Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 76
Filter
1.
Neurosci Biobehav Rev ; 77: 32-47, 2017 06.
Article in English | MEDLINE | ID: mdl-28223129

ABSTRACT

Huntington's disease (HD) is an inherited neurodegenerative disorder, characterized by impairment in motor, cognitive and psychiatric domains. Currently, there is no specific therapy to act on the onset or progression of HD. The marked neuronal death observed in HD is a main argument in favour of stem cells (SCs) transplantation as a promising therapeutic perspective to replace the population of lost neurons and restore the functionality of the damaged circuitry. The availability of rodent models of HD encourages the investigation of the restorative potential of SCs transplantation longitudinally. However, the results of preclinical studies on SCs therapy in HD are so far largely inconsistent; this hampers the individuation of the more appropriate model and precludes the comparative analysis of transplant efficacy on behavioural end points. Thus, this review will describe the state of the art of in vivo research on SCs therapy in HD, analysing in a translational perspective the strengths and weaknesses of animal studies investigating the therapeutic potential of cell transplantation on HD progression.


Subject(s)
Huntington Disease , Animals , Disease Models, Animal , Humans , Neurons , Regenerative Medicine , Stem Cell Transplantation
2.
Neuropharmacology ; 103: 155-62, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26631535

ABSTRACT

Niemann Pick C 1 (NPC1) disease is an incurable, devastating lysosomal-lipid storage disorder characterized by hepatosplenomegaly, progressive neurological impairment and early death. Current treatments are very limited and the research of new therapeutic targets is thus mandatory. We recently showed that the stimulation of adenosine A2A receptors (A2ARs) rescues the abnormal phenotype of fibroblasts from NPC1 patients suggesting that A2AR agonists could represent a therapeutic option for this disease. However, since all NPC1 patients develop severe neurological symptoms which can be ascribed to the complex pathology occurring in both neurons and oligodendrocytes, in the present paper we tested the effects of the A2AR agonist CGS21680 in human neuronal and oligodendroglial NPC1 cell lines (i.e. neuroblastoma SH-SY5Y and oligodendroglial MO3.13 transiently transfected with NPC1 small interfering RNA). The down-regulation of the NPC1 protein effectively resulted in intracellular cholesterol accumulation and altered mitochondrial membrane potential. Both effects were significantly attenuated by CGS21680 (500 nM). The protective effects of CGS were prevented by the selective A2AR antagonist ZM241385 (500 nM). The involvement of calcium modulation was demonstrated by the ability of Bapta-AM (5-7 µM) in reverting the effect of CGS. The A2A-dependent activity was prevented by the PKA-inhibitor KT5720, thus showing the involvement of the cAMP/PKA signaling. These findings provide a clear in vitro proof of concept that A2AR agonists are promising potential drugs for NPC disease.


Subject(s)
Adenosine A2 Receptor Agonists/pharmacology , Cholesterol/metabolism , Mitochondria/metabolism , Neurons/metabolism , Niemann-Pick Disease, Type C/metabolism , Receptor, Adenosine A2A/metabolism , Adenosine/analogs & derivatives , Adenosine/pharmacology , Calcium/metabolism , Carrier Proteins/genetics , Cell Line, Tumor , Cell Membrane/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Humans , Intracellular Signaling Peptides and Proteins , Membrane Glycoproteins/genetics , Niemann-Pick C1 Protein , Oligodendroglia/metabolism , Phenethylamines/pharmacology
3.
J Neurosci Res ; 89(8): 1276-83, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21538464

ABSTRACT

Branched-chain amino acids (BCAAs), valine, isoleucine, and leucine, are widely used among athletes as dietary integrators. Although the occurrence of untoward effects of BCCA supplementation, with particular regard to neurological disturbances, cannot be excluded, no specific studies have been performed so far. The aim of this work was to evaluate the effects of a diet enriched in BCAAs on the expression of oxidative stress pathway genes in the brain of C57Bl/6J mice. Animals were fed a standard or a BCAA diet for 95 days starting from postnatal day 21 until sacrifice. BCAA treatment, at doses comparable to human usage, significantly down-regulated the expression of some antioxidant genes, while up-regulating the expression of some oxygen transporters. In conclusion, it appears that BCAAs administered by diet could alter some specific oxidative stress pathways in the brain. Caution should thus be exercised in the widespread use of BCAAs as dietary integrators in sports practice.


