Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 12 de 12
1.
ACS Chem Neurosci ; 14(7): 1299-1309, 2023 04 05.
Article En | MEDLINE | ID: mdl-36881648

Increased ATP release and its extracellular catabolism through CD73 (ecto-5'-nucleotidase) lead to the overactivation of adenosine A2A receptors (A2AR), which occurs in different brain disorders. A2AR blockade blunts mood and memory dysfunction caused by repeated stress, but it is unknown if increased ATP release coupled to CD73-mediated formation of extracellular adenosine is responsible for A2AR overactivation upon repeated stress. This was now investigated in adult rats subject to repeated stress for 14 consecutive days. Frontocortical and hippocampal synaptosomes from stressed rats displayed an increased release of ATP upon depolarization, coupled to an increased density of vesicular nucleotide transporters and of CD73. The continuous intracerebroventricular delivery of the CD73 inhibitor α,ß-methylene ADP (AOPCP, 100 µM) during restraint stress attenuated mood and memory dysfunction. Slice electrophysiological recordings showed that restraint stress decreased long-term potentiation both in prefrontocortical layer II/III-layer V synapses and in hippocampal Schaffer fibers-CA1 pyramid synapses, which was prevented by AOPCP, an effect occluded by adenosine deaminase and by the A2AR antagonist SCH58261. These results indicate that increased synaptic ATP release coupled to CD73-mediated formation of extracellular adenosine contributes to mood and memory dysfunction triggered by repeated restraint stress. This prompts considering interventions decreasing ATP release and CD73 activity as novel strategies to mitigate the burden of repeated stress.


5'-Nucleotidase , Adenosine , Animals , Rats , 5'-Nucleotidase/metabolism , Adenosine/metabolism , Adenosine Triphosphate/metabolism , Receptor, Adenosine A2A/metabolism , Synapses/metabolism , Synaptosomes/metabolism , Stress, Physiological , Electrophysiological Phenomena
2.
Mol Neurobiol ; 60(3): 1659-1674, 2023 Mar.
Article En | MEDLINE | ID: mdl-36547848

Extracellular ATP can be a danger signal, but its role in striatal circuits afflicted in Parkinson's disease (PD) is unclear and was now investigated. ATP was particularly released at high stimulation intensities from purified striatal nerve terminals of mice, which were endowed with different ATP-P2 receptors (P2R), although P2R antagonists did not alter corticostriatal transmission or plasticity. Instead, ATP was extracellularly catabolized into adenosine through CD73 to activate adenosine A2A receptors (A2AR) modulating corticostriatal long-term potentiation (LTP) in mice. In the presymptomatic phase of a 6-hydroxydopamine rat model of PD, ATP release from striatal nerve terminals was increased and was responsible for a greater impact of CD73 and A2AR on corticostriatal LTP. These observations identify increased ATP release and ATP-derived formation of extracellular adenosine bolstering A2AR activation as a key pathway responsible for abnormal synaptic plasticity in circuits involved in the onset of PD motor symptoms. The translation of these findings to humans prompts extending the use of A2AR antagonists from only co-adjuvants of motor control in Parkinsonian patients to neuroprotective drugs delaying the onset of motor symptoms.


Adenosine , Parkinson Disease , Rats , Humans , Mice , Animals , Adenosine/metabolism , Adenosine Triphosphate/metabolism , Long-Term Potentiation , Neuronal Plasticity
3.
Front Pharmacol ; 9: 133, 2018.
Article En | MEDLINE | ID: mdl-29615897

Adenosine A2A receptors (A2AR) are activated upon increased synaptic activity to assist in the implementation of long-term plastic changes at synapses. While it is reported that A2AR are involved in the control of prefrontal cortex (PFC)-dependent behavior such as working memory, reversal learning and effort-based decision making, it is not known whether A2AR control glutamatergic synapse plasticity within the medial PFC (mPFC). To elucidate that, we tested whether A2AR blockade affects long-term plasticity (LTP) of excitatory post-synaptic potentials in pyramidal neurons and fast spiking (FS) interneurons in layer 5 of the mPFC and of population spikes. Our results show that A2AR are enriched at mPFC synapses, where their blockade reversed the direction of plasticity at excitatory synapses onto layer 5 FS interneurons from LTP to long-term depression, while their blockade had no effect on the induction of LTP at excitatory synapses onto layer 5 pyramidal neurons. At the network level, extracellularly induced LTP of population spikes was reduced by A2AR blockade. The interneuron-specificity of A2AR in controlling glutamatergic synapse LTP may ensure that during periods of high synaptic activity, a proper excitation/inhibition balance is maintained within the mPFC.

