Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 6 de 6
1.
Mol Neurobiol ; 54(1): 423-436, 2017 01.
Article En | MEDLINE | ID: mdl-26742520

Accumulating evidences indicate that endogenous modulators of excitatory synapses in the mammalian brain are potential targets for treating neuropsychiatric disorders. Indeed, glutamatergic and adenosinergic neurotransmissions were recently highlighted as potential targets for developing innovative anxiolytic drugs. Accordingly, it has been shown that guanine-based purines are able to modulate both adenosinergic and glutamatergic systems in mammalian central nervous system. Here, we aimed to investigate the potential anxiolytic-like effects of guanosine and its effects on the adenosinergic and glutamatergic systems. Acute/systemic guanosine administration (7.5 mg/kg) induced robust anxiolytic-like effects in three classical anxiety-related paradigms (elevated plus maze, light/dark box, and round open field tasks). These guanosine effects were correlated with an enhancement of adenosine and a decrement of glutamate levels in the cerebrospinal fluid. Additionally, pre-administration of caffeine (10 mg/kg), an unspecific adenosine receptors' antagonist, completely abolished the behavioral and partially prevented the neuromodulatory effects exerted by guanosine. Although the hippocampal glutamate uptake was not modulated by guanosine (both ex vivo and in vitro protocols), the synaptosomal K+-stimulated glutamate release in vitro was decreased by guanosine (100 µM) and by the specific adenosine A1 receptor agonist, 2-chloro-N 6-cyclopentyladenosine (CCPA, 100 nM). Moreover, the specific adenosine A1 receptor antagonist 8-cyclopentyl-1,3-dipropylxanthine (DPCPX, 100 nM) fully reversed the inhibitory guanosine effect in the glutamate release. The pharmacological modulation of A2a receptors has shown no effect in any of the evaluated parameters. In summary, the guanosine anxiolytic-like effects seem closely related to the modulation of adenosinergic (A1 receptors) and glutamatergic systems.


Adenosine A1 Receptor Antagonists/pharmacology , Adenosine/metabolism , Anti-Anxiety Agents/therapeutic use , Glutamic Acid/metabolism , Guanosine/therapeutic use , Receptor, Adenosine A1/metabolism , Animals , Anti-Anxiety Agents/pharmacology , Anxiety/drug therapy , Anxiety/metabolism , Guanosine/pharmacology , Hippocampus/drug effects , Hippocampus/metabolism , Male , Neurotransmitter Agents/metabolism , Rats , Rats, Wistar
2.
Purinergic Signal ; 12(1): 149-59, 2016 Mar.
Article En | MEDLINE | ID: mdl-26695181

In addition to its intracellular roles, the nucleoside guanosine (GUO) also has extracellular effects that identify it as a putative neuromodulator signaling molecule in the central nervous system. Indeed, GUO can modulate glutamatergic neurotransmission, and it can promote neuroprotective effects in animal models involving glutamate neurotoxicity, which is the case in brain ischemia. In the present study, we aimed to investigate a new in vivo GUO administration route (intranasal, IN) to determine putative improvement of GUO neuroprotective effects against an experimental model of permanent focal cerebral ischemia. Initially, we demonstrated that IN [(3)H] GUO administration reached the brain in a dose-dependent and saturable pattern in as few as 5 min, presenting a higher cerebrospinal GUO level compared with systemic administration. IN GUO treatment started immediately or even 3 h after ischemia onset prevented behavior impairment. The behavior recovery was not correlated to decreased brain infarct volume, but it was correlated to reduced mitochondrial dysfunction in the penumbra area. Therefore, we showed that the IN route is an efficient way to promptly deliver GUO to the CNS and that IN GUO treatment prevented behavioral and brain impairment caused by ischemia in a therapeutically wide time window.


Brain Ischemia/drug therapy , Guanosine/administration & dosage , Guanosine/therapeutic use , Neuroprotective Agents/administration & dosage , Neuroprotective Agents/therapeutic use , Stroke/drug therapy , Administration, Intranasal , Animals , Behavior, Animal , Brain Ischemia/psychology , Cerebral Infarction/pathology , Cerebral Infarction/prevention & control , Dose-Response Relationship, Drug , Exploratory Behavior/drug effects , Guanosine/cerebrospinal fluid , Guanosine/pharmacokinetics , Male , Mitochondria/drug effects , Neuroprotective Agents/cerebrospinal fluid , Neuroprotective Agents/pharmacokinetics , Rats , Rats, Wistar , Stroke/psychology
3.
PLoS One ; 9(2): e90693, 2014.
Article En | MEDLINE | ID: mdl-24587409

