Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
Add more filters










Publication year range
1.
Sci Rep ; 13(1): 755, 2023 01 14.
Article in English | MEDLINE | ID: mdl-36641518

ABSTRACT

Mice with deletion of the FMR1 gene show episodic memory impairments and exhibit dendritic spines and synaptic plasticity defects prevalently identified in non-training conditions. Based on evidence that synaptic changes associated with normal or abnormal memory emerge when mice are cognitively challenged, here we examine whether, and how, fragile entorhinal and hippocampal synapses are remodeled when mice succeed or fail to learn. We trained Fmr1 knockout (KO) and wild-type C57BL/6J (WT) mice in the novel object recognition (NOR) paradigm with 1 h or 24 h training-to-test intervals and then assessed whether varying the time between the presentation of similar and different objects modulates NOR performance and plasticity along the entorhinal cortex-hippocampus axis. At the 1 h-interval, KO mice failed to discriminate the novel object, showed a collapse of spines in the lateral entorhinal cortex (LEC), and of long-term potentiation (LTP) in the lateral perforant path (LPP), but a normal increase in hippocampal spines. At the 24 h, they exhibited intact NOR performance, typical LEC and hippocampal spines, and exaggerated LPP-LTP. Our findings reveal that the inability of mice to detect object novelty primarily stands in their impediment to elaborate, and convey to the hippocampus, sensory/perceptive object representations.


Subject(s)
Hippocampus , Neuronal Plasticity , Animals , Mice , Mice, Knockout , Mice, Inbred C57BL , Hippocampus/metabolism , Neuronal Plasticity/genetics , Long-Term Potentiation/genetics , Synapses/metabolism , Fragile X Mental Retardation Protein/genetics
2.
Int J Mol Sci ; 23(7)2022 Mar 22.
Article in English | MEDLINE | ID: mdl-35408815

ABSTRACT

Niemann Pick type C disease (NPC) is a rare disorder characterized by lysosomal lipid accumulation that damages peripheral organs and the central nervous system. Currently, only miglustat is authorized for NPC treatment in Europe, and thus the identification of new therapies is necessary. The hypothesis addressed in this study is that increasing adenosine levels may represent a new therapeutic approach for NPC. In fact, a reduced level of adenosine has been shown in the brain of animal models of NPC; moreover, the compound T1-11, which is able to weakly stimulate A2A receptor and to increase adenosine levels by blocking the equilibrative nucleoside transporter ENT1, significantly ameliorated the pathological phenotype and extended the survival in a mouse model of the disease. To test our hypothesis, fibroblasts from NPC1 patients were treated with dipyridamole, a clinically-approved drug with inhibitory activity towards ENT1. Dipyridamole significantly reduced cholesterol accumulation in fibroblasts and rescued mitochondrial deficits; the mechanism elicited by dipyridamole relies on activation of the adenosine A2AR subtype subsequent to the increased levels of extracellular adenosine due to the inhibition of ENT1. In conclusion, our results provide the proof of concept that targeting adenosine tone could be beneficial in NPC.


Subject(s)
Niemann-Pick Disease, Type C , Adenosine/pharmacology , Animals , Dipyridamole/pharmacology , Dipyridamole/therapeutic use , Drug Repositioning , Humans , Mice , Niemann-Pick Disease, Type C/pathology , Proof of Concept Study
3.
Int J Mol Sci ; 22(16)2021 Aug 17.
Article in English | MEDLINE | ID: mdl-34445564

ABSTRACT

Niemann-Pick type C (NPC) disease is a wide-spectrum clinical condition classified as a neurovisceral disorder affecting mainly the liver and the brain. It is caused by mutations in one of two genes, NPC1 and NPC2, coding for proteins located in the lysosomes. NPC proteins are deputed to transport cholesterol within lysosomes or between late endosome/lysosome systems and other cellular compartments, such as the endoplasmic reticulum and plasma membrane. The first trait of NPC is the accumulation of unesterified cholesterol and other lipids, like sphingosine and glycosphingolipids, in the late endosomal and lysosomal compartments, which causes the blockade of autophagic flux and the impairment of mitochondrial functions. In the brain, the main consequences of NPC are cerebellar neurodegeneration, neuroinflammation, and myelin defects. This review will focus on myelin defects and the pivotal importance of cholesterol for myelination and will offer an overview of the molecular targets and the pharmacological strategies so far proposed, or an object of clinical trials for NPC. Finally, it will summarize recent data on a new and promising pharmacological perspective involving A2A adenosine receptor stimulation in genetic and pharmacological NPC dysmyelination models.


