptor proteins in striatum and substantia nigra: an immunohistological study. Purinergic Signal. 2007;3:389–398. doi: 10.1007/s11302-007-9069-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Moore D, Chambers J, Waldvogel H, et al. Regional and cellular distribution of the P2Y1 purinergic receptor in the human brain: striking neuronal localization. J Comp Neurol. 2000;421:374–384. doi: 10.1002/(sici)1096-9861(20000605)421:3<374::aid-cne6>3.0.co;2-z. [DOI] [PubMed] [Google Scholar]
  • 89.Miras-Portugal MT, Marìn-García P, Carrasquera LM, et al. Physiological role of extracellular nucleotides at the central nervous system: signaling through P2X and P2Y receptors. An R Acad Nac Farm. 2007;73:1127–1157. [Google Scholar]
  • 90.Verkhratsky A, Krishtal OA, Burnstock G. Purinoreceptors in neuroglia. Mol Neurobiol. 2009;39:190–208. doi: 10.1007/s12035-009-8063-2. [DOI] [PubMed] [Google Scholar]
  • 91.Fukumitsu N, Ishii K, Kimura Y, et al. Adenosine A1 receptor mapping of the human brain by PET with 8-dicyclopropylmethyl-1-11C-methyl-3-propylxanthine. J Nucl Med. 2005;46:32–37. [PubMed] [Google Scholar]
  • 92.Ishiwata K, Mishina M, Kimura Y, et al. First visualization of adenosine A(2A) receptors in the human brain by positron emission tomography with [11C]TMSX. Synapse. 2005;55:133–136. doi: 10.1002/syn.20099. [DOI] [PubMed] [Google Scholar]
  • 93.Sheth S, Brito R, Mukherjea D, et al. Adenosine receptors: expression, function and regulation. Int J Mol Sci. 2014;15:2024–2052. doi: 10.3390/ijms15022024. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Abbracchio MP, Burnstock G. Purinergic signaling: pathophysiological roles. Jpn J Pharmacol. 1998;78:113–145. doi: 10.1254/jjp.78.113. [DOI] [PubMed] [Google Scholar]
  • 95.Fields D, Burnstock G. Purinergic signaling in neuron–glial interactions. Nat Neurosci Rev. 2006;7:423–436. doi: 10.1038/nrn1928. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Parpura V, Zorec R. Gliotransmission: exocytotic release from astrocytes. Brain Res Rev. 2010;63:83–92. doi: 10.1016/j.brainresrev.2009.11.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Matute C, Cavaliere F. Neuroglial interactions mediated by purinergic signaling in the pathophysiology of CNS disorders. Semin Cell Dev Biol. 2011;22:252–259. doi: 10.1016/j.semcdb.2011.02.011. [DOI] [PubMed] [Google Scholar]
  • 98.Verderio C, Matteoli M. ATP in neuron–glia bidirectional signaling. Brain Res Rev. 2011;66:106–114. doi: 10.1016/j.brainresrev.2010.04.007. [DOI] [PubMed] [Google Scholar]
  • 99.Cunha RA. How does adenosine control neuronal dysfunction and neurodegeneration? J Neurochem. 2016;139:1019–1055. doi: 10.1111/jnc.13724. [DOI] [PubMed] [Google Scholar]
  • 100.Borea PA, Gessi S, Merighi S, et al. Pathological overproduction: the bad side of adenosine. Br J Pharmacol. 2017;174:1945–1960. doi: 10.1111/bph.13763. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Faas MM, Sáez T, de Vos P. Extracellular ATP and adenosine: the Yin and Yang in immune responses? Mol Asp Med. 2017;55:9–19. doi: 10.1016/j.mam.2017.01.002. [DOI] [PubMed] [Google Scholar]