BACKGROUND AND PURPOSE: Stroke is a devastating disease. Both excitotoxicity and oxidative stress play important roles in ischemic brain injury, along with harmful impacts on ischemic cerebral tissue. As guanosine plays an important neuroprotective role in the central nervous system, the purpose of this study was to evaluate the neuroprotective effects of guanosine and putative cerebral events following the onset of permanent focal cerebral ischemia. METHODS: Permanent focal cerebral ischemia was induced in rats by thermocoagulation. Guanosine was administered immediately, 1 h, 3 h and 6 h after surgery. Behavioral performance was evaluated by cylinder testing for a period of 15 days after surgery. Brain oxidative stress parameters, including levels of ROS/RNS, lipid peroxidation, antioxidant non-enzymatic levels (GSH, vitamin C) and enzymatic parameters (SOD expression and activity and CAT activity), as well as glutamatergic parameters (EAAC1, GLAST and GLT1, glutamine synthetase) were analyzed. RESULTS: After 24 h, ischemic injury resulted in impaired function of the forelimb, caused brain infarct and increased lipid peroxidation. Treatment with guanosine restored these parameters. Oxidative stress markers were affected by ischemic insult, demonstrated by increased ROS/RNS levels, increased SOD expression with reduced SOD activity and decreased non-enzymatic (GSH and vitamin C) antioxidant defenses. Guanosine prevented increased ROS/RNS levels, decreased SOD activity, further increased SOD expression, increased CAT activity and restored vitamin C levels. Ischemia also affected glutamatergic parameters, illustrated by increased EAAC1 levels and decreased GLT1 levels; guanosine reversed the decreased GLT1 levels and did not affect the EAAC1 levels. CONCLUSION: The effects of brain ischemia were strongly attenuated by guanosine administration. The cellular mechanisms involved in redox and glutamatergic homeostasis, which were both affected by the ischemic insult, were also modulated by guanosine. These observations reveal that guanosine may represent a potential therapeutic agent in cerebral ischemia by preventing oxidative stress and excitotoxicity.


Brain Injuries/prevention & control , Brain Injuries/physiopathology , Brain Ischemia/physiopathology , Guanosine/pharmacology , Animals , Ascorbic Acid/metabolism , Blotting, Western , Brain Injuries/metabolism , Brain Ischemia/etiology , Catalase/metabolism , Dose-Response Relationship, Drug , Drug Administration Schedule , Electrocoagulation/adverse effects , Excitatory Amino Acid Transporter 3/metabolism , Exploratory Behavior/drug effects , Exploratory Behavior/physiology , Forelimb/drug effects , Forelimb/physiopathology , Glutamate Plasma Membrane Transport Proteins/metabolism , Glutamate-Ammonia Ligase/metabolism , Glutathione/metabolism , Guanosine/administration & dosage , Lipid Peroxidation/drug effects , Male , Neuroprotective Agents/pharmacology , Nitric Oxide/metabolism , Rats, Wistar , Reactive Oxygen Species/metabolism , Superoxide Dismutase/metabolism
4.
Neurol Sci ; 33(5): 985-97, 2012 Oct.
Article En | MEDLINE | ID: mdl-22167652

Chronic cerebral hypoperfusion contributes to a cognitive decline related to brain disorders. Its experimental model in rats is a permanent bilateral common carotid artery occlusion (2VO). Overstimulation of the glutamatergic system excitotoxicity due to brain energetic disturbance in 2VO animals seems to play a pivotal role as a mechanism of cerebral damage. The nucleoside guanosine (GUO) exerts extracellular effects including antagonism of glutamatergic activity. Accordingly, our group demonstrated several neuroprotective effects of GUO against glutamatergic excitotoxicity. Therefore, in this study, we evaluated a chronic GUO treatment effects in rats submitted to 2VO. We evaluated the animals performance in the Morris water maze and hippocampal damage by neurons and astrocytes immunohistochemistry. In addition, we investigated the cerebrospinal fluid (CSF) brain derived neurotrophic factor (BDNF) and serum S100B levels. Additionally, the purine CSF and plasma levels were determined. GUO treatment did not prevent the cognitive impairment promoted by 2VO. However, none of the 2VO animals treated with GUO showed differences in the hippocampal regions compared to control, while 20% of 2VO rats not treated with GUO presented loss of pyramidal neurons and increased glial labeling cells in CA1 hippocampal region. In addition, we did not observe differences in CSF BDNF nor serum S100B levels among the groups. Of note, both the 2VO surgery and GUO treatment changed the purine CSF and plasma profile. In conclusion, GUO treatment did not prevent the cognitive impairment observed in 2VO animals, but our data suggest that GUO could be neuroprotective against hippocampal damage induced by 2VO.