Subject(s)
Amino Acids, Branched-Chain/administration & dosage , Brain/drug effects , Diet , Oxidative Stress/drug effects , Amyotrophic Lateral Sclerosis/etiology , Animals , Apolipoproteins E/genetics , Apolipoproteins E/metabolism , Brain/metabolism , Down-Regulation/drug effects , Down-Regulation/genetics , Globins/genetics , Globins/metabolism , Male , Mice , Mice, Inbred C57BL , Oxidative Stress/genetics , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism , Superoxide Dismutase-1 , Up-Regulation/genetics
4.
Neurol Sci ; 31 Suppl 3: 295-7, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20644975

ABSTRACT

At the end of 2006, a pharmacovigilance program on natalizumab was settled by the Italian Pharmaceutical Agency, and on January 2007, multiple sclerosis patients poorly responding to the immunomodulating therapies or with an aggressive clinical form of disease from onset initiated to be registered and to receive the medication. On February 2010, almost 3,000 cases have been treated with natalizumab. The drop-out rate is 10%. Almost 800 cases received cycles of natalizumab for more than 18 months. One case of PML was reported and other adverse events are similar to those described in phase III studies. The majority of cases remained stable, while in 25% of cases, an improvement of disability was documented.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Multiple Sclerosis/drug therapy , Product Surveillance, Postmarketing/trends , Registries , Adult , Antibodies, Monoclonal, Humanized/adverse effects , Female , Humans , Italy/epidemiology , Male , Multiple Sclerosis/epidemiology , Natalizumab , Registries/statistics & numerical data
5.
Neurol Sci ; 30 Suppl 2: S163-5, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19882367

ABSTRACT

At the end of 2006 a country-based surveillance program on natalizumab therapy in multiple sclerosis was settled in Italy by a collaborative effort of the Italian Drug Agency (AIFA) and a group of experts and neurologists appointed by the National Society of Neurology (SIN). After 2 years, 1,818 patients are registered in the database. The majority of cases (88.6%) failed the therapy with beta interferon or glatiramer acetate and had relapses or accumulated disability during immunomodulating treatment, while 11.4% of patients enrolled in the surveillance study were not previously treated with immunomodulating therapies and had a rapidly evolving clinical course. Almost 10% of the patients treated with natalizumab interrupted, for various different reasons, the therapy. Treatment was well tolerated and side effects were similar to those reported in the registrative studies. The majority of treated cases are stable or ameliorated.


Subject(s)
Antibodies, Monoclonal/adverse effects , Multiple Sclerosis/drug therapy , Product Surveillance, Postmarketing , Adult , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal, Humanized , Databases, Factual , Female , Follow-Up Studies , Humans , Italy , Magnetic Resonance Imaging , Male , Natalizumab , Patient Dropouts
6.
Curr Pharm Des ; 14(15): 1468-74, 2008.
Article in English | MEDLINE | ID: mdl-18537670

ABSTRACT

Adenosine A(2A)-dopamine D(2) receptor interactions play a very important role in striatal function. A(2A)-D(2) receptor interactions provide an example of the capabilities of information processing by just two different G protein-coupled receptors. Thus, there is evidence for the coexistence of two reciprocal antagonistic interactions between A(2A) and D(2) receptors in the same neurons, the GABAergic enkephalinergic neurons. An antagonistic A(2A)-D(2) intramembrane receptor interaction, which depends on A(2A)-D(2) receptor heteromerization and G(q/11)-PLC signaling, modulates neuronal excitability and neurotransmitter release. On the other hand, an antagonistic A(2A)-D(2) receptor interaction at the adenylyl-cyclase level, which depends on G(s/olf)- and G(i/o)-type V adenylyl-cyclase signaling, modulates protein phosphorylation and gene expression. Finally, under conditions of upregulation of an activator of G protein signaling (AGS3), such as during chronic treatment with addictive drugs, a synergistic A(2A)-D(2) receptor interaction can also be demonstrated. AGS3 facilitates a synergistic interaction between G(s/olf) - and G(i/o)-coupled receptors on the activation of types II/IV adenylyl cyclase, leading to a paradoxical increase in protein phosphorylation and gene expression upon co-activation of A(2A) and D(2) receptors. The analysis of A(2)-D(2) receptor interactions will have implications for the pathophysiology and treatment of basal ganglia disorders and drug addiction.