  • 102.Miras-Portugal MT, Sebastian-Serrano Á, de Diego GarcíaL, et al. Neuronal P2X7 receptor: involvement in neuronal physiology and pathology. J Neurosci. 2017;37:7063–7072. doi: 10.1523/JNEUROSCI.3104-16.2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Burnstock G. An introduction to the roles of purinergic signaling in neurodegeneration, neuroprotection and neuroregeneration. Neuropharmacology. 2016;104:4–17. doi: 10.1016/j.neuropharm.2015.05.031. [DOI] [PubMed] [Google Scholar]
  • 104.Popat RA, Van Den Eeden SK, Tanner C, et al. Coffee, ADORA2A, and CYP1A2: the caffeine connection in Parkinson’s disease. Eur J Neurol. 2011;18:756–765. doi: 10.1111/j.1468-1331.2011.03353.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Kumar PM, Paing SS, Li H, et al. Differential effect of caffeine intake in subjects with genetic susceptibility to Parkinson’s disease. Sci Rep. 2015;5:15492. doi: 10.1038/srep15492. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Yamada-Fowler N, Frekdrikson M, Söderkvist P. Caffeine interaction with glutamate receptor gene GRIN2A: Parkinson’s disease in Swedish population. PLoS ONE. 2014;9:e99294. doi: 10.1371/journal.pone.0099294. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Simon DK, Wu C, Tilley BC, et al. Caffeine, creatine, GRIN2A and Parkinson’s disease progression. J Neurol Sci. 2017;375:355–359. doi: 10.1016/j.jns.2017.02.032. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Liu H, Han X, Li Y, et al. Association of P2X7 receptor gene polymorphisms with sporadic Parkinson’s disease in a Han Chinese population. Neurosci Lett. 2013;546:42–45. doi: 10.1016/j.neulet.2013.04.049. [DOI] [PubMed] [Google Scholar]
  • 109.Schiffmann SN, Fisone G, Moresco R, et al. Adenosine A2A receptors and basal ganglia physiology. Prog Neurobiol. 2007;83:277–292. doi: 10.1016/j.pneurobio.2007.05.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Fuxe K, Marcellino D, Genedani S, et al. Adenosine A(2A) receptors, dopamine D(2) receptors and their interactions in Parkinson’s disease. Mov Disord. 2007;22:1990–2017. doi: 10.1002/mds.21440. [DOI] [PubMed] [Google Scholar]
  • 111.Olivieira-Giacomelli Á, Naaldijk Y, Sardá-Arroyo L, et al. Purinergic receptors in neurological diseases with motor symptoms: targets for therapy. Front Pharmacol. 2018;9:325. doi: 10.3389/fphar.2018.00325. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Fuxe K, Borroto-Escuela DO, Marcellino D, et al. GPCR heteromers and their allosteric receptor–receptor interactions. Curr Med Chem. 2012;19:356–363. doi: 10.2174/092986712803414259. [DOI] [PubMed] [Google Scholar]
  • 113.Armentero MT, Pinna A, Ferre S, et al. Past, present and future of A(2A) adenosine receptor antagonists in the therapy of Parkinson’s disease. Pharmacol Ther. 2011;132:280–299. doi: 10.1016/j.pharmthera.2011.07.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Bogenpohl JW, Ritter SL, Hall RA, et al. Adenosine A2A receptor in the monkey basal ganglia: ultrastructural localization and colocalization with the metabotropic glutamate receptor 5 in the striatum. J Comp Neurol. 2012;520:570–589. doi: 10.1002/cne.22751. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Łukasiewicz S, Blasiak E, Faron-Gorecka A, et al. Fluorescence studies of homooligomerization of adenosine A2A and serotonin 5-HT1A receptors reveal the specificity of receptor interactions in the plasma membrane. Pharmacol Rep. 2007;59:379–392. [PubMed] [Google Scholar]