Subject(s)
Adenosine A2 Receptor Agonists/pharmacology , Cholesterol/metabolism , Myelin Sheath/pathology , Niemann-Pick Disease, Type C/pathology , Receptor, Adenosine A2A/metabolism , Animals , Humans , Myelin Sheath/drug effects , Niemann-Pick Disease, Type C/drug therapy , Niemann-Pick Disease, Type C/metabolism
4.
Front Pharmacol ; 12: 647742, 2021.
Article in English | MEDLINE | ID: mdl-33953681

ABSTRACT

The STriatal-Enriched protein tyrosine phosphatase STEP is a brain-specific tyrosine phosphatase that plays a pivotal role in the mechanisms of learning and memory, and it has been demonstrated to be involved in several neuropsychiatric diseases. Recently, we found a functional interaction between STEP and adenosine A2A receptor (A2AR), a subtype of the adenosine receptor family widely expressed in the central nervous system, where it regulates motor behavior and cognition, and plays a role in cell survival and neurodegeneration. Specifically, we demonstrated the involvement of STEP in A2AR-mediated cocaine effects in the striatum and, more recently, we found that in the rat striatum and hippocampus, as well as in a neuroblastoma cell line, the overexpression of the A2AR, or its stimulation, results in an increase in STEP activity. In the present article we will discuss the functional implication of this interaction, trying to examine the possible mechanisms involved in this relation between STEP and A2ARs.

5.
Transl Psychiatry ; 11(1): 112, 2021 02 05.
Article in English | MEDLINE | ID: mdl-33547274

ABSTRACT

In fragile X syndrome (FXS) the lack of the fragile X mental retardation protein (FMRP) leads to exacerbated signaling through the metabotropic glutamate receptors 5 (mGlu5Rs). The adenosine A2A receptors (A2ARs), modulators of neuronal damage, could play a role in FXS. A synaptic colocalization and a strong permissive interaction between A2A and mGlu5 receptors in the hippocampus have been previously reported, suggesting that blocking A2ARs might normalize the mGlu5R-mediated effects of FXS. To study the cross-talk between A2A and mGlu5 receptors in the absence of FMRP, we performed extracellular electrophysiology experiments in hippocampal slices of Fmr1 KO mouse. The depression of field excitatory postsynaptic potential (fEPSPs) slope induced by the mGlu5R agonist CHPG was completely blocked by the A2AR antagonist ZM241385 and strongly potentiated by the A2AR agonist CGS21680, suggesting that the functional synergistic coupling between the two receptors could be increased in FXS. To verify if chronic A2AR blockade could reverse the FXS phenotypes, we treated the Fmr1 KO mice with istradefylline, an A2AR antagonist. We found that hippocampal DHPG-induced long-term depression (LTD), which is abnormally increased in FXS mice, was restored to the WT level. Furthermore, istradefylline corrected aberrant dendritic spine density, specific behavioral alterations, and overactive mTOR, TrkB, and STEP signaling in Fmr1 KO mice. Finally, we identified A2AR mRNA as a target of FMRP. Our results show that the pharmacological blockade of A2ARs partially restores some of the phenotypes of Fmr1 KO mice, both by reducing mGlu5R functioning and by acting on other A2AR-related downstream targets.


Subject(s)
Fragile X Syndrome , Receptor, Adenosine A2A , Adenosine , Animals , Cognition , Disease Models, Animal , Fragile X Mental Retardation Protein/genetics , Fragile X Syndrome/drug therapy , Fragile X Syndrome/genetics , Hippocampus/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptor, Adenosine A2A/genetics
6.
Pharmacol Res ; 147: 104338, 2019 09.
Article in English | MEDLINE | ID: mdl-31276772

ABSTRACT

Adenosine A2A receptor (A2AR) is a G-protein coupled receptor that regulates several important functions in the central nervous system. Large amount of preclinical data suggests that the A2AR could represent a target for the development of new therapeutic strategies for different neuropsychiatric conditions. In this review we will recapitulate and discuss the most relevant studies on the role of A2ARs in neurodegenerative, neurodevelopmental and psychiatric diseases, which led to suggest a therapeutic use of A2AR agonists in certain diseases (Niemann-Pick disease, autism-spectrum disorders, schizophrenia) and A2AR antagonists in others (Alzheimer's disease, Parkinson's disease, attention-deficit hyperactivity disorder, fragile X syndrome, depression, anxiety). Moreover, we will try to analyze which are the main obstacles to the conduction of clinical trials with A2AR ligands for the treatment of neuropsychiatric disease.


Subject(s)
Mental Disorders/metabolism , Neurodegenerative Diseases/metabolism , Receptor, Adenosine A2A/metabolism , Animals , Humans , Mental Disorders/drug therapy , Neurodegenerative Diseases/drug therapy
7.
Sci Rep ; 9(1): 9782, 2019 07 05.
Article in English | MEDLINE | ID: mdl-31278313

ABSTRACT

Niemann Pick type C (NPC) disease is a rare neurovisceral disorder. Mutations in npc1 gene induce an intracellular accumulation of unesterified cholesterol in the endosomal/lysosomal system causing cell death. We recently showed that stimulation of adenosine A2A receptors (A2AR) restores cholesterol accumulation in late endosomes/lysosomes in human NPC fibroblasts and neural cell lines transiently transfected with NPC1 siRNA, suggesting that these receptors might be targeted to contrast the disease. Since NPC1 disease is characterized by dysmyelination and maturational arrest of oligodendrocyte progenitors (OPs), in this study, we investigated whether A2AR stimulation could promote oligodendrocyte differentiation and myelin formation, thus overcoming these important neurological abnormalities. We developed a NPC1 pharmacological model, in which primary cultures of OPs are exposed to a cholesterol transport inhibitor to induce a NPC1-like phenotype characterized by several typical features such as (i) cholesterol accumulation, (ii) altered mitochondrial morphology and membrane potential, (iii) defect of autophagy and (iv) maturation arrest. The A2AR agonist CGS21680 normalized all NPC1-like features. The ability of CGS21680 of rescuing OP from maturational arrest and promoting their differentiation to mature OL, suggests that A2AR stimulation might be exploited to correct dysmyelination in NPC1, further supporting their therapeutic potential in the disease.