4.
Eur J Neurosci ; 47(9): 1127-1134, 2018 05.
Article En | MEDLINE | ID: mdl-29570875

Prefrontal cortex (PFC) circuits are modulated by dopamine acting on D1 - and D2 -like receptors, which are pharmacologically exploited to manage neuropsychiatric conditions. Adenosine A2A receptors (A2A R) also control PFC-related responses and A2A R antagonists are potential anti-psychotic drugs. As tight antagonistic A2A R-D2 R and synergistic A2A R-D1 R interactions occur in other brain regions, we now investigated the crosstalk between A2A R and D1 /D2 R controlling synaptic transmission between layers II/III and V in mouse PFC coronal slices. Dopamine decreased synaptic transmission, a presynaptic effect based on the parallel increase in paired-pulse responses. Dopamine inhibition was prevented by the D2 R-like antagonist sulpiride but not by the D1 R antagonist SCH23390 and was mimicked by the D2 R agonist sumanirole, but not by the agonists of either D4 R (A-412997) or D3 R (PD128907). Dopamine inhibition was prevented by the A2A R antagonist, SCH58261, and attenuated in A2A R knockout mice. Accordingly, triple-labelling immunocytochemistry experiments revealed the co-localization of A2A R and D2 R immunoreactivity in glutamatergic (vGluT1-positive) nerve terminals of the PFC. This reported positive A2A R-D2 R interaction controlling PFC synaptic transmission provides a mechanistic justification for the anti-psychotic potential of A2A R antagonists.


Dopamine Agonists/pharmacology , Prefrontal Cortex/drug effects , Receptor, Adenosine A2A/drug effects , Receptors, Dopamine D2/drug effects , Adenosine/pharmacology , Animals , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Dopamine/pharmacology , Glutamic Acid/pharmacology , Male , Mice, Inbred C57BL , Prefrontal Cortex/metabolism , Receptor, Adenosine A2A/metabolism , Receptors, Dopamine D1/metabolism , Receptors, Dopamine D2/metabolism , Synaptic Transmission/physiology
5.
Cell Death Dis ; 9(3): 297, 2018 02 20.
Article En | MEDLINE | ID: mdl-29463792

Despite the characteristic etiologies and phenotypes, different brain disorders rely on common pathogenic events. Glutamate-induced neurotoxicity is a pathogenic event shared by different brain disorders. Another event occurring in different brain pathological conditions is the increase of the extracellular ATP levels, which is now recognized as a danger and harmful signal in the brain, as heralded by the ability of P2 receptors (P2Rs) to affect a wide range of brain disorders. Yet, how ATP and P2R contribute to neurodegeneration remains poorly defined. For that purpose, we now examined the contribution of extracellular ATP and P2Rs to glutamate-induced neurodegeneration. We found both in vitro and in vivo that ATP/ADP through the activation of P2Y1R contributes to glutamate-induced neuronal death in the rat hippocampus. We found in cultured rat hippocampal neurons that the exposure to glutamate (100 µM) for 30 min triggers a sustained increase of extracellular ATP levels, which contributes to NMDA receptor (NMDAR)-mediated hippocampal neuronal death through the activation of P2Y1R. We also determined that P2Y1R is involved in excitotoxicity in vivo as the blockade of P2Y1R significantly attenuated rat hippocampal neuronal death upon the systemic administration of kainic acid or upon the intrahippocampal injection of quinolinic acid. This contribution of P2Y1R fades with increasing intensity of excitotoxic conditions, which indicates that P2Y1R is not contributing directly to neurodegeneration, rather behaving as a catalyst decreasing the threshold from which glutamate becomes neurotoxic. Moreover, we unraveled that such excitotoxicity process began with an early synaptotoxicity that was also prevented/attenuated by the antagonism of P2Y1R, both in vitro and in vivo. This should rely on the observed glutamate-induced calpain-mediated axonal cytoskeleton damage, most likely favored by a P2Y1R-driven increase of NMDAR-mediated Ca2+ entry selectively in axons. This may constitute a degenerative mechanism shared by different brain diseases, particularly relevant at initial pathogenic stages.