  • 116.Carriba P, Navarro G, Ciruela F, et al. Detection of heteromerization of more than two proteins by sequential BRET–FRET. Nat Methods. 2008;5:727–733. doi: 10.1038/nmeth.1229. [DOI] [PubMed] [Google Scholar]
  • 117.Navarro G, Carriba P, Gandía J, et al. Detection of heteromers formed by cannabinoid CB1, dopamine D2, and adenosine A2A G-protein-coupled receptors by combining bimolecular fluorescence complementation and bioluminescence energy transfer. Sci World J. 2008;8:1088–1097. doi: 10.1100/tsw.2008.136. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Fuxe K, Ungerstedt U. Action of caffeine and theophyllamine on supersensitive dopamine receptors: considerable enhancement of receptor response to treatment with DOPA and dopamine receptor agonists. Med Biol. 1974;52:48–54. [PubMed] [Google Scholar]
  • 119.Fredholm BB, Fuxe K, Agnati L. Effect of some phosphodiesterase inhibitors on central dopamine mechanisms. Eur J Pharmacol. 1976;38:31–38. doi: 10.1016/0014-2999(76)90198-9. [DOI] [PubMed] [Google Scholar]
  • 120.Kanda T, Jackson MJ, Smith LA, et al. Combined use of the adenosine A2A antagonist KW-6002 with L-DOPA or with selective D1 or D2 dopamine agonists increases antiparkinsonian activity but not dyskinesia in MPTP-treated monkeys. Exp Neurol. 2000;162:321–327. doi: 10.1006/exnr.2000.7350. [DOI] [PubMed] [Google Scholar]
  • 121.Fuzzati-Armentero MT, Cerri S, Levandis G, et al. Dual target strategy: combining distinct non-dopaminergic treatments reduces neuronal cell loss and synergistically modulates l-DOPA-induced rotational behavior in a rodent model of Parkinson’s disease. J Neurochem. 2015;134:740–747. doi: 10.1111/jnc.13162. [DOI] [PubMed] [Google Scholar]
  • 122.Canals M, Burgueno J, Marcellino D, et al. Homodimerization of adenosine A2A receptors: qualitative and quantitative assessment by fluorescence and bioluminescence energy transfer. J Neurochem. 2004;88:726–734. doi: 10.1046/j.1471-4159.2003.02200.x. [DOI] [PubMed] [Google Scholar]
  • 123.Lee SP, O’Dowd BF, George SR. Homo- and hetero-oligomerization of G protein-coupled receptors. Life Sci. 2003;74:173–180. doi: 10.1016/j.lfs.2003.09.028. [DOI] [PubMed] [Google Scholar]
  • 124.Guo W, Urizar E, Kralikova M, et al. Dopamine D2 receptors form higher order oligomers at physiological expression levels. EMBO J. 2008;27:2293–2304. doi: 10.1038/emboj.2008.153. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Antonelli T, Fuxe K, Agnati L, et al. Experimental studies and theoretical aspects on A2A/D2 receptor interactions in a model of Parkinson’s disease. Relevance for l-DOPA induced dyskinesias. J Neurol Sci. 2006;248:16–22. doi: 10.1016/j.jns.2006.05.019. [DOI] [PubMed] [Google Scholar]
  • 126.Fuxe K, Marcellino D, Borroto-Escuela DO, et al. Adenosine-dopamine interactions in the pathophysiology and treatment of CNS disorders. CNS Neurosci Ther. 2010;16:e18–e42. doi: 10.1111/j.1755-5949.2009.00126.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Fuxe K, Guidolin D, Agnati LF, et al. Dopamine heteroreceptor complexes as therapeutic targets in Parkinson’s disease. Expert Opin Ther Targets. 2015;19:377–398. doi: 10.1517/14728222.2014.981529. [DOI] [PubMed] [Google Scholar]