Brain Ischemia/drug therapy , Cognition/drug effects , Guanosine/pharmacology , Hippocampus/drug effects , Neuroprotective Agents/pharmacology , Animals , Brain Ischemia/metabolism , Brain Ischemia/pathology , Chromatography, High Pressure Liquid , Cognition Disorders/drug therapy , Cognition Disorders/metabolism , Cognition Disorders/pathology , Hippocampus/metabolism , Hippocampus/pathology , Immunohistochemistry , Male , Maze Learning/drug effects , Purines/blood , Purines/cerebrospinal fluid , Rats , Rats, Wistar
5.
Life Sci ; 87(9-10): 325-32, 2010 Aug 28.
Article En | MEDLINE | ID: mdl-20655932

AIMS: To investigate the effect of N(omega)-Nitro-L-arginine methyl ester (l-NAME) treatment, known to induce a sustained elevation of blood pressure, on ectonucleotidase activities in kidney membranes of rats. MAIN METHODS: L-NAME (30 mg/kg/day) was administered to Wistar rats for 14 days in the drinking water. Enzyme activities were determined colorimetrically and their gene expression patterns were analyzed by semi-quantitative RT-PCR. The metabolism of ATP and the accumulation of adenosine were evaluated by HPLC in kidney membranes from control and hypertensive rats. PKC phosphorylation state was investigated by Western blot. KEY FINDINGS: We observed an increase in systolic blood pressure from 115+/-12 mmHg (control group) to 152+/-18 mmHg (l-NAME-treated group). Furthermore, the hydrolysis of ATP, ADP, AMP, and p-Nph-5'TMP was also increased (17%, 35%, 27%, 20%, respectively) as was the gene expression of NTPDase2, NTPDase3 and NPP3 in kidneys of hypertensive animals. Phospho-PKC was increased in hypertensive rats. SIGNIFICANCE: The general increase in ATP hydrolysis and in ecto-5'-nucleotidase activity suggests a rise in renal adenosine levels and in renal autoregulatory responses in order to protect the kidney against the threat presented by hypertension.


Adenosine Triphosphatases/metabolism , Cell Membrane/enzymology , Hypertension/enzymology , Kidney/enzymology , NG-Nitroarginine Methyl Ester/pharmacology , Adenosine Triphosphatases/genetics , Animals , Blood Pressure/drug effects , Cell Membrane/drug effects , Chromatography, High Pressure Liquid , Creatinine/blood , Disease Models, Animal , Gene Expression/drug effects , Heart Rate/drug effects , Hypertension/blood , Hypertension/physiopathology , Kidney/drug effects , Kidney/physiopathology , Male , Rats , Rats, Wistar , Reverse Transcriptase Polymerase Chain Reaction
6.
Blood Cells Mol Dis ; 41(2): 223-9, 2008.
Article En | MEDLINE | ID: mdl-18559295

It is well known that hypertension is closely associated to the development of vascular diseases and that the inhibition of nitric oxide biosynthesis by administration of Nomega-Nitro-L-arginine methyl ester hydrochloride(L-NAME) leads to arterial hypertension. In the vascular system, extracellular purines mediate several effects;thus, ADP is the most important platelet agonist and recruiting ag ent, while adenosine, an end product of nucleotide metabolism, is a vasodilator and inhibitor of platelet activation and recruitment. Members of several families of enzymes, known as ectonucleotidases, including E-NTPDases (ecto-nucleoside triphosphate diphosphohydrolase), E-NPP (ecto-nucleotide pyrophosphatase/phosphodiesterase) and 5'-nucleotidase are able to hydrolyze extracellular nucleotides until their respective nucleosides. We investigated the ectonucleotidase activities of serum and platelets from rats made hypertensive by oral administration of L-NAME (30 mg/kg/day for 14 days or 30 mg/kg/day for 14 days plus 7 days of L-NAME washout, in the drinking water) in comparison to normotensive control rats. L-NAME promoted a significant rise in systolic blood pressure from 112 +/- 9.8 to 158 +/- 23 mmHg. The left ventricle weight index (LVWI) was increased in rats treated with L-NAME for 14 days when compared to control animals. In serum samples, ATP, ADP and AMP hydrolysis were reduced by about 27%, 36% and 27%, respectively. In platelets, the decrease in ATP, ADP and AMP hydrolysis was approximately 27%, 24% and 32%, respectively. All parameters recovered after 7 days of L-NAME washout. HPLC demonstrated a reduction in ADP, AMP and hypoxanthine levels by about 64%, 69% and 87%,respectively. In this study, we showed that ectonucleotidase activities are decreased in serum and platelets from L-NAME-treated rats, which should represent an additional risk for the development of hypertension. The modulation of ectonucleotidase activities may represent an approach to antihypertensive therapy via inhibition of spontaneous platelet activation and recruitment, as well as thrombus formation.


Adenosine Triphosphatases/metabolism , Hypertension/etiology , NG-Nitroarginine Methyl Ester/pharmacology , Adenine Nucleotides/analysis , Adenine Nucleotides/metabolism , Adenosine Triphosphatases/drug effects , Animals , Blood Platelets/enzymology , Hypertension/chemically induced , Hypertrophy, Left Ventricular , NG-Nitroarginine Methyl Ester/administration & dosage , Rats , Serum/enzymology
...