Subject(s)
Receptor, Adenosine A2A/physiology , Receptors, Dopamine D2/physiology , Adenosine A2 Receptor Agonists , Adenosine A2 Receptor Antagonists , Adenylyl Cyclases/metabolism , Animals , Basal Ganglia/physiology , Basal Ganglia Diseases/drug therapy , Basal Ganglia Diseases/physiopathology , Dopamine D2 Receptor Antagonists , Enkephalins/metabolism , Enzyme Activation , GTP-Binding Proteins/physiology , Humans , Neurons/metabolism , Phosphorylation , Receptors, Dopamine D2/agonists , Substance-Related Disorders/drug therapy , Substance-Related Disorders/physiopathology , gamma-Aminobutyric Acid/metabolism
7.
Eur J Nucl Med Mol Imaging ; 35(4): 704-15, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18080815

ABSTRACT

PURPOSE: Huntington's disease (HD) is a progressive neurodegenerative disorder, which is characterised by prominent neuronal cell loss in the basal ganglia with motor and cognitive disturbances. One of the most well-studied pharmacological models of HD is produced by local injection in the rat brain striatum of the excitotoxin quinolinic acid (QA), which produces many of the distinctive features of this human neurodegenerative disorder. Here, we report a detailed analysis, obtained both in vivo and in vitro of this pharmacological model of HD. MATERIALS AND METHODS: By combining emission tomography (PET) with autoradiographic and immunocytochemical confocal laser techniques, we quantified in the QA-injected striatum the temporal behavior (from 1 to 60 days from the excitotoxic insult) of neuronal cell density and receptor availability (adenosine A(2A) and dopamine D(2) receptors) together with the degree of microglia activation. RESULTS: Both approaches showed a loss of adenosine A(2A) and dopamine D(2) receptors paralleled by an increase of microglial activation. CONCLUSION: This combined longitudinal analysis of the disease progression, which suggested an impairment of neurotransmission, neuronal integrity and a reversible activation of brain inflammatory processes, might represent a more quantitative approach to compare the differential effects of treatments in slowing down or reversing HD in rodent models with potential applications to human patients.


Subject(s)
Corpus Striatum/physiology , Microglia/physiology , Nerve Degeneration/chemically induced , Raclopride/pharmacology , Animals , Carbon Radioisotopes , Corpus Striatum/drug effects , Isoquinolines/pharmacokinetics , Kinetics , Microglia/drug effects , Quinolinic Acid/toxicity , Raclopride/pharmacokinetics , Radioisotope Dilution Technique , Rats , Rats, Wistar , Receptors, Dopamine D2/drug effects , Receptors, Dopamine D2/physiology , Receptors, Purinergic P1/drug effects , Receptors, Purinergic P1/physiology , Reference Values , Stereotaxic Techniques
8.
J Neurochem ; 104(1): 279-86, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18005343

ABSTRACT

Brain-derived neurotrophic factor (BDNF), a member of neurotrophin family, enhances synaptic transmission and regulates neuronal proliferation and survival. Both BDNF and its tyrosine kinase receptors (TrkB) are highly expressed in the hippocampus, where an interaction with adenosine A(2A) receptors (A(2A)Rs) has been recently reported. In the present paper, we evaluated the role of A(2A)Rs in mediating functional effects of BDNF in hippocampus using A(2A)R knock-out (KO) mice. In hippocampal slices from WT mice, application of BDNF (10 ng/mL) increased the slope of excitatory post-synaptic field potentials (fEPSPs), an index of synaptic facilitation. This increase of fEPSP slope was abolished by the selective A(2A) antagonist ZM 241385. Similarly, genetic deletion of the A(2A)Rs abolished BDNF-induced increase of the fEPSP slope in slices from A(2A)R KO mice The reduced functional ability of BDNF in A(2A)R KO mice was correlated with the reduction in hippocampal BDNF levels. In agreement, the pharmacological blockade of A(2)Rs by systemic ZM 241385 significantly reduced BDNF levels in the hippocampus of normal mice. These results indicate that the tonic activation of A(2A)Rs is required for BDNF-induced potentiation of synaptic transmission and for sustaining a normal BDNF tone in the hippocampus.


Subject(s)
Brain-Derived Neurotrophic Factor/metabolism , Brain-Derived Neurotrophic Factor/pharmacology , Hippocampus/physiology , Receptor, Adenosine A2A/physiology , Synaptic Transmission/drug effects , Adenosine A2 Receptor Antagonists , Animals , Dose-Response Relationship, Drug , Drug Interactions , Enzyme-Linked Immunosorbent Assay/methods , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/physiology , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , In Vitro Techniques , Mice , Mice, Knockout , Patch-Clamp Techniques/methods , Receptor, Adenosine A2A/deficiency , Triazines/pharmacology , Triazoles/pharmacology
9.
Neurobiol Dis ; 28(2): 197-205, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17720507