Subject(s)
Niemann-Pick Disease, Type C/etiology , Niemann-Pick Disease, Type C/metabolism , Oligodendroglia/metabolism , Receptor, Adenosine A2A/metabolism , Animals , Autophagy , Cell Differentiation , Cholesterol/metabolism , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Fibroblasts/metabolism , Humans , Mitochondria/metabolism , Niemann-Pick Disease, Type C/pathology , Oligodendroglia/pathology
8.
J Neurochem ; 149(2): 211-230, 2019 04.
Article in English | MEDLINE | ID: mdl-30614535

ABSTRACT

Cerebral ischemia is the second most common cause of death and a major cause of disability worldwide. Available therapies are based only on anticoagulants or recombinant tissue plasminogen activator. Extracellular adenosine increases during ischemia and acts as a neuroprotective endogenous agent mainly by activating adenosine A1 receptors (A1 Rs) which control calcium influx, glutamate release, membrane potential, and metabolism. Accordingly, in many experimental paradigms it has been already demonstrated that the stimulation of A1 R with full agonists is able to reduce ischemia-related structural and functional brain damage; unfortunately, cardiovascular side effects and desensitization of A1 R induced by these compounds have strongly limited their exploitation in stroke therapy so far. Among the newly emerging compounds, A1 R partial agonists could be almost free of side effects and equally effective. Therefore, we decided to evaluate the neuroprotective potential of two A1 R partial agonists, namely 2'-dCCPA and 3'-dCCPA, in in vitro and ex vivo experimental models of cerebral ischemia. Within the experimental paradigm of oxygen-glucose deprivation in vitro in human neuroblastoma (SH-SY5Y) cells both A1 R partial agonists increased cell viability. Considering the high level of expression of A1 Rs in the hippocampus and the susceptibility of CA1 region to hypoxia, we performed electrophysiological experiments in this subfield. The application of 7 min of oxygen-glucose deprivation constantly produces an irreversible synaptic failure in all the C57Bl/6 mice hippocampal slices evaluated; both tested compounds allowed a significant recovery of synaptic transmission. These findings demonstrate that A1 R and its partial agonists are still of interest for cerebral ischemia therapy. OPEN SCIENCE BADGES: This article has received a badge for *Open Materials* because it provided all relevant information to reproduce the study in the manuscript. The complete Open Science Disclosure form for this article can be found at the end of the article. More information about the Open Practices badges can be found at https://cos.io/our-services/open-science-badges/.


Subject(s)
Adenosine A1 Receptor Agonists/pharmacology , Brain Ischemia , Neuroprotective Agents/pharmacology , Animals , Hippocampus/drug effects , Humans , Mice , Mice, Inbred C57BL , Models, Theoretical , Receptor, Adenosine A1/drug effects , Synaptic Transmission/drug effects
9.
Purinergic Signal ; 14(3): 235-243, 2018 09.
Article in English | MEDLINE | ID: mdl-29770921

ABSTRACT

The A2A adenosine receptor (A2AR) is widely distributed on different cellular types in the brain, where it exerts a broad spectrum of pathophysiological functions, and for which a role in different neurodegenerative diseases has been hypothesized or demonstrated. To investigate the role of neuronal A2ARs in neurodegeneration, we evaluated in vitro and in vivo the effect of the neurotoxin 3-nitropropionic acid (3-NP) in a transgenic rat strain overexpressing A2ARs under the control of the neural-specific enolase promoter (NSEA2A rats). We recorded extracellular field potentials (FP) in corticostriatal slice and found that the synaptotoxic effect of 3-NP was significantly reduced in NSEA2A rats compared with wild-type animals (WT). In addition, after exposing corticostriatal slices to 3-NP 10 mM for 2 h, we found that striatal cell viability was significantly higher in NSEA2A rats compared to control rats. These in vitro results were confirmed by in vivo experiments: daily treatment of female rats with 3-NP 10 mg/kg for 8 days induced a selective bilateral lesion in the striatum, which was significantly reduced in NSEA2A compared to WT rats. These results demonstrate that the overexpression of the A2AR selectively at the neuronal level reduced 3-NP-induced neurodegeneration, and suggest an important function of the neuronal A2AR in the modulation of neurodegeneration.