  • 128.Poewe W, Mahlknecht P, Jankovic J. Emerging therapies for Parkinson’s disease. Curr Opin Neurol. 2012;25:448–459. doi: 10.1097/WCO.0b013e3283542fde. [DOI] [PubMed] [Google Scholar]
  • 129.Hauser RA. Future treatments for Parkinson’s disease: surfing the PD pipeline. Int J Neurosci. 2011;121(Suppl 2):53–62. doi: 10.3109/00207454.2011.620195. [DOI] [PubMed] [Google Scholar]
  • 130.Zhu C, Wang G, Li J, et al. Adenosine A2A receptor antagonist istradefylline 20 versus 40 mg/day as augmentation for Parkinson’s disease: a meta-analysis. Neurol Res. 2014;36:1028–1034. doi: 10.1179/1743132814Y.0000000375. [DOI] [PubMed] [Google Scholar]
  • 131.Dungo R, Deeks ED. Istradefylline: first global approval. Drugs. 2013;73:875–882. doi: 10.1007/s40265-013-0066-7. [DOI] [PubMed] [Google Scholar]
  • 132.Hauser RA, Stocchi F, Rascol O, et al. Preladenant as an adjunctive therapy with levodopa in Parkinson disease: two randomized clinical trials and lessons learned. JAMA Neurol. 2015;72:1491–1500. doi: 10.1001/jamaneurol.2015.2268. [DOI] [PubMed] [Google Scholar]
  • 133.Pinna A. Adenosine A2A receptor antagonists in Parkinson’s disease: progress in clinical trials from the newly approved istradefylline to drugs in early development and those already discontinued. CNS Drugs. 2014;28:455–474. doi: 10.1007/s40263-014-0161-7. [DOI] [PubMed] [Google Scholar]
  • 134.Gillespie RJ, Bamford SJ, Botting R, et al. Antagonists of the human A(2A) adenosine receptor. 4. Design, synthesis, and preclinical evaluation of 7-aryltriazolol[4,5-d]pyrimidines. J Med Chem. 2009;52:33–47. doi: 10.1021/jm800961g. [DOI] [PubMed] [Google Scholar]
  • 135.Sitkovsky MV, Hatfield S, Abbott R, et al. Hostile, hypoxia-A2-adenosinergic tumor biology as the next barrier to overcome for tumor immunologists. Cancer Immunol Res. 2014;2:598–605. doi: 10.1158/2326-6066.CIR-14-0075. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Hove-Madsen L, Prat-Vidal C, Llach A, et al. Adenosine A2A receptors are expressed in human atrial myocytes and modulate spontaneous sarcoplasmic reticulum calcium release. Cardiovasc Res. 2006;72:292–302. doi: 10.1016/j.cardiores.2006.07.020. [DOI] [PubMed] [Google Scholar]
  • 137.Llach A, Molina CE, Prat-Vidal C, et al. Abnormal calcium handling in atrial fibrillation is linked to up-regulation of adenosine A2A receptors. Eur Heart J. 2011;32:721–729. doi: 10.1093/eurheartj/ehq464. [DOI] [PubMed] [Google Scholar]
  • 138.Hauser RA, Olanow CW, Kieburtz KD, et al. Tozadenant (SYN115) in patients with Parkinson’s disease who have motor fluctuations on levodopa: a phase 2b, double-blind, randomised trial. Lancet Neurol. 2014;13:767–776. doi: 10.1016/S1474-4422(14)70148-6. [DOI] [PubMed] [Google Scholar]
  • 139.LeWitt PA, Guttman M, Tetrud JW, et al. Adenosine A2A receptor antagonist istradefylline (KW-6002) reduces “off” time in Parkinson’s disease: a double-blind, randomized, multicenter clinical trial (6002-US-005) Ann Neurol. 2008;63:295–302. doi: 10.1002/ana.21315. [DOI] [PubMed] [Google Scholar]
  • 140.Fernandez HH, Greeley DR, Zweig RM, et al. Istradefylline as monotherapy for Parkinson disease: results of the 6002-US-051 trial. Parkinsonism Relat Disord. 2010;16:16–20. doi: 10.1016/j.parkreldis.2009.06.008. [DOI] [PubMed] [Google Scholar]