ABSTRACT

The effect of chronic treatment with the selective adenosine A2A receptor antagonist SCH 58261 on the behavioral and electrophysiological alterations typical of R6/2 mice (a transgenic mouse model of Huntington's disease, HD), has been studied. Starting from 5 weeks of age, R6/2 and wild type (WT) mice were treated daily with SCH 58261 (0.01 mg/kg i.p.) for 7 days. In the following weeks, the ability of mice to perform in the rotarod, plus maze and open field tests were evaluated. In addition, with electrophysiological experiments in corticostriatal slices we tested whether the well-known increased NMDA vulnerability of R6/2 mice was prevented by SCH 58261 treatment. We found that chronic treatment with SCH 58262: i) fully prevented the alterations in emotional/anxious responses displayed by R6/2 mice; ii) did not prevent the impairment in motor coordination; iii) abolished the increase in NMDA-induced toxicity observed in the striatum of HD mice. On balance, targeting A2A receptors seems to have some beneficial effects in HD even though, given the complexity of A2A receptor pharmacology and HD pathogenesis, further studies are necessary to clarify whether A2A receptor antagonists have therapeutic potential in HD.


Subject(s)
Adenosine A2 Receptor Antagonists , Brain/drug effects , Huntington Disease/drug therapy , Neuroprotective Agents/pharmacology , Pyrimidines/pharmacology , Triazoles/pharmacology , Animals , Behavior, Animal/drug effects , Behavior, Animal/physiology , Brain/metabolism , Brain/physiopathology , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Corpus Striatum/physiopathology , Disease Models, Animal , Excitatory Amino Acid Agonists/toxicity , Glutamic Acid/metabolism , Huntington Disease/metabolism , Huntington Disease/physiopathology , Learning Disabilities/drug therapy , Learning Disabilities/etiology , Maze Learning/drug effects , Maze Learning/physiology , Mental Disorders/drug therapy , Mental Disorders/etiology , Mice , Mice, Transgenic , Motor Activity/drug effects , Motor Activity/physiology , Organ Culture Techniques , Receptor, Adenosine A2A/metabolism
10.
Purinergic Signal ; 3(4): 333-8, 2007 Sep.
Article in English | MEDLINE | ID: mdl-18404446

ABSTRACT

Brain-derived neurotrophic factor (BDNF), a member of the neurotrophin family, enhances synaptic transmission and regulates neuronal proliferation and survival. Functional interactions between adenosine A(2A) receptors (A(2A)Rs) and BDNF have been recently reported. In this article, we report some recent findings from our group showing that A(2A)Rs regulate both BDNF functions and levels in the brain. Whereas BDNF (10 ng/ml) increased the slope of excitatory postsynaptic field potentials (fEPSPs) in hippocampal slices from wild-type (WT) mice, it was completely ineffective in slices taken from A(2A)R knock-out (KO) mice. Furthermore, enzyme immunoassay studies showed a significant reduction in hippocampal BDNF levels in A(2A)R KO vs. WT mice. Having found an even marked reduction in the striatum of A(2A)R KO mice, and as both BDNF and A(2A)Rs have been implicated in the pathogenesis of Huntington's disease (HD), an inherited striatal neurodegenerative disease, we then evaluated whether the pharmacological blockade of A(2A)Rs could influence striatal levels of BDNF in an experimental model of HD-like striatal degeneration (quinolinic acid-lesioned rats) and in a transgenic mice model of HD (R6/2 mice). In both QA-lesioned rats and early symptomatic R6/2 mice (8 weeks), the systemic administration of the A(2A)R antagonist SCH58261 significantly reduced striatal BDNF levels. These results indicate that the presence and the tonic activation of A(2A)Rs are necessary to allow BDNF-induced potentiation of synaptic transmission and to sustain a normal BDNF tone. The possible functional consequences of reducing striatal BDNF levels in HD models need further investigation.

11.
Neuropharmacology ; 51(5): 1004-12, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16895732

ABSTRACT

The ability of CB(1) receptors to regulate the release of glutamate in the striatum, together with the finding that, in experimental models of Huntington disease (HD), both endocannabinoid levels and CB(1) receptor densities are reduced, has prompted the investigation on the neuroprotective role of the cannabinoids in HD. Quinolinic acid (QA) is an excitotoxin that, when injected in the rat striatum reproduces many features of HD and that acts by stimulating glutamate outflow. The aim of the present study was to test the ability of the cannabinoid receptor agonist WIN 55,212-2 to prevent the effects induced by QA in the rat striatum. In microdialysis experiments, probe perfusion with WIN 55,212-2 significantly and dose-dependently prevented the increase in extracellular glutamate induced by QA. In electrophysiological recordings in corticostriatal slices, the application of WIN 55,212-2 prevented QA-induced reduction of the field potential amplitude. Both effects of WIN 55,212-2 were prevented by the CB(1) receptor antagonist AM 251. In in vivo experiments, intrastriatal WIN 55,212-2 significantly attenuated the striatal damage induced by QA, although no significant effects were observed on a behavioural ground. These data demonstrate that the stimulation of CB(1) receptors might lead to neuroprotective effects against excitotoxic striatal toxicity.