Subject(s)
Corpus Striatum/metabolism , Huntington Disease/metabolism , Nerve Degeneration/metabolism , Receptor, Adenosine A2A/metabolism , Animals , Convulsants/toxicity , Corpus Striatum/drug effects , Disease Models, Animal , Humans , Male , Nitro Compounds/toxicity , Propionates/toxicity , Rats , Rats, Sprague-Dawley , Rats, Transgenic
10.
Neurobiol Dis ; 110: 1-11, 2018 02.
Article in English | MEDLINE | ID: mdl-29079454

ABSTRACT

Niemann-Pick C is a fatal neurovisceral disorder caused, in 95% of cases, by mutation of NPC1 gene. Therapeutic options are extremely limited and new "druggable" targets are highly warranted. We previously demonstrated that the stimulation of the adenosine A2A receptor (A2AR) normalized the pathological phenotype of cellular models of NPC1. Since the validation of A2ARs as a therapeutic target for NPC1 can be obtained only conducting studies in in vivo models of the disease, in the present paper, the effects of two agonists of A2ARs were evaluated in the mouse model Balb/c Npc1nih, hereafter indicated as NPC1-/-. The agonists CGS21680 (2.5 and 5mg/kg/day by intraperitoneal injection) and T1-11 (50mg/kg/day in drinking water) were administered at a presymptomatic stage of the disease of NPC1-/- mice (PN28 and PN30, respectively); the experimental groups were the following: vehicle-treated WT mice (N=16 for both CGS and T1-11 treatments); vehicle-treated NPC1-/- mice (N=14 for CGS and 12 for T1-11 treatment); CGS-treated NPC1-/- mice (N=7) and T1-11-treated NPC1-/- mice (N=11). The efficacy of the treatments was evaluated by comparing vehicle-treated and CGS or T1-11-treated NPC1-/- mice for their motor deficits (analyzed by both rotarod and footprint tests), hippocampal cognitive impairment (by Novel Object Recognition (NOR) test), cerebellar neurodegeneration (Purkinje neurons counting), and cholesterol and sphingomyelin accumulation in spleen and liver. Finally, the effect of both agonists on survival was evaluated by applying a humane late endpoint (weight loss >30% of peak weight, punched posture and reduced activity in the cage). The results demonstrated that, while CGS21680 only slightly attenuated cognitive deficits, T1-11 ameliorated motor coordination, significantly improved cognitive impairments, increased the survival of Purkinje neurons and reduced sphingomyelin accumulation in the liver. More importantly, it significantly prolonged the lifespan of NPC1-/- mice. In vitro experiments conducted in a neuronal model of NPC1 demonstrated that the ability of T1-11 to normalize cell phenotype was mediated by the selective activation of A2ARs and modulation of intracellular calcium levels. In conclusion, our results fully confirm the validity of A2ARs as a new target for NPC1 treatment. As soon as new ligands with improved pharmacokinetic characteristics (i.e. orally active, with brain bioavailability and metabolic stability) will be obtained, A2AR agonists could represent a breakthrough in the treatment of NPC.


Subject(s)
Adenosine/analogs & derivatives , Longevity/drug effects , Niemann-Pick Disease, Type C/pathology , Adenosine/pharmacology , Animals , Cerebellum/drug effects , Cerebellum/pathology , Disease Models, Animal , Liver/drug effects , Liver/pathology , Mice , Mice, Inbred BALB C , Mice, Knockout , Purinergic P1 Receptor Agonists/pharmacology , Purkinje Cells/drug effects , Receptor, Adenosine A2A/metabolism
11.
J Neurochem ; 136(5): 907-17, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26526685

ABSTRACT

Adenosine A2A receptors (A2 A Rs) and cannabinoid CB1 receptors (CB1 Rs) are highly expressed in the striatum, where they functionally interact and form A2A /CB1 heteroreceptor complexes. We investigated the effects of CB1 R stimulation in a transgenic rat strain over-expressing A2 A Rs under the control of the neural-specific enolase promoter (NSEA2A rats) and in age-matched wild-type (WT) animals. The effects of the CB1 R agonist WIN 55,212-2 (WIN) were significantly lower in NSEA2A rats than in WT animals, as demonstrated by i) electrophysiological recordings of synaptic transmission in corticostriatal slices; ii) the measurement of glutamate outflow from striatal synaptosomes and iii) in vivo experiments on locomotor activity. Moreover, while the effects of WIN were modulated by both A2 A R agonist (CGS 21680) and antagonists (ZM 241385, KW-6002 and SCH-442416) in WT animals, the A2 A R antagonists failed to influence WIN-mediated effects in NSEA2A rats. The present results demonstrate that in rats with genetic neuronal over-expression of A2 A Rs, the effects mediated by CB1 R activation in the striatum are significantly reduced, suggesting a change in the stoichiometry of A2A and CB1 receptors and providing a strategy to dissect the involvement of A2 A R forming or not forming heteromers in the modulation of striatal functions. These findings add additional evidence for the existence of an interaction between striatal A2 A Rs and CB1 Rs, playing a fundamental role in the regulation of striatal functions. We studied A2A -CB1 receptor interaction in transgenic rats over-expressing adenosine A2A receptors under the control of the neuron-specific enolase promoter (NSEA2A ). In these rats, we demonstrated a reduced effect of the CB1 receptor agonist WIN 55,212-2 in the modulation of corticostriatal synaptic transmission and locomotor activity, while CB1 receptor expression level did not change with respect to WT rats. A reduction in the expression of A2A -CB1 receptor heteromers is postulated.