  • 141.Gyoneva S, Shapiro L, Lazo C, et al. Adenosine A2A receptor antagonism reverses inflammation-induced impairment of microglial process extension in a model of Parkinson’s disease. Neurobiol Dis. 2014;67:191–202. doi: 10.1016/j.nbd.2014.03.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Golembiowska K, Wardas J, Noworyta-Sokolowska K, Kaminska K, et al. Effects of adenosine receptor antagonists on the in vivo LPS-induced inflammation model of Parkinson’s disease. Neurotox Res. 2013;24:29–40. doi: 10.1007/s12640-012-9372-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143.Villar-Menéndez I, Porta S, Buira SP, et al. Increased striatal adenosine A2A receptor levels is an early event in Parkinson’s disease-related pathology and it is potentially regulated by miR-34b. Neurobiol Dis. 2014;69:206–214. doi: 10.1016/j.nbd.2014.05.030. [DOI] [PubMed] [Google Scholar]
  • 144.Hu Q, Ren X, Liu Y, et al. Aberrant adenosine A2A receptor signaling contributes to neurodegeneration and cognitive impairments in a mouse model of synucleinopathy. Exp Neurol. 2016;283:213–223. doi: 10.1016/j.expneurol.2016.05.040. [DOI] [PubMed] [Google Scholar]
  • 145.Kachroo A, Schwarzschild MA. Adenosine A2A receptor gene disruption protects in an α-synuclein model of Parkinson’s disease. Ann Neurol. 2012;71:278–282. doi: 10.1002/ana.22630. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146.Ferreira DG, Batalha VL, Vicente Miranda H, et al. Adenosine A2A receptors modulate α-synuclein aggregation and toxicity. Cereb Cortex. 2015 doi: 10.1093/cercor/bhv268. [DOI] [PubMed] [Google Scholar]
  • 147.Pintor J, Diaz-Rey MA, Miras-Portugal MT. Ap4A and ADP-beta-S binding to P2 purinoreceptors present on rat brain synaptic terminals. Br J Pharmacol. 1993;108:1094–1099. doi: 10.1111/j.1476-5381.1993.tb13510.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148.Rodriguez-Pascual F, Cortes R, Torres M, et al. Distribution of [3H]diadenosine tetraphosphate binding sites in rat brain. Neuroscience. 1997;77:247–255. doi: 10.1016/s0306-4522(96)00424-1. [DOI] [PubMed] [Google Scholar]
  • 149.Burnstock G. Purinergic signalling and disorders of the central nervous system. Nat Rev Drug Discov. 2008;7:575–590. doi: 10.1038/nrd2605. [DOI] [PubMed] [Google Scholar]
  • 150.Heine C, Wegner A, Grosche J, et al. P2 receptor expression in the dopaminergic system of the rat brain during development. Neuroscience. 2007;149:165–181. doi: 10.1016/j.neuroscience.2007.07.015. [DOI] [PubMed] [Google Scholar]
  • 151.Krügel U, Kittner H, Franke H, et al. Stimulation of P2 receptors in the ventral tegmental area enhances dopaminergic mechanisms in vivo. Neuropharmacology. 2001;40:1084–1093. doi: 10.1016/s0028-3908(01)00033-8. [DOI] [PubMed] [Google Scholar]
  • 152.Krügel U, Kittner H, Illes P. Mechanisms of adenosine 5′-triphosphate-induced dopamine release in the rat nucleus accumbens in vivo. Synapse. 2001;39:222–232. doi: 10.1002/1098-2396(20010301)39:3<222::AID-SYN1003>3.0.CO;2-R. [DOI] [PubMed] [Google Scholar]