Subject(s)
Corpus Striatum/drug effects , Morpholines/pharmacology , Naphthalenes/pharmacology , Neuroprotective Agents/pharmacology , Neurotoxins/pharmacology , Quinolinic Acid/pharmacology , Receptor, Cannabinoid, CB1/physiology , Animals , Behavior, Animal/drug effects , Benzoxazines , Cerebral Cortex/cytology , Cerebral Ventricles/drug effects , Dose-Response Relationship, Drug , Drug Interactions , Exploratory Behavior/drug effects , In Vitro Techniques , Male , Maze Learning/drug effects , Membrane Potentials/drug effects , Motor Activity/drug effects , Neurons/drug effects , Patch-Clamp Techniques/methods , Rats , Rats, Wistar
12.
Purinergic Signal ; 2(4): 619-25, 2006 Nov.
Article in English | MEDLINE | ID: mdl-18404464

ABSTRACT

The aim of the present paper was to examine, in a comparative way, the occurrence and the mechanisms of the interactions between adenosine A(2A) receptors (A(2A)Rs) and metabotropic glutamate 5 receptors (mGlu5Rs) in the hippocampus and the striatum. In rat hippocampal and corticostriatal slices, combined ineffective doses of the mGlu5R agonist 2-chloro-5-hydroxyphenylglycine (CHPG) and the A(2A)R agonist CGS 21680 synergistically reduced the slope of excitatory postsynaptic field potentials (fEPSPs) recorded in CA1 and the amplitude of field potentials (FPs) recorded in the dorsomedial striatum. The cyclic adenosine monophosphate (cAMP)/protein kinase A (PKA) pathway appeared to be involved in the effects of CGS 21680 in corticostriatal but not in hippocampal slices. In both areas, a postsynaptic locus of interaction appeared more likely. N-methyl-D: -aspartate (NMDA) reduced the fEPSP slope and FP amplitude in hippocampal and corticostriatal slices, respectively. Such an effect was significantly potentiated by CHPG in both areas. Interestingly, the A(2A)R antagonist ZM 241385 significantly reduced the NMDA-potentiating effect of CHPG. In primary cultures of rat hippocampal and striatal neurons (ED 17, DIV 14), CHPG significantly potentiated NMDA-induced lactate dehydrogenase (LDH) release. Again, such an effect was prevented by ZM 241385. Our results show that A(2A) and mGlu5 receptors functionally interact both in the hippocampus and in the striatum, even though different mechanisms seem to be involved in the two areas. The ability of A(2A)Rs to control mGlu5R-dependent effects may thus be a general feature of A(2A)Rs in different brain regions (irrespective of their density) and may represent an additional target for the development of therapeutic strategies against neurological disorders.

13.
J Neurochem ; 95(4): 1188-200, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16271052

ABSTRACT

Hippocampal metabotropic glutamate 5 receptors (mGlu5Rs) regulate both physiological and pathological responses to glutamate. Because mGlu5R activation enhances NMDA-mediated effects, and given the role played by NMDA receptors in synaptic plasticity and excitotoxicity, modulating mGlu5R may influence both the physiological and the pathological effects elicited by NMDA receptor stimulation. We evaluated whether adenosine A2A receptors (A(2A)Rs) modulated mGlu5R-dependent effects in the hippocampus, as they do in the striatum. Co-application of the A(2A)R agonist CGS 21680 with the mGlu5R agonist (RS)-2-chloro-s-hydroxyphenylglycine(CHPG) synergistically reduced field excitatory postsynaptic potentials in the CA1 area of rat hippocampal slices. Endogenous tone at A(2A)Rs seemed to be required to enable mGlu5R-mediated effects, as the ability of CHPG to potentiate NMDA effects was antagonized by the selective A(2A)R antagonist ZM 241385 in rat hippocampal slices and cultured hippocampal neurons, and abolished in the hippocampus of A(2A)R knockout mice. Evidence for the interaction between A(2A)Rs and mGlu5Rs was further strengthened by demonstrating their co-localization in hippocampal synapses. This is the first evidence showing that hippocampal A(2A)Rs and mGlu5Rs are co-located and act synergistically, and that A(2A)Rs play a permissive role in mGlu5R receptor-mediated potentiation of NMDA effects in the hippocampus.