Glutamic Acid/toxicity , Neurodegenerative Diseases/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Receptors, Purinergic P2Y1/metabolism , Adenosine Triphosphate/metabolism , Animals , Cell Death , Female , Glutamic Acid/metabolism , Hippocampus/cytology , Hippocampus/metabolism , Humans , Male , Neurodegenerative Diseases/etiology , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/physiopathology , Neurons/cytology , Neurons/metabolism , Rats , Rats, Wistar , Receptors, N-Methyl-D-Aspartate/genetics , Receptors, Purinergic P2Y1/genetics
6.
Front Mol Neurosci ; 11: 475, 2018.
Article En | MEDLINE | ID: mdl-30618621

Adenosine A2A receptors (A2ARs) were recently described to control synaptic plasticity and network activity in the prefrontal cortex (PFC). We now probed the role of these PFC A2AR by evaluating the behavioral performance (locomotor activity, anxiety-related behavior, cost-benefit decision making and working memory) of rats upon downregulation of A2AR selectively in the prelimbic medial PFC (PLmPFC) via viral small hairpin RNA targeting the A2AR (shA2AR). The most evident alteration observed in shA2AR-treated rats, when compared to sh-control (shCTRL)-treated rats, was a decrease in the choice of the large reward upon an imposed delay of 15 s assessed in a T-maze-based cost-benefit decision-making paradigm, suggestive of impulsive decision making. Spontaneous locomotion in the open field was not altered, suggesting no changes in exploratory behavior. Furthermore, rats treated with shA2AR in the PLmPFC also displayed a tendency for higher anxiety levels in the elevated plus maze (less entries in the open arms), but not in the open field test (time spent in the center was not affected). Finally, working memory performance was not significantly altered, as revealed by the spontaneous alternation in the Y-maze test and the latency to reach the platform in the repeated trial Morris water maze. These findings constitute the first direct demonstration of a role of PFC A2AR in the control of behavior in physiological conditions, showing their major contribution for the control of delay-based cost-benefit decisions.

7.
eNeuro ; 5(6)2018.
Article En | MEDLINE | ID: mdl-30627646

Neurodegeneration is a process transversal to neuropsychiatric diseases and the understanding of its mechanisms should allow devising strategies to prevent this irreversible step in brain diseases. Neurodegeneration caused by seizures is a critical step in the aggravation of temporal lobe epilepsy, but its mechanisms remain undetermined. Convulsions trigger an elevation of extracellular adenosine and upregulate adenosine A2A receptors (A2AR), which have been associated with the control of neurodegenerative diseases. Using the rat and mouse kainate model of temporal lobe epilepsy, we now tested whether A2AR control convulsions-induced hippocampal neurodegeneration. The pharmacological or genetic blockade of A2AR did not affect kainate-induced convulsions but dampened the subsequent neurotoxicity. This neurotoxicity began with a rapid A2AR upregulation within glutamatergic synapses (within 2 h), through local translation of synaptic A2AR mRNA. This bolstered A2AR-mediated facilitation of glutamate release and of long-term potentiation (LTP) in CA1 synapses (4 h), triggered a subsequent synaptotoxicity, heralded by decreased synaptic plasticity and loss of synaptic markers coupled to calpain activation (12 h), that predated overt neuronal loss (24 h). All modifications were prevented by the deletion of A2AR selectively in forebrain neurons. This shows that synaptic A2AR critically control synaptic excitotoxicity, which underlies the development of convulsions-induced neurodegeneration.


Convulsants/toxicity , Kainic Acid/toxicity , Nerve Degeneration/etiology , Nerve Degeneration/metabolism , Neurons/metabolism , Receptor, Adenosine A2A/metabolism , Adenosine A2 Receptor Antagonists/therapeutic use , Amygdala/physiology , Animals , Cells, Cultured , Epilepsy/complications , Epilepsy/drug therapy , Epilepsy/etiology , Hippocampus/drug effects , Hippocampus/physiology , Kindling, Neurologic/drug effects , Kindling, Neurologic/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Nerve Degeneration/prevention & control , Neurons/drug effects , Protein Binding/drug effects , Pyrimidines/therapeutic use , Rats , Rats, Wistar , Receptor, Adenosine A2A/genetics , Synaptic Transmission/drug effects , Synaptic Transmission/genetics , Triazoles/therapeutic use
8.
Cereb Cortex ; 27(1): 718-730, 2017 01 01.
Article En | MEDLINE | ID: mdl-26534909

Abnormal accumulation of aggregated α-synuclein (aSyn) is a hallmark of sporadic and familial Parkinson's disease (PD) and related synucleinopathies. Recent studies suggest a neuroprotective role of adenosine A2A receptor (A2AR) antagonists in PD. Nevertheless, the precise molecular mechanisms underlying this neuroprotection remain unclear. We assessed the impact of A2AR blockade or genetic deletion (A2AR KO) on synaptic plasticity and neuronal cell death induced by aSyn oligomers. We found that impairment of LTP associated with aSyn exposure was rescued in A2AR KO mice or upon A2AR blockade, through an NMDA receptor-dependent mechanism. The mechanisms underlying these effects were evaluated in SH-SY5Y cells overexpressing aSyn and rat primary neuronal cultures exposed to aSyn. Cell death in both conditions was prevented by selective A2AR antagonists. Interestingly, blockade of these receptors did not interfere with aSyn oligomerization but, instead, reduced the percentage of cells displaying aSyn inclusions. Altogether, our data raise the possibility that the well-documented effects of A2AR antagonists involve the control of the latter stages of aSyn aggregation, thereby preventing the associated neurotoxicity. These findings suggest that A2AR represent an important target for the development of effective drugs for the treatment of PD and related synucleinopathies.