Subject(s)
Adenosine/metabolism , Cannabinoids/metabolism , Corpus Striatum/metabolism , Receptor, Adenosine A2A/metabolism , Adenosine A2 Receptor Antagonists/pharmacology , Animals , Corpus Striatum/drug effects , Male , Rats, Sprague-Dawley , Receptor, Cannabinoid, CB1/metabolism , Synaptic Transmission/drug effects
12.
Eur Neuropsychopharmacol ; 25(6): 889-901, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25890884

ABSTRACT

Rho GTPases are molecules critically involved in neuronal plasticity and cognition. We have previously reported that modulation of brain Rho GTPases by the bacterial toxin CNF1 rescues the neurobehavioral phenotype in MeCP2-308 male mice, a model of Rett syndrome (RTT). RTT is a rare X-linked neurodevelopmental disorder and a genetic cause of intellectual disability, for which no effective therapy is available. Mitochondrial dysfunction has been proposed to be involved in the mechanism of the disease pathogenesis. Here we demonstrate that modulation of Rho GTPases by CNF1 rescues the reduced mitochondrial ATP production via oxidative phosphorylation in the brain of MeCP2-308 heterozygous female mice, the condition which more closely recapitulates that of RTT patients. In RTT mouse brain, CNF1 also restores the alterations in the activity of the mitochondrial respiratory chain (MRC) complexes and of ATP synthase, the molecular machinery responsible for the majority of cell energy production. Such effects were achieved through the upregulation of the protein content of those MRC complexes subunits, which were defective in RTT mouse brain. Restored mitochondrial functionality was accompanied by the rescue of deficits in cognitive function (spatial reference memory in the Barnes maze), synaptic plasticity (long-term potentiation) and Tyr1472 phosphorylation of GluN2B, which was abnormally enhanced in the hippocampus of RTT mice. Present findings bring into light previously unknown functional mitochondrial alterations in the brain of female mice modeling RTT and provide the first evidence that RTT brain mitochondrial dysfunction can be rescued by modulation of Rho GTPases.


Subject(s)
Cognition Disorders/etiology , Cognition Disorders/metabolism , Mitochondrial Diseases/etiology , Mitochondrial Diseases/metabolism , Neuronal Plasticity/genetics , Rett Syndrome/complications , rho GTP-Binding Proteins/metabolism , Animals , Bacterial Toxins/therapeutic use , Brain/drug effects , Brain/metabolism , Cognition Disorders/drug therapy , Disease Models, Animal , Electron Transport Chain Complex Proteins/metabolism , Escherichia coli Proteins/therapeutic use , Female , Hippocampus/cytology , Hippocampus/drug effects , Hippocampus/physiology , Injections, Intraventricular , Maze Learning/drug effects , Maze Learning/physiology , Methyl-CpG-Binding Protein 2/genetics , Mice , Mice, Transgenic , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondrial Diseases/drug therapy , Neuronal Plasticity/drug effects , Receptors, Glutamate/metabolism , Rett Syndrome/pathology , Rett Syndrome/physiopathology , Visual Acuity/drug effects , Visual Acuity/genetics
13.
Neurobiol Dis ; 71: 193-204, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25132555

ABSTRACT

Adenosine A1 receptor (A1R) stimulation exerts beneficial effects in response to various insults to the brain and, although it was found neuroprotective in a lesional model of Huntington's disease (HD), the features of this receptor in genetic models of HD have never been explored. In the present study we characterized the expression, affinity and functional effects of A1Rs in R6/2 mice (the most widely used transgenic model of HD) and in a cellular model of HD. Binding studies revealed that the density of A1Rs was significantly reduced in the cortex and the striatum of R6/2 mice compared to age-matched wild-type (WT), while receptor affinity was unchanged. The selective A1R agonist cyclopentyladenosine (CPA, 300nM) was significantly more effective in reducing synaptic transmission in corticostriatal slices from symptomatic R6/2 than in age-matched WT mice. Such an effect was due to a stronger inhibition of glutamate release from the pre-synaptic terminal. The different functional activities of A1Rs in HD mice were associated also to a different intracellular signaling pathway involved in the synaptic effect of CPA. In fact, while the PKA pathway was involved in both genotypes, p38 MAPK inhibitor SB203580 partially prevented synaptic effects of CPA in R6/2, but not in WT, mice; moreover, CPA differently modulated the phosphorylation status of p38 in the two genotypes. In vitro studies confirmed a different behavior of A1Rs in HD: CPA (100 nM for 5h) modulated cell viability in STHdh(Q111/Q111) (mhttHD cells), without affecting the viability of STHdh(Q7/Q7) (wthtt cells). This effect was prevented by the application of SB203580. Our results demonstrate that in the presence of the HD mutation A1Rs undergo profound changes in terms of expression, pharmacology and functional activity. These changes have to be taken in due account when considering A1Rs as a potential therapeutic target for this disease.