Subject(s)
Hippocampus/cytology , N-Methylaspartate/pharmacology , Neurons/metabolism , Receptors, Adenosine A2/physiology , Receptors, Metabotropic Glutamate/physiology , Adenosine/analogs & derivatives , Adenosine/pharmacology , Adenosine A2 Receptor Agonists , Animals , Bicuculline/pharmacology , Blotting, Western/methods , Colforsin/pharmacology , Disks Large Homolog 4 Protein , Dose-Response Relationship, Drug , Drug Interactions , Electric Stimulation/methods , Embryo, Mammalian , Enzyme Inhibitors/pharmacology , Excitatory Amino Acid Agonists/pharmacology , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/physiology , Excitatory Postsynaptic Potentials/radiation effects , Female , Fluorescent Antibody Technique/methods , GABA Antagonists/pharmacology , Glycine/analogs & derivatives , Glycine/pharmacology , In Vitro Techniques , Intracellular Signaling Peptides and Proteins/metabolism , Male , Membrane Proteins/metabolism , Patch-Clamp Techniques/methods , Phenethylamines/pharmacology , Phenylacetates/pharmacology , Pregnancy , Presynaptic Terminals/metabolism , Pyridines/pharmacology , Qa-SNARE Proteins/metabolism , Rats , Rats, Wistar , Receptor, Metabotropic Glutamate 5 , Synaptophysin/metabolism , Vesicular Glutamate Transport Protein 1/metabolism
14.
J Neurochem ; 90(5): 1276-9, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15312183

ABSTRACT

The metabotropic glutamate receptors 5 (mGlu5Rs) and the adenosine A2A receptors (A2ARs) have been reported to functionally interact in the striatum. The aim of the present work was to verify the hypothesis that the state of activation of A2A Rs could influence mGlu5R-mediated effects in the striatum. In electrophysiological experiments (extracellular recording in rat corticostriatal slices), the ability of the selective mGlu5R agonist CHPG to potentiate the reduction of the field potential amplitude induced by NMDA was prevented not only by the selective mGlu5R antagonist MPEP, but also by the selective A2AR antagonist ZM 241385. Analogously, the application of CHPG potentiated NMDA-induced toxicity (measured by LDH release) in cultured striatal neurons, an effect that was abolished by both MPEP and ZM 241385. Finally, the A2AR agonist CGS 21680 potentiated CHGP effects, an action that was reproduced and abolished, respectively, by forskolin (an activator of the cAMP/protein kinase A, PKA, pathway) and KT 5720 (a PKA inhibitor). The results indicate that A2ARs exert a permissive role on mGlu5R-induced effects in the striatum. Such an interaction may represent an additional target for the development of therapeutic strategies towards striatal disorders.


Subject(s)
Adenosine/analogs & derivatives , Corpus Striatum/physiology , Glycine/analogs & derivatives , Receptors, Adenosine A2/physiology , Receptors, Metabotropic Glutamate/physiology , Adenosine/pharmacology , Animals , Carbazoles/pharmacology , Cells, Cultured , Corpus Striatum/drug effects , Dose-Response Relationship, Drug , Drug Interactions , Electrophysiology/methods , Embryo, Mammalian , Enzyme Inhibitors/pharmacology , Excitatory Amino Acid Agonists/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Female , Glycine/pharmacology , In Vitro Techniques , Indoles/pharmacology , Male , Membrane Potentials/drug effects , Membrane Potentials/physiology , N-Methylaspartate/pharmacology , Phenethylamines/pharmacology , Phenylacetates/pharmacology , Pregnancy , Purinergic P1 Receptor Agonists , Pyridines/pharmacology , Pyrroles/pharmacology , Rats , Rats, Wistar , Receptor, Metabotropic Glutamate 5 , Triazines/pharmacology , Triazoles/pharmacology
15.
J Neurochem ; 89(1): 152-6, 2004 Apr.
Article in English | MEDLINE | ID: mdl-15030399

ABSTRACT

Active uptake by neurons and glial cells is the main mechanism for maintaining extracellular glutamate at low, non-toxic concentrations. Activation of adenosine A(2A) receptors increases extracellular glutamate levels, while A(2A) receptor antagonists reduce stimulated glutamate outflow. Whether a modulation of the glutamate uptake system is involved in the effects elicited by A(2A) receptor blockers has never been investigated. This study examined the ability of adenosine A(2A) receptor antagonists to prevent the increase in glutamate levels induced by blockade of the glutamate uptake. In rats implanted with a microdialysis probe in the dorsal striatum, perfusion with 4 mm l-trans-pyrrolidine-2,4-dicarboxylic acid (PDC, a transportable competitive inhibitor of glutamate uptake), or 10 mm dihydrokainic acid (DHK, a non-transportable competitive inhibitor that mainly blocks the glial glutamate transporter GLT-1), significantly increased extracellular glutamate levels. The effects of PDC and DHK were completely prevented by the adenosine A(2A) receptor antagonists SCH 58261 (0.01 mg/kg i.p.) and/or ZM 241385 (5 nm via probe). Since an impairment in glutamate transporter function is thought to play a major role in neurodegenerative disorders, the regulation of glutamate uptake may be one of the mechanisms of the neuroprotective effects of A(2A) receptor antagonists.