Neurons/metabolism , Receptor, Adenosine A2A/metabolism , alpha-Synuclein/metabolism , Adenosine A2 Receptor Antagonists/toxicity , Animals , Cell Death/drug effects , Cell Death/physiology , Cell Line, Tumor , Excitatory Postsynaptic Potentials , Humans , Male , Mice, Inbred C57BL , Mice, Knockout , Neurons/drug effects , Neurons/pathology , Rats, Wistar , Receptor, Adenosine A2A/genetics , Recombinant Proteins/metabolism , Tissue Culture Techniques , alpha-Synuclein/genetics
9.
Neurosci Lett ; 638: 162-166, 2017 01 18.
Article En | MEDLINE | ID: mdl-28007645

GPR37 is an orphan G protein-coupled receptor highly expressed in the brain. The precise function of GPR37 is still unknown, but a number of evidences indicate it modulates the dopaminergic system. Here, we aimed to determine the role of GPR37 on the control of cocaine-mediated electrophysiological effects (synaptic transmission and short-term plasticity) in corticostriatal synapses. Accordingly, we evaluated basal synaptic transmission and paired-pulse stimulation (PPS) in wild-type and GPR37KO mice slices. Regardless of the genotype, a low concentration of cocaine (2µM) did not modify basal synaptic transmission. Conversely, a higher dose of cocaine (30µM) decreased synaptic transmission in both genotypes, although with different intensities: approximately 30% in slices from wild-type mice and 45% in slices from GPR37-KO mice. On the other hand, no differences in PPS ratio were observed between wild-type and GPR37-KO cocaine-treated mice. Overall, our data suggest that GPR37 is involved in cocaine-induced modification of basal synaptic transmission without modifying cocaine effects in short-term plasticity.


Cocaine/pharmacology , Corpus Striatum/drug effects , Dopamine Uptake Inhibitors/pharmacology , Receptors, G-Protein-Coupled/metabolism , Synaptic Transmission/drug effects , Animals , Corpus Striatum/physiology , Male , Mice, Inbred C57BL , Mice, Knockout , Neuronal Plasticity/drug effects , Receptors, G-Protein-Coupled/genetics
10.
Behav Brain Res ; 301: 43-54, 2016 Mar 15.
Article En | MEDLINE | ID: mdl-26707254

Parkinson's disease (PD) is characterized by motor dysfunction associated with dopaminergic degeneration in the dorsolateral striatum (DLS). However, motor symptoms in PD are often preceded by short-term memory deficits, which have been argued to involve deregulation of medial prefrontal cortex (mPFC). We now used a 6-hydroxydopamine (6-OHDA) rat PD model to explore if alterations of synaptic plasticity in DLS and mPFC underlie short-term memory impairments in PD prodrome. The bilateral injection of 6-OHDA (20µg/hemisphere) in the DLS caused a marked loss of dopaminergic neurons in the substantia nigra (>80%) and decreased monoamine levels in the striatum and PFC, accompanied by motor deficits evaluated after 21 days in the open field and accelerated rotarod. A lower dose of 6-OHDA (10µg/hemisphere) only induced a partial degeneration (about 60%) of dopaminergic neurons in the substantia nigra with no gross motor impairments, thus mimicking an early premotor stage of PD. Notably, 6-OHDA (10µg)-lesioned rats displayed decreased monoamine levels in the PFC as well as short-term memory deficits evaluated in the novel object discrimination and in the modified Y-maze tasks; this was accompanied by a selective decrease in the amplitude of long-term potentiation in the mPFC, but not in DLS, without changes of synaptic transmission in either brain regions. These results indicate that the short-term memory dysfunction predating the motor alterations in the 6-OHDA model of PD is associated with selective changes of information processing in PFC circuits, typified by persistent changes of synaptic plasticity.