Subject(s)
Adenine/analogs & derivatives , Cyclopentanes/pharmacology , Gene Expression Regulation/genetics , Huntington Disease/metabolism , Receptor, Adenosine A1/metabolism , Action Potentials/drug effects , Action Potentials/genetics , Adenine/pharmacology , Adenosine A1 Receptor Antagonists/pharmacokinetics , Animals , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Cyclic AMP/metabolism , Disease Models, Animal , Dose-Response Relationship, Drug , Electric Stimulation , Enzyme Inhibitors/pharmacology , Female , Gene Expression Regulation/drug effects , Glutamic Acid/metabolism , Huntingtin Protein , Huntington Disease/genetics , Huntington Disease/pathology , In Vitro Techniques , Male , Mice , Mice, Transgenic , Nerve Tissue Proteins/genetics , Neurons/drug effects , Neurons/metabolism , Nuclear Proteins/genetics , Potassium Chloride/pharmacology , Protein Binding/drug effects , Signal Transduction/drug effects , Signal Transduction/genetics , Statistics, Nonparametric , Synaptic Transmission/drug effects , Synaptic Transmission/genetics , Synaptosomes/drug effects , Synaptosomes/metabolism , Transfection , Trinucleotide Repeat Expansion/genetics , Tritium/pharmacokinetics , Xanthines/pharmacokinetics
14.
Neuropsychopharmacology ; 39(3): 569-78, 2014 Feb.
Article in English | MEDLINE | ID: mdl-23989619

ABSTRACT

The striatum is a brain area implicated in the pharmacological action of drugs of abuse. Adenosine A2A receptors (A2ARs) are highly expressed in the striatum and mediate, at least in part, cocaine-induced psychomotor effects in vivo. Here we studied the synaptic mechanisms implicated in the pharmacological action of cocaine in the striatum and investigated the influence of A2ARs. We found that synaptic transmission was depressed in corticostriatal slices after perfusion with cocaine (10 µM). This effect was reduced by the A2AR antagonist ZM241385 and almost abolished in striatal A2AR-knockout mice (mice lacking A2ARs in striatal neurons, stA2ARKO). The effect of cocaine on synaptic transmission was also prevented by the protein tyrosine phosphatases (PTPs) inhibitor sodium orthovanadate (Na3VO4). In synaptosomes prepared from striatal slices, we found that the activity of striatal-enriched protein tyrosine phosphatase (STEP) was upregulated by cocaine, prevented by ZM241385, and absent in synaptosomes from stA2ARKO. The role played by STEP in cocaine modulation of synaptic transmission was investigated in whole-cell voltage clamp recordings from medium spiny neurons of the striatum. We found that TAT-STEP, a peptide that renders STEP enzymatically inactive, prevented cocaine-induced reduction in AMPA- and NMDA-mediated excitatory post-synaptic currents, whereas the control peptide, TAT-myc, had no effect. These results demonstrate that striatal A2ARs modulate cocaine-induced synaptic depression in the striatum and highlight the potential role of PTPs and specifically STEP in the effects of cocaine.


Subject(s)
Cocaine/pharmacology , Corpus Striatum/drug effects , Dopamine Uptake Inhibitors/pharmacology , Protein Tyrosine Phosphatases, Non-Receptor/metabolism , Receptor, Adenosine A2A/metabolism , Synaptic Transmission/drug effects , Animals , Cerebral Cortex/cytology , Corpus Striatum/cytology , Enzyme Inhibitors/pharmacology , Gene Expression Regulation/drug effects , Humans , In Vitro Techniques , Inhibitory Postsynaptic Potentials/drug effects , Inhibitory Postsynaptic Potentials/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neural Pathways/physiology , Neurons/drug effects , Neurons/ultrastructure , Receptor, Adenosine A2A/genetics , Synaptosomes/drug effects , Synaptosomes/metabolism , Vanadates/pharmacology
15.
J Neurosci ; 33(39): 15388-93, 2013 Sep 25.
Article in English | MEDLINE | ID: mdl-24068806

ABSTRACT

Niemann-Pick type C1 (NPC1) disease is a rare neurovisceral disorder characterized by intracellular accumulation of unesterified cholesterol, sphingolipids, and other lipids in the lysosomal compartment. A deregulation of lysosomal calcium has been identified as one of the earliest steps of the degenerative process. Since adenosine A2A receptors (A2ARs) control lysosome trafficking and pH, which closely regulates lysosomal calcium, we hypothesized a role for these receptors in NPC1. The aim of this study was to evaluate the effects of the A2AR agonist CGS21680 on human control and NPC1 fibroblasts. We show that CGS21680 raises lysosomal calcium levels and rescues mitochondrial functionality (mitochondrial inner membrane potential and expression of the complex IV of the mitochondrial respiratory chain), which is compromised in NPC1 cells. These effects are prevented by the selective blockade of A2ARs by the antagonist ZM241385. The effects of A2AR activation on lysosomal calcium are not mediated by the cAMP/PKA pathway but they appear to involve the phosphorylation of ERK1/2. Finally, CGS21680 reduces cholesterol accumulation (Filipin III staining), which is the main criterion currently used for identification of a compound or pathway that would be beneficial for NPC disease, and such an effect is prevented by the Ca(2+) chelator BAPTA-AM. Our findings strongly support the hypothesis that A2AR agonists may represent a therapeutic option for NPC1 and provide insights on their mechanisms of action.