Subject(s)
Adenosine A2 Receptor Antagonists , Corpus Striatum/metabolism , Glutamic Acid/metabolism , Kainic Acid/analogs & derivatives , Neurotransmitter Uptake Inhibitors/pharmacology , Pyrimidines/pharmacology , Triazines/pharmacology , Triazoles/pharmacology , Animals , Corpus Striatum/drug effects , Dicarboxylic Acids/pharmacology , Dose-Response Relationship, Drug , Excitatory Amino Acid Antagonists/pharmacology , Excitatory Amino Acid Transporter 2/antagonists & inhibitors , Kainic Acid/pharmacology , Male , Microdialysis , Neuroprotective Agents/pharmacology , Pyrrolidines/pharmacology , Rats , Rats, Wistar , Receptor, Adenosine A2A/metabolism
16.
Neurology ; 61(11 Suppl 6): S19-23, 2003 Dec 09.
Article in English | MEDLINE | ID: mdl-14663004

ABSTRACT

Recently evidence has been presented that adenosine A2A and dopamine D2 receptors form functional heteromeric receptor complexes as demonstrated in human neuroblastoma cells and mouse fibroblast Ltk- cells. These A2A/D2 heteromeric receptor complexes undergo coaggregation, cointernalization, and codesensitization on D2 or A2A receptor agonist treatments and especially after combined agonist treatment. It is hypothesized that the A2A/D2 receptor heteromer represents the molecular basis for the antagonistic A2A/D2 receptor interactions demonstrated at the biochemical and behavioral levels. Functional heteromeric complexes between A2A and metabotropic glutamate 5 receptors (mGluR5) have also recently been demonstrated in HEK-293 cells and rat striatal membrane preparations. The A2A/mGluR5 receptor heteromer may account for the synergism found after combined agonist treatments demonstrated in different in vitro and in vivo models. D2, A2A, and mGluR5 receptors are found together in the dendritic spines of the striatopallidal GABA neurons. Therefore, possible D2/A2A/mGluR5 multimeric receptor complexes and the receptor interactions within them may have a major role in controlling the dorsal and ventral striatopallidal GABA neurons involved in Parkinson's disease and in schizophrenia and drug addiction, respectively.


Subject(s)
Corpus Striatum/metabolism , Parkinson Disease/metabolism , Receptor, Adenosine A2A/metabolism , Signal Transduction/physiology , Animals , Cell Line , Dimerization , Humans , Macromolecular Substances , Mice , Parkinson Disease/therapy , Receptor, Metabotropic Glutamate 5 , Receptors, Dopamine D2/metabolism , Receptors, Metabotropic Glutamate/metabolism , gamma-Aminobutyric Acid/metabolism
17.
Neurology ; 61(11 Suppl 6): S69-71, 2003 Dec 09.
Article in English | MEDLINE | ID: mdl-14663014

ABSTRACT

Because an increased glutamate outflow is thought to play a crucial role in triggering excitotoxic neuronal death, drugs able to regulate glutamate release could be effective for the management of neurodegenerative diseases. In this article, the authors discuss the hypothesis that adenosine A2A receptor antagonists (A2A antagonists) may belong to the aforementioned category. In rats bilaterally lesioned with the excitotoxin quinolinic acid (QA) in the striatum, the A2A antagonist SCH 58261 significantly reduced the motor, EEG, and neuropathologic changes induced by the lesion. Such effects of SCH 58261 occurred only at low doses and were paralleled by an inhibition of QA-stimulated glutamate release. The role played by A2A antagonists in the regulation of glutamate outflow was also confirmed by preliminary results obtained in the model of paired-pulse stimulation in corticostriatal slices. Conversely, based on data obtained in cultured striatal neurons, A2A antagonists appear unable to directly inhibit NMDA effects. In conclusion, A2A antagonists show clear neuroprotective effects in models of brain injury, although their actual therapeutic potential needs to be confirmed in a wider range of doses and in models of neurodegenerative diseases in which presynaptic and postsynaptic effects play different relative roles.