  • 153.Gan M, Moussaud S, Jiang P, et al. Extracellular ATP induces intracellular alpha-synuclein accumulation via P2X1 receptor-mediated lysosomal dysfunction. Neurobiol Aging. 2015;36:1209–1220. doi: 10.1016/j.neurobiolaging.2014.10.037. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154.Marcellino D, Suarez-Boomgaard D, Sanchez-Reina MD, et al. On the role of P2X7 receptors in dopamine nerve cell degeneration in a rat model of Parkinson’s disease: studies with the P2X7 receptor antagonist A-438079. J Neural Transm. 2010;117:681–687. doi: 10.1007/s00702-010-0400-0. [DOI] [PubMed] [Google Scholar]
  • 155.Carmo MR, Menezes AP, Nunes AC, et al. The P2X7 receptor antagonist Brilliant Blue G attenuates contralateral rotations in a rat model of Parkinsonism through a combined control of synaptotoxicity, neurotoxicity and gliosis. Neuropharmacology. 2014;81:142–152. doi: 10.1016/j.neuropharm.2014.01.045. [DOI] [PubMed] [Google Scholar]
  • 156.Hracskó Z, Baranyi M, Csölle C, et al. Lack of neuroprotection in the absence of P2X7 receptors in toxin-induced animal models of Parkinson’s disease. Mol Neurodegener. 2011;6:28. doi: 10.1186/1750-1326-6-28. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 157.Wilkaniec A, Gassowska M, Czapski GA, et al. P2X7 receptor-pennexin 1 interaction mediates extracellular alpha-synuclein-induced ATP release in neuroblastoma SH-SY5Y cells. Purinergic Signal. 2017;13:347–361. doi: 10.1007/s11302-017-9567-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158.Jiang T, Hoekstra J, Heng X, et al. P2X7 receptor is critical in α-synuclein-mediated microglial NADPH oxidase activation. Neurobiol Aging. 2015;36:2304–2318. doi: 10.1016/j.neurobiolaging.2015.03.015. [DOI] [PubMed] [Google Scholar]
  • 159.Coppi E, Pedata F, Gibb AJ. P2Y1 receptor modulation of Ca2+-activated K+ currents in medium-sized neurons from neonatal rat striatal slices. J Neurophysiol. 2012;107:1009–1021. doi: 10.1152/jn.00816.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 160.Wang Y, Chang CF, Morales M, et al. Diadenosine tetraphosphate protects against injuries induced by ischaemia and 6-hydroxidopamine in rat brain. J Neurosci. 2013;23:7958–7965. doi: 10.1523/JNEUROSCI.23-21-07958.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 161.Yang X, Lou Y, Liu G, et al. Microglia P2Y6 receptor is related to Parkinson’s disease through neuroinflammatory process. J Neuroinflamm. 2017;14:38. doi: 10.1186/s12974-017-0795-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162.Qian Y, Xu S, Yang X, et al. Purinergic receptor P2Y6 contributes to 1-methyl-4-phenylpyridinium-induced oxidative stress and cell death in neuronal SH-SY5Y cells. J Neurosci Res. 2017;96:253–264. doi: 10.1002/jnr.24119. [DOI] [PubMed] [Google Scholar]
  • 163.Neher JJ, Neniskyte U, Hornik T, et al. Inhibition of UDP/P2Y6 purinergic signaling prevents phagocytosis of viable neurons by activated microglia in vitro and in vivo. Glia. 2014;62:1463–1465. doi: 10.1002/glia.22693. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 164.Dunkel P, Chai CL, Sperlágh B, et al. Clinical utility of neuroprotective agents in neurodegenerative diseases: current status of drug development for Alzheimer’s, Parkinson’s and Huntington’s diseases, and amyotrophic lateral sclerosis. Expert Opin Investig Drugs. 2012;21:1267–1308. doi: 10.1517/13543784.2012.703178. [DOI] [PubMed] [Google Scholar]

Articles from Neurochemical Research are provided here courtesy of Springer

RESOURCES