Subject(s)
Adenosine A2 Receptor Agonists/pharmacology , Adenosine/analogs & derivatives , Fibroblasts/drug effects , Niemann-Pick Disease, Type C/metabolism , Phenethylamines/pharmacology , Phenotype , Receptor, Adenosine A2A/metabolism , Adenosine/pharmacology , Adenosine A2 Receptor Antagonists/pharmacology , Calcium/metabolism , Case-Control Studies , Cell Line , Cholesterol/metabolism , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Electron Transport Complex IV/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Fibroblasts/metabolism , Fibroblasts/pathology , Humans , Lysosomes/metabolism , Male , Membrane Potential, Mitochondrial , Mitochondria/metabolism , Niemann-Pick Disease, Type C/pathology , Triazines/pharmacology , Triazoles/pharmacology
16.
J Neurochem ; 125(2): 225-35, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23363456

ABSTRACT

NMDA receptor-mediated excitotoxicity is thought to play a pivotal role in the pathogenesis of Huntington's disease (HD). The neurotrophin brain-derived neurotrophic factor (BDNF), which is also highly involved in HD and whose effects are modulated by adenosine A2 ARs, influences the activity and expression of striatal NMDA receptors. In electrophysiology experiments, we investigated the role of BDNF toward NMDA-induced effects in HD models, and the possible involvement of A2ARs. In corticostriatal slices from wild-type mice and age-matched symptomatic R6/2 mice (a model of HD), NMDA application (75 µM) induced a transient or a permanent (i.e., toxic) reduction of field potential amplitude, respectively. BDNF (10 ng/mL) potentiated NMDA effects in wild-type, while it protected from NMDA toxicity in R6/2 mice. Both effects of BDNF were prevented by A2 AR blockade. The protective effect of BDNF against NMDA-induced toxicity was reproduced in a cellular model of HD. These findings may have very important implications for the neuroprotective potential of BDNF and A2 AR ligands in HD.


Subject(s)
Brain-Derived Neurotrophic Factor/metabolism , Huntington Disease/metabolism , N-Methylaspartate/toxicity , Receptor, Adenosine A2A/metabolism , Synaptic Transmission/physiology , Animals , Disease Models, Animal , Female , Genotype , Huntington Disease/genetics , Male , Mice , Mice, Inbred C57BL , Patch-Clamp Techniques
17.
J Neurosci Res ; 91(4): 585-92, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23361938

ABSTRACT

Caffeine is a nonselective adenosine receptor antagonist; chronic consumption has proved protective toward neurodegenerative diseases such as Parkinson's and Alzheimer's diseases. The present study was designed to determine whether caffeine intake affected survival and/or motor performance in a transgenic model of amyotrophic lateral sclerosis (ALS). SOD1(G93A) mice received caffeine through drinking water from 70 days of age until death. Body weight, motor performance and survival were evaluated. Furthermore, the expression of adenosine A(2A) receptors (A(2A) Rs), glial glutamate transporter (GLT1), and glial fibrillar acidic protein (GFAP) were evaluated by Western blotting. The results showed that caffeine intake significantly shortened the survival of SOD1(G93A) mice (log rank test, P = 0.01) and induced a nonsignificant advancing of disease onset. The expression of A(2A) R, GLT1, and GFAP was altered in the spinal cords of ALS mice, but caffeine did not influence their expression in either wild-type or SOD1(G93) mice. These data indicate that adenosine receptors may play an important role in ALS.


Subject(s)
Amyotrophic Lateral Sclerosis/metabolism , Caffeine/administration & dosage , Longevity/drug effects , Motor Neurons/drug effects , Motor Skills/drug effects , Spinal Cord/drug effects , Administration, Oral , Amyotrophic Lateral Sclerosis/genetics , Animals , Body Weight/drug effects , Body Weight/physiology , Disease Models, Animal , Mice , Mice, Transgenic , Motor Neurons/metabolism , Receptor, Adenosine A2A/metabolism , Rotarod Performance Test , Spinal Cord/metabolism , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism , Superoxide Dismutase-1
18.
Neurobiol Dis ; 45(3): 983-91, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22207189

ABSTRACT

Cannabinoid CB1 receptors (CB1Rs) are known to be downregulated in patients and in animal models of Huntington's disease (HD). However, the functional meaning of this reduction, if any, is still unclear. Here, the effects of the cannabinoid receptor agonist WIN 55,212-2 (WIN) were investigated on striatal synaptic transmission and on glutamate and GABA release in symptomatic R6/2 mice, a genetic model of HD. The expression levels of CB1Rs in glutamatergic and GABAergic synapses were also evaluated. We found that in R6/2 mice, WIN effects on synaptic transmission and glutamate release were significantly increased with respect to wild type mice. On the contrary, a decrease in WIN-induced reduction of GABA release was found in R6/2 versus WT mice. The expression of CB1Rs in GABAergic neurons was drastically reduced, while CB1Rs levels in glutamatergic neurons were unchanged. These results demonstrate that the expression and functionality of CB1Rs are differentially affected in GABAergic and glutamatergic neurons in R6/2 mice. As a result, the balance between CB1Rs expressed by the two neuronal populations and, thus, the net effect of CB1R stimulation, is profoundly altered in HD mice.