Subject(s)
Glutamic Acid/metabolism , Neurotoxins/metabolism , Receptor, Adenosine A2A/metabolism , Adenosine A2 Receptor Antagonists , Animals , Cells, Cultured , Corpus Striatum/cytology , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Drug Administration Routes , Electric Stimulation , Electroencephalography/drug effects , Excitatory Amino Acid Antagonists/pharmacology , Glutamic Acid/toxicity , In Vitro Techniques , Microdialysis , Motor Activity/drug effects , Neurons/drug effects , Neurons/metabolism , Neuroprotective Agents/pharmacology , Neurotoxins/toxicity , Pyrimidines/pharmacology , Quinolinic Acid/metabolism , Quinolinic Acid/toxicity , Rats , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Synapses/drug effects , Synapses/metabolism , Triazoles/pharmacology
19.
Neuropsychopharmacology ; 25(4): 505-13, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11557164

ABSTRACT

In 6-hydroxydopamine-lesioned rats, the selective mGlu(5) receptor agonist (RS)-2-Cholro-5-Hydroxyphenylglycine (CHPG, 1-6 microg/10 microl intracerebroventricularly) significantly inhibited contralateral turning induced by quinpirole and, to a lesser extent, that induced by SKF 38393. The inhibitory effects of CHPG on quinpirole-induced turning were significantly potentiated by an adenosine A(2A) receptor agonist (CGS 21680, 0.2 mg/kg IP) and attenuated by an A(2A) receptor antagonist (SCH 58261, 1 mg/kg IP). In rat striatal membranes, CHPG (100-1,000 nM) significantly reduced the affinity of the high-affinity state of D(2) receptors for the agonist, an effect potentiated by CGS 21680 (30 nM). These results show the occurrence of functional interactions among mGlu(5), adenosine A(2A), and dopamine D(2) receptors in the regulation of striatal functioning, and suggest that mGlu(5) receptors may be regarded as alternative/integrative targets for the development of therapeutic strategies in the treatment of Parkinson's disease.


Subject(s)
Dopamine Agonists/pharmacology , Excitatory Amino Acid Agonists/pharmacology , Glycine/pharmacology , Neostriatum/metabolism , Phenylacetates/pharmacology , Quinpirole/antagonists & inhibitors , Receptors, Dopamine D2/metabolism , Receptors, Metabotropic Glutamate/agonists , Receptors, Purinergic P1/metabolism , Stereotyped Behavior/drug effects , Sympathectomy, Chemical , 2,3,4,5-Tetrahydro-7,8-dihydroxy-1-phenyl-1H-3-benzazepine/pharmacology , Animals , Dopamine/metabolism , Excitatory Amino Acid Agonists/administration & dosage , Extracellular Space/metabolism , Functional Laterality , Glycine/administration & dosage , Glycine/analogs & derivatives , Injections, Intraventricular , Male , Microdialysis , Motor Activity/drug effects , Neostriatum/drug effects , Oxidopamine , Phenylacetates/administration & dosage , Quinpirole/pharmacology , Rats , Rats, Sprague-Dawley , Receptor, Adenosine A2A , Receptor, Metabotropic Glutamate 5 , Receptors, Dopamine D1/agonists , Receptors, Dopamine D2/drug effects , Receptors, Purinergic P1/drug effects
20.
Eur J Pharmacol ; 421(3): 177-80, 2001 Jun 15.
Article in English | MEDLINE | ID: mdl-11516434

ABSTRACT

The aim of the present work was to determine whether systemic administration of the adenosine A(2A) receptor antagonist, SCH 58261 (7-(2-phenylethyl)-5-amino-2-(2-furyl)-pyrazolo-[4,3-e]-1,2,4,triazolo[1,5-c]pyrimidine), could modulate striatal glutamate outflow in the rat. Microdialysis experiments were performed in male Wistar rats implanted with microdialysis probes in the striatum. Pretreatment (15 min before) with SCH 58261 (0.01 and 0.1, but not 1 mg/kg intraperitoneally) significantly prevented K(+)-stimulated glutamate release. These results suggest that SCH 58261 could possess neuroprotective effects in the low dose range, while, at higher doses, the occurrence of additional mechanisms may limit the neuroprotective potential of this drug.


Subject(s)
Corpus Striatum/drug effects , Glutamic Acid/drug effects , Neuroprotective Agents/pharmacology , Potassium/pharmacology , Pyrimidines/pharmacology , Triazoles/pharmacology , Animals , Corpus Striatum/metabolism , Dose-Response Relationship, Drug , Glutamic Acid/metabolism , Male , Microdialysis , Purinergic P1 Receptor Antagonists , Rats , Rats, Wistar , Receptor, Adenosine A2A , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...