Long-Term Potentiation/physiology , Memory Disorders/physiopathology , Memory, Short-Term/physiology , Parkinsonian Disorders/physiopathology , Prefrontal Cortex/physiopathology , Animals , Discrimination, Psychological/physiology , Double-Blind Method , Male , Maze Learning/physiology , Memory Disorders/pathology , Motor Activity , Oxidopamine , Parkinsonian Disorders/pathology , Parkinsonian Disorders/psychology , Prefrontal Cortex/pathology , Rats, Wistar , Rotarod Performance Test , Spatial Memory/physiology , Synaptic Transmission/physiology , Tissue Culture Techniques
11.
Mol Neurobiol ; 53(6): 3891-3899, 2016 08.
Article En | MEDLINE | ID: mdl-26164273

The dorsolateral striatum (DLS) processes motor and non-motor functions and undergoes extensive dopaminergic degeneration in Parkinson's disease (PD). The nigrostriatal dopaminergic degeneration also affects other brain areas including the pre-frontal cortex (PFC), which has been associated with the appearance of anhedonia and depression at pre-motor phases of PD. Using behavioral, neurochemical, and electrophysiological approaches, we investigated the temporal dissociation between the role of the DLS and PFC in the appearance of anhedonia and defense behaviors relevant to depression in rats submitted to bilateral DLS lesions with 6-hydroxydopamine (6-OHDA; 10 µg/hemisphere). 6-OHDA induced partial dopaminergic nigrostriatal damage with no gross motor impairments. Anhedonic-like behaviors were observed in the splash and sucrose consumption tests only 7 days after 6-OHDA lesion. By contrast, defense behaviors relevant to depression evaluated in the forced swimming test and social withdrawal only emerged 21 days after 6-OHDA lesion when anhedonia was no longer present. These temporally dissociated behavioral alterations were coupled to temporal- and structure-dependent alterations in dopaminergic markers such as dopamine D1 and D2 receptors and dopamine transporter, leading to altered dopamine sensitivity in DLS and PFC circuits, evaluated electrophysiologically. These results provide the first demonstration of a dissociated involvement of the DLS and PFC in anhedonic-like and defense behaviors relevant to depression in 6-OHDA-lesioned rats, which was linked with temporal fluctuations in dopaminergic receptor density, leading to altered dopaminergic system sensitivity in these two brain structures. This sheds new light to the duality between depressive and anhedonic symptoms in PD.


Anhedonia , Behavior, Animal , Corpus Striatum/pathology , Depression/chemically induced , Depression/pathology , Prefrontal Cortex/pathology , Animals , Corpus Striatum/metabolism , Corpus Striatum/physiopathology , Depression/physiopathology , Dopamine Plasma Membrane Transport Proteins/metabolism , Dopaminergic Neurons/metabolism , Dopaminergic Neurons/pathology , Male , Motor Activity , Oxidopamine , Prefrontal Cortex/metabolism , Prefrontal Cortex/physiopathology , Rats, Wistar , Receptors, Dopamine/metabolism , Time Factors , Tyrosine 3-Monooxygenase/metabolism
12.
Front Cell Neurosci ; 9: 521, 2015.
Article En | MEDLINE | ID: mdl-26834566

Recent studies combining pharmacological, behavioral, electrophysiological and molecular approaches indicate that depression results from maladaptive neuroplastic processes occurring in defined frontolimbic circuits responsible for emotional processing such as the prefrontal cortex, hippocampus, amygdala and ventral striatum. However, the exact mechanisms controlling synaptic plasticity that are disrupted to trigger depressive conditions have not been elucidated. Since glial cells (astrocytes and microglia) tightly and dynamically interact with synapses, engaging a bi-directional communication critical for the processing of synaptic information, we now revisit the role of glial cells in the etiology of depression focusing on a dysfunction of the "quad-partite" synapse. This interest is supported by the observations that depressive-like conditions are associated with a decreased density and hypofunction of astrocytes and with an increased microglia "activation" in frontolimbic regions, which is expected to contribute for the synaptic dysfunction present in depression. Furthermore, the traditional culprits of depression (glucocorticoids, biogenic amines, brain-derived neurotrophic factor, BDNF) affect glia functioning, whereas antidepressant treatments (serotonin-selective reuptake inhibitors, SSRIs, electroshocks, deep brain stimulation) recover glia functioning. In this context of a quad-partite synapse, systems modulating glia-synapse bidirectional communication-such as the purinergic neuromodulation system operated by adenosine 5'-triphosphate (ATP) and adenosine-emerge as promising candidates to "re-normalize" synaptic function by combining direct synaptic effects with an ability to also control astrocyte and microglia function. This proposed triple action of purines to control aberrant synaptic function illustrates the rationale to consider the interference with glia dysfunction as a mechanism of action driving the design of future pharmacological tools to manage depression.

...