Subject(s)
Glutamates/metabolism , Huntington Disease/pathology , Neurons/metabolism , Receptor, Cannabinoid, CB1/metabolism , gamma-Aminobutyric Acid/metabolism , Action Potentials/drug effects , Action Potentials/genetics , Action Potentials/physiology , Analysis of Variance , Animals , Benzoxazines/pharmacology , Brain/pathology , Disease Models, Animal , Dronabinol/analogs & derivatives , Dronabinol/pharmacology , Drug Interactions , Electric Stimulation , Excitatory Amino Acid Agents/pharmacology , Humans , Huntingtin Protein , Huntington Disease/physiopathology , In Vitro Techniques , Mice , Mice, Transgenic , Microscopy, Immunoelectron , Morpholines/pharmacology , Motor Activity/drug effects , Motor Activity/genetics , Naphthalenes/pharmacology , Nerve Tissue Proteins/genetics , Neurons/drug effects , Neurons/ultrastructure , Nuclear Proteins/genetics , Patch-Clamp Techniques , Piperidines/pharmacology , Potassium/pharmacology , Pyrazoles/pharmacology , Receptor, Cannabinoid, CB1/agonists , Receptor, Cannabinoid, CB1/antagonists & inhibitors , Statistics, Nonparametric , Synaptosomes/drug effects , Synaptosomes/metabolism , Trinucleotide Repeats/genetics , Tritium/metabolism , Vesicular Glutamate Transport Protein 1/metabolism , Vesicular Inhibitory Amino Acid Transport Proteins/metabolism
19.
Mol Nutr Food Res ; 55(4): 541-52, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21462321

ABSTRACT

SCOPE: We hypothesized that chronic supplementation with branched chain amino acids (BCAAs) affects neurobehavioral development in vulnerable gene backgrounds. METHODS AND RESULTS: A murine model of amyotrophic lateral sclerosis (ALS), G93A mice bearing the mutated human superoxide dismutase 1 (SOD1) gene, and control mice received from 4 to 16 wk of age dietary supplementation with BCAAs at doses comparable to human usage. Motor coordination, exploratory behaviors, pain threshold, synaptic activity and response to glutamatergic stimulation in primary motor cortex slices were evaluated between the 8th and 16th week. The glial glutamate transporter 1 (GLT-1) and metabotropic glutamate 5 receptor (mGlu5R) were analyzed by immunoblotting in cortex, hippocampus and striatum. BCAAs induced hyperactivity, decreased pain threshold in wild-type mice and exacerbated the motor deficits of G93A mice while counteracting their abnormal pain response. Electrophysiology on G93A brain slices showed impaired synaptic function, reduced toxicity of GLT-1 blocking and increased glutamate toxicity prevented by BCAAs. Immunoblotting indicated down-regulation of GLT-1 and mGlu5R in G93A, both effects counteracted by BCAAs. CONCLUSION: These results, though not fully confirming a role of BCAAs in ALS-like etiology in the genetic model, clearly indicate that BCAAs' complex effects on central nervous system depend on gene background and raise alert over their spread use.


Subject(s)
Amino Acids, Branched-Chain/adverse effects , Amyotrophic Lateral Sclerosis/physiopathology , Diet/adverse effects , Hyperkinesis/etiology , Synaptic Transmission , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/metabolism , Animals , Behavior, Animal , Brain/drug effects , Brain/metabolism , Dietary Supplements/adverse effects , Glucose Transporter Type 1/antagonists & inhibitors , Glucose Transporter Type 1/metabolism , Glutamic Acid/toxicity , In Vitro Techniques , Male , Mice , Mice, Transgenic , Nerve Tissue Proteins/metabolism , Neurons/drug effects , Neurons/metabolism , Organ Specificity , Pain Threshold , Receptor, Metabotropic Glutamate 5 , Receptors, Metabotropic Glutamate/metabolism , Severity of Illness Index , Synaptic Transmission/drug effects , Time Factors
20.
ScientificWorldJournal ; 10: 1768-82, 2010 Sep 01.
Article in English | MEDLINE | ID: mdl-20842321

ABSTRACT

In the last few years, accumulating evidence has shown the existence of an important cross-talk between adenosine A(2A) receptors (A(2A)Rs) and brain-derived neurotrophic factor (BDNF). Not only are A(2A)Rs involved in the mechanism of transactivation of BDNF receptor TrkB, they also modulate the effect of BDNF on synaptic transmission, playing a facilitatory and permissive role. The cAMP-PKA pathway, the main transduction system operated by A(2A)Rs, is involved in such effects. Furthermore, a basal tonus of A(2A)Rs is required to allow the regulation of BDNF physiological levels in the brain, as demonstrated by the reduced protein levels measured in A(2A)Rs KO mice. The crucial role of adenosine A(2A)Rs in the maintenance of synaptic functions and BDNF levels will be reviewed here and discussed in the light of possible implications for Huntington's disease therapy, in which a joint impairment of BDNF and A(2A)Rs seems to play a pathogenetic role.


Subject(s)
Brain-Derived Neurotrophic Factor/metabolism , Brain/physiology , Receptor, Adenosine A2A/physiology , Synaptic Transmission/physiology , Animals , Brain/metabolism , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Humans , Huntington Disease/metabolism , Huntington Disease/physiopathology , Models, Neurological , Receptor, Adenosine A2